Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 118(14): 3901-10, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21835956

ABSTRACT

Multiple myeloma is a plasma cell malignancy that is heterogeneous with respect to its causative molecular abnormalities and the treatment response of patients. The Bcl-2 protein family is critical for myeloma cell survival. ABT-737 is a cell-permeant compound that binds to Bcl-2 and Bcl-x(L) but not to Mcl-1. Using a myeloma cell line collection (n = 25) representative of different molecular translocations, we showed that ABT-737 effectively kills a subset of cell lines (n = 6), with a median lethal dose ranging from 7 ± 0.4 nM to 150 ± 7.5 nM. Of interest, all sensitive cell lines harbored a t(11;14). We demonstrated that ABT-737-sensitive and ABT-737-resistant cell lines could be differentiated by the BCL2/MCL1 expression ratio. A screen of a public expression database of myeloma patients indicates that the BCL2/MCL1 ratio of t(11;14) and hyperdiploid patients was significantly higher than in all other groups (P < .001). ABT-737 first induced the disruption of Bcl-2/Bax, Bcl-2/Bik, or Bcl-2/Puma complexes, followed by the disruption of Bcl-2 heterodimers with Bak and Bim. Altogether, the identification of a subset of cell lines and primary cells effectively killed by ABT-737 alone supported the evaluation of ABT-263, an orally active counterpart to ABT-737, for the treatment of t(11;14) and hyperdiploid groups of myeloma harboring a Bcl-2(high)/Mcl-1(low) profile.


Subject(s)
Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Gene Expression Regulation, Neoplastic , Multiple Myeloma/drug therapy , Nitrophenols/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/therapeutic use , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cyclin D1/metabolism , Humans , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/pharmacology , Piperazines/pharmacology , Piperazines/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Tumor Cells, Cultured
2.
Biology (Basel) ; 12(7)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37508420

ABSTRACT

Multiple Myeloma (MM) and its preexisting stage, termed Monoclonal Gammopathy of Undetermined Significance (MGUS), have long been considered mainly as genomic diseases. However, the bone changes observed in both conditions have led to a reassessment of the role of the bone microenvironment, mainly the endosteal niche in their genesis. Here, we consider the disruption of the endosteal niche in the bone marrow, that is, the shift of the endosteal niche from an osteoblastic to an osteoclastic profile produced by bone senescence and inflammaging, as the key element. Thus, this disrupted endosteal niche is proposed to represent the permissive microenvironment necessary not only for the emergence of MM from MGUS but also for the emergence and maintenance of MGUS. Moreover, the excess of osteoclasts would favor the presentation of antigens (Ag) into the endosteal niche because osteoclasts are Ag-presenting cells. As such, they could significantly stimulate the presentation of some specific Ag and the clonal expansion of the stimulated cells as well as favor the expansion of such selected clones because osteoclasts are immunosuppressive. We also discuss this scenario in the Gaucher disease, in which the high incidence of MGUS and MM makes it a good model both at the bone level and the immunological level. Finally, we envisage that this endosteal niche disruption would increase the stochasticity (epigenetic and genetic instability) in the selected clones, according to our Tissue Disruption-induced cell Stochasticity (TiDiS) theory.

3.
Blood Cells Mol Dis ; 48(4): 247-53, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22341562

ABSTRACT

The purpose of this study was to identify the pathways associated with the ability of CD138(+) human myeloma cells to form colonies in a serum-free semi-solid human collagen-based assay. Only 26% (7 of 27) of human myeloma cell lines were able to spontaneously form colonies. This spontaneous clonogenic growth correlated with the expression of the NOTCH ligand JAG2 (p<0.001). Blocking JAG-NOTCH interactions with NOTCH-Fc chimeric molecules impaired self-colony formation, indicating a role for JAG-NOTCH pathway in colony formation. In two cell lines, silencing of JAG2 blocked both colony formation and in vivo tumor formation in immunocompromised mice. RT-PCR and flow cytometry analysis revealed that JAG2 is often expressed by CD138(+) primary cells. Our results indicate that spontaneous clonogenic growth of myeloma cells requires the expression of JAG2.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Multiple Myeloma/metabolism , Receptors, Notch/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-2 Protein , Ligands , Membrane Proteins/genetics , Mice , Mice, SCID , Multiple Myeloma/genetics , Signal Transduction , Syndecan-1/metabolism
4.
Crit Rev Oncol Hematol ; 172: 103640, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35183697

ABSTRACT

The standard model of multiple myeloma (MM) oncogenesis from monoclonal gammopathy of undetermined significance (MGUS) relies on genetic instability in the normal counterparts of MM cells. However, the importance of both MGUS-associated and MM-induced bone changes has been recently re-appraised, emphasizing the bone microenvironment (BME) as a tissue of significance. In this review, we propose that early BME alterations (bone senescence and inflammation, i.e., bone inflamm'aging) at the pre-MGUS stage could be causal, and not simply permissive, and creative of phenotypic instability and genetic alterations thanks to the concept of tissue disruption-induced cell stochasticity (TiDiS). This article offers a bone scenario challenging the chromosome-and-gene-centric standard model of MM oncogenesis. The high incidence of both MGUS and MM in Gaucher disease supports such a scenario.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Paraproteinemias , Cell Transformation, Neoplastic/genetics , Humans , Monoclonal Gammopathy of Undetermined Significance/epidemiology , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Tumor Microenvironment
5.
Haematologica ; 96(4): 574-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21173094

ABSTRACT

BACKGROUND: Multiple myeloma is a plasma-cell tumor with heterogeneity in molecular abnormalities and treatment response. DESIGN AND METHODS: We have assessed whether human myeloma cell lines have kept patients' heterogeneity using Affymetrix gene expression profiling of 40 human myeloma cell lines obtained with or without IL6 addition and could provide a signature for stratification of patient risk. RESULTS: Human myeloma cell lines, especially those derived in the presence of IL6, displayed a heterogeneity that overlaps that of the patients with multiple myeloma. Human myeloma cell lines segregated into 6 groups marked by overexpression of MAF, MMSET, CCND1, FRZB with or without overexpression of cancer testis antigens (CTA). Cell lines of CTA/MAF and MAF groups have a translocation involving C-MAF or MAFB, cell lines of groups CCND1-1 and CCND1-2like have a t(11;14) and cell lines of group MMSET have a t(4;14). The CTA/FRZB group comprises cell lines that had no or no recurrent 14q32 translocation. Expression of 248 genes accounted for human myeloma cell line molecular heterogeneity. Human myeloma cell line heterogeneity genes comprise genes with prognostic value for survival of patients making it possible to build a powerful prognostic score involving a total of 13 genes. CONCLUSIONS: Human myeloma cell lines derived in the presence of IL6 recapitulate the molecular diversity of multiple myeloma that made it possible to design, using human myeloma cell line heterogeneity genes, a high-risk signature for patients at diagnosis. We propose this classification to be used when addressing the physiopathology of multiple myeloma with human myeloma cell lines.


Subject(s)
Gene Expression Profiling , Multiple Myeloma/diagnosis , Cell Line, Tumor , Cluster Analysis , Female , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Genetic Markers , Humans , Interleukin-6/metabolism , Male , Multiple Myeloma/genetics , Prognosis , Risk Factors , Survival Analysis
6.
J Immunol ; 182(7): 4471-8, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299748

ABSTRACT

Multiple myeloma (MM) cells express TLR. It has been shown that TLR ligands induce the proliferation, survival, and immune surveillance escape of MM cells through MyD88-TLR pathways. Deciphering TLR function in MM cells will help in understanding the mechanisms of tumor cell growth. In this study, we examined the response of MM cells to the MyD88-independent/TIR-domain-containing adapter-inducing IFN-beta-dependent TLR3. Deregulation of NF-kappaB pathway is a feature of MM cells, and we wondered whether TLR3 activation could mobilize the NF-kappaB pathway. We show that five of seven human myeloma cell line (HMCL) cells expressed TLR3. In the presence of the synthetic TLR3 ligand (poly(I:C)), activation of NF-kappaB pathway was observed in three of five selected TLR3(+) HMCL, NCI-H929, RPMI 8226, and KMM1. In agreement with NF-kappaB activation, only these three HMCL responded to poly(I:C), although by either an increase (KMM1) or a decrease (NCI-H929, RPMI 8226) of proliferation. We show that KMM1 increase of proliferation was prevented by NF-kappaB inhibitor. In contrast, inhibition of proliferation in both NCI-H929 and RPMI 8226 was due to IFN-alpha-induced apoptosis. We next demonstrated that p38 MAPK pathway controlled both IFN-alpha secretion and IFN-alpha-mediated cell death. Moreover, cell death also involved activation of ERK1/2 pathway. In conclusion, our results show that TLR3 ligand induces NF-kappaB pathway activation in MM and support a switching function of type I IFN in the functional outcome of TLR3 triggering in tumor cells.


Subject(s)
Interferon-alpha/biosynthesis , Multiple Myeloma/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/immunology , Apoptosis/immunology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Fluorescent Antibody Technique , Humans , Interferon-alpha/immunology , Ligands , Multiple Myeloma/metabolism , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Poly I-C/immunology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 3/metabolism
7.
J Immunol ; 183(7): 4371-7, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734228

ABSTRACT

Hypomethylated CpG oligodeoxynucleotides (CpG ODNs) target TLR9 expressed by immune cells and are currently being evaluated as adjuvants in clinical trials. However, TLR signaling can promote some tumor growth and immune evasion, such as in multiple myeloma (MM). Therefore, deciphering the effects of CpG ODNs on cancer cells will help in preventing these adverse effects and in designing future clinical trials. TLR activation induces multiple signaling pathways, notably NF-kappaB that has been involved in the resistance to TRAIL. Thus, we wondered if CpG ODNs could modulate TRAIL-induced apoptosis in different models of tumors. Here, we show that TLR9+ (NCI-H929, NAN6, KMM1) and TLR9- MM cells (MM1S) were protected by CpG ODNs against recombinant TRAIL-induced apoptosis. By using two fully human, agonist mAbs directed against TRAIL receptors DR4 and DR5 (mapatumumab and lexatumumab, respectively), we show that the protection was restricted to DR5-induced apoptosis. Similar results were observed for two colon cancer (C45 and Colo205) and two breast cancer cell lines (HCC1569 and Cal51). The protection of CpG ODNs was mediated by its nuclease-resistant phosphorothioate backbone independent of TLR9. We next demonstrated by surface plasmon resonance that phosphorothioate-modified CpG ODNs directly bound to either TRAIL or lexatumumab and then decreased their binding to DR5. Finally, NK cell lysis of a DR5-sensitive MM cell line (NCI-H929) through TRAIL was partially inhibited by phosphorothioate-modified CpG ODNs. In conclusion, our results suggest that the phosphorothioate modification of CpG ODNs could dampen the clinical efficacy of CpG ODN-based adjuvants by altering TRAIL/TRAIL receptor interaction.


Subject(s)
Adjuvants, Immunologic/physiology , Apoptosis/immunology , Breast Neoplasms/prevention & control , Colonic Neoplasms/prevention & control , Multiple Myeloma/prevention & control , Oligodeoxyribonucleotides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , Toll-Like Receptor 9/metabolism , Adjuvants, Immunologic/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , CpG Islands/immunology , Female , Humans , Ligands , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Phosphorothioate Oligonucleotides/pharmacology , Toll-Like Receptor 9/biosynthesis
8.
Kidney Int ; 78(5): 503-13, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20531452

ABSTRACT

Several transplant patients maintain stable kidney graft function in the absence of immunosuppression. Here we compared the characteristics of their peripheral B cells to that of others who had stable graft function but were under pharmacologic immunosuppression, to patients with chronic rejection and to healthy volunteers. In drug-free long-term graft function (DF) there was a significant increase in both absolute cell number and frequency of total B cells; particularly activated, memory and early memory B cells. These increased B-cell numbers were associated with a significantly enriched transcriptional B-cell profile. Costimulatory/migratory molecules (B7-2/CD80, CD40, and CD62L) were upregulated in B cells; particularly in memory CD19(+)IgD(-)CD38(+/-)CD27(+) B cells in these patients. Their purified B cells, however, responded normally to a polyclonal stimulation and did not have cytokine polarization. This phenotype was associated with the following specific characteristics which include an inhibitory signal (decreased FcgammaRIIA/FcgammaRIIB ratio); a preventive signal of hyperactive B-cell response (an increase in BANK1, which negatively modulates CD40-mediated AKT activation); an increased number of B cells expressing CD1d and CD5; an increased BAFF-R/BAFF ratio that could explain why these patients have more peripheral B cells; and a specific autoantibody profile. Thus, our findings show that patients with DF have a particular blood B-cell phenotype that may contribute to the maintenance of long-term graft function.


Subject(s)
B-Lymphocytes/immunology , Immune Tolerance , Immunologic Memory , Kidney Transplantation/immunology , ADP-ribosyl Cyclase 1/analysis , Adaptor Proteins, Signal Transducing/physiology , Adult , Aged , Aged, 80 and over , Antigens, CD19/analysis , Autoantibodies/biosynthesis , Female , Gene Expression Profiling , Humans , Isoantibodies/biosynthesis , Lymphocyte Activation , Male , Membrane Glycoproteins/analysis , Membrane Proteins/physiology , Middle Aged , Phenotype
9.
Blood ; 112(6): 2205-13, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18591383

ABSTRACT

The humoral immune system senses microbes via recognition of specific microbial molecular motifs by Toll-like receptors (TLRs). These encounters promote plasma cell differentiation and antibody production. Recent studies have demonstrated the importance of the TLR system in enhancing antibody-mediated defense against infections and maintaining memory B cells. These results have led the way to the design of vaccines that target B cells by engaging TLRs. In hematologic malignancies, cells often retain B cell-specific receptors and associated functions. Among these, TLRs are currently exploited to target different subclasses of B-cell leukemia, and TLR agonists are currently being evaluated in clinical trials. However, accumulating evidence suggests that endogenous TLR ligands or chronic infections promote tumor growth, thus providing a need for further investigations to decipher the exact function of TLRs in the B-cell lineage and in neoplastic B cells. The aim of this review is to present and discuss the latest advances with regard to the expression and function of TLRs in both healthy and malignant B cells. Special attention will be focused on the growth-promoting effects of TLR ligands on leukemic B cells and their potential clinical impact.


Subject(s)
B-Lymphocytes/immunology , Toll-Like Receptors/immunology , Antibody Formation , Humans , Leukemia, B-Cell/pathology , Ligands , Lymphoma, B-Cell/pathology
10.
J Immunol ; 181(10): 6837-42, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981102

ABSTRACT

IL-21 is a member of the type I cytokine family related most closely to IL-2 and IL-15. IL-21 is a pleiotropic cytokine, produced by T, NKT, and dendritic cells, which modulates lymphoid and myeloid cell functions. Besides its activities on normal lymphoid cells, it has been shown that IL-21 is a growth factor for myeloma cells. In the present study, we demonstrate that IL-21 generated myeloma colonies from 9 of 24 human myeloma cell lines (HMCL) in a collagen-based assay. Of major interest, the capacity of IL-21 to stimulate clonogenicity was restricted to CD45(-) HMCL. We found that IL-21 induced tyrosine phosphorylation of STAT-3, STAT-1, and Erk1/2. Interestingly, an Akt activation was observed lately after 30 min to 1 h of IL-21 stimulation, indicating that this Akt phosphorylation could be due to an IGF-1 autocrine loop. This hypothesis was sustained both by the fact that IL-21 treatment induced an IGF-1 mRNA synthesis and that an antagonistic anti-IGF-1 receptor mAb (AVE1642) strongly inhibits the IL-21-induced clonogenicity. Thus, we demonstrated by quantitative PCR that IL-21 induced clonogenicity through an autocrine IGF-1 secretion in HMCL and primary myeloma cells. Because we have previously demonstrated that CD45 phosphatase inhibits the IGF-1 signaling, this inhibitory effect of CD45 explains why the IL-21-induced clonogenicity was restricted to CD45(-) HMCL. These results support that therapy against IGF-1R, which are presently under investigation in multiple myeloma, could be beneficial, not only to suppress IGF-1-mediated myeloma cell growth, but also IL-21-mediated myeloma cell growth.


Subject(s)
Autocrine Communication/physiology , Cell Proliferation , Insulin-Like Growth Factor I/metabolism , Interleukins/metabolism , Multiple Myeloma/metabolism , Blotting, Western , Cell Line, Tumor , Clone Cells , Flow Cytometry , Fluorescent Antibody Technique , Humans , Leukocyte Common Antigens/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
11.
Cancers (Basel) ; 12(8)2020 Aug 04.
Article in English | MEDLINE | ID: mdl-32759688

ABSTRACT

The standard model of multiple myeloma (MM) relies on genetic instability in the normal counterparts of MM cells. MM-induced lytic bone lesions are considered as end organ damages. However, bone is a tissue of significance in MM and bone changes could be at the origin/facilitate the emergence of MM. We propose the tissue disruption-induced cell stochasticity (TiDiS) theory for MM oncogenesis that integrates disruption of the microenvironment, differentiation, and genetic alterations. It starts with the observation that the bone marrow endosteal niche controls differentiation. As decrease in cellular stochasticity occurs thanks to cellular interactions in differentiating cells, the initiating role of bone disruption would be in the increase of cellular stochasticity. Thus, in the context of polyclonal activation of B cells, memory B cells and plasmablasts would compete for localizing in endosteal niches with the risk that some cells cannot fully differentiate if they cannot reside in the niche because of a disrupted microenvironment. Therefore, they would remain in an unstable state with residual proliferation, with the risk that subclones may transform into malignant cells. Finally, diagnostic and therapeutic perspectives are provided.

12.
Br J Haematol ; 145(2): 180-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19298592

ABSTRACT

Survivin is selectively expressed in most of common human cancers and is now viewed as a potent modulator of the cell death/proliferation balance in tumour cells. We previously found that myeloma cells expressed high levels of Survivin protein in correlation with disease progression and that Survivin knock-down by RNA interference decreased myeloma cell growth. We now demonstrate that Survivin overexpression promotes the proliferation and survival of human myeloma cells both in vitro and in vivo in the absence of their major growth factor, interleukin 6. Of particular interest, this effect correlates with the down regulation of Bim, a critical BH3-only cell death activator during cytokine deprivation, mainly at transcriptional level. The tight link between Survivin and Bim expression, reported for the first time here in myeloma cells and in other cell lines, is further confirmed in a panel of newly diagnosed patients with myeloma, and BIRC5 is validated as a gene significantly associated with short survival in these patients. Altogether, our findings provide evidence that Survivin directly contributes to malignant progression of myeloma and strongly suggest that targeting Survivin may disrupt the delicate balance controlling cell survival and proliferation, opening new avenues for the therapy of this still difficult-to-treat cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Multiple Myeloma/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Proliferation , Clone Cells , Gene Expression , Humans , Inhibitor of Apoptosis Proteins , Interleukin-6/metabolism , Membrane Proteins/genetics , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Neoplasm Transplantation , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Survival Rate , Survivin , Tumor Escape/immunology
13.
Breast Cancer Res Treat ; 116(3): 509-20, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19020972

ABSTRACT

Currently, no prognostic gene-expression signature (GES) established from node-positive breast cancer cohorts, able to predict evolution after systemic adjuvant chemotherapy, exists. Gene-expression profiles of 252 node-positive breast cancer patients (median follow-up: 7.7 years), mostly included in a randomized clinical trial (PACS01), receiving systemic adjuvant regimen, were determined by means of cDNA custom array. In the training cohort, we established a GES composed of 38 genes (38-GES) for the purpose of predicting metastasis-free survival. The 38-GES yielded unadjusted hazard ratio (HR) of 4.86 (95% confidence interval = 2.76-8.56). Even when adjusted with the best two clinicopathological prognostic indexes: Nottingham prognostic index (NPI) and Adjuvant!, 38-GES HRs were 3.30 (1.81-5.99) and 3.40 (1.85-6.24), respectively. Furthermore, 38-GES improved NPI and Adjuvant! classification. In particular, NPI intermediate-risk patients were divided into 2/3 close to low-risk group and 1/3 close to high-risk group (HR = 6.97 [2.51-19.36]). Similarly, Adjuvant! intermediate-risk patients were divided into 2/3 close to low-risk group and 1/3 close to high-risk group (HR = 4.34 [1.64-11.48]). The 38-GES was validated on gene-expression datasets from three external node-positive breast cancer subcohorts (n = 224) generated from different microarray platforms, with HR = 2.95 (1.74-5.01). Moreover, 38-GES showed prognostic performance in supplementary cohorts with different lymph-node status and endpoints (1,040 new patients). The 38-GES represents a robust tool able to type systemic adjuvant treated node-positive patients at high risk of metastatic relapse, and is especially powerful to refine NPI and Adjuvant! classification for those patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/secondary , Gene Expression Profiling , Lymph Nodes/pathology , Aged , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Chemotherapy, Adjuvant , Clinical Trials, Phase III as Topic , Cyclophosphamide/administration & dosage , Double-Blind Method , Epirubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Lymph Nodes/drug effects , Lymphatic Metastasis , Multicenter Studies as Topic , Oligonucleotide Array Sequence Analysis , Postmenopause , Prognosis , Survival Rate , Treatment Outcome
14.
Haematologica ; 94(1): 87-93, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001278

ABSTRACT

BACKGROUND: XIAP is the best characterized and the most potent direct endogenous caspase inhibitor and is considered a key actor in the control of apoptotic threshold in cancer cells. In this report, we specifically addressed XIAP regulation and function in myeloma cells. DESIGN AND METHODS: XIAP and its endogenous inhibitor XAF-1 protein levels and their regulation were assessed by immunoblot analysis in myeloma cell lines or primary myeloma cells. XIAP knockdown by RNA interference was used to evaluate XIAP impact on in vitro drug sensitivity and in vivo tumor growth. RESULTS: Our results indicate that myeloma cells expressed high levels of XIAP protein that were tightly regulated during growth factor stimulation or stress condition. Of note, an increased XIAPlevel was evidenced during the blockade of the canonical cap-dependent translation by the mTOR inhibitor rapamycin, supporting the hypothesis of a functional IRES sequence in XIAP mRNA. In addition, caspase-mediated XIAP cleavage correlated to an apoptotic process occurring upon cell treatment with the proteasome inhibitor bortezomib. Importantly, XIAP knockdown using RNA interference enhanced drug sensitivity and decreased tumor formation in NOD/SCID mice. Finally, myeloma cells also expressed the XIAP inhibitor XAF-1 that interacted with XIAP in viable myeloma cells. CONCLUSIONS: Altogether, our data argue for a delicate control of XIAP function in myeloma cells and stimulate interest in targeting XIAP in myeloma treatment.


Subject(s)
Multiple Myeloma/metabolism , Multiple Myeloma/pathology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm/drug effects , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Mice, SCID , Multiple Myeloma/genetics , RNA Interference , X-Linked Inhibitor of Apoptosis Protein/genetics , Xenograft Model Antitumor Assays
15.
Int Immunol ; 20(6): 775-82, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18397910

ABSTRACT

The aim of this in vitro study was to evaluate if osteoclasts (OCs) and dendritic cells (DCs), both of monocyte origin, can support the survival of normal human plasma cells (PCs). PCs differentiate from plasmablasts (PBs) arising from activated B cells, essentially memory B cells. To study the survival of both PBs (CD20(low)CD38(high)CD138(neg)) and PCs (CD20(neg)CD38(bright)CD138(bright)), we generated pre-PBs (CD20(low)CD38(pos)CD138(neg)) from CD40-activated B cells (CD20(high)CD38(neg)CD138(neg)) and cultured them on DCs or OCs in the presence of added IL-6. By quantitative and qualitative study, we showed that DCs support the survival of PBs and early PCs, but not that of PCs. In contrast, OCs support the survival of PBs, early PCs and PCs. PCs surviving on OCs 12 days after pre-PB input display phenotypic features of bone marrow PCs, CD138(bright)CD38(bright)HLA-DR(low)CD45(dim). The ability for OCs to support the survival of PCs was fully dependent on cell-cell contact and not inhibited by BCMA-Fc suggesting that secreted BAFF and APRIL were not involved.


Subject(s)
Antigens, CD/biosynthesis , Dendritic Cells/cytology , Osteoclasts/cytology , Plasma Cells/cytology , Antigens, CD/immunology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , Cell Communication , Cell Differentiation/immunology , Cell Separation , Cell Survival/immunology , Coculture Techniques , Contact Inhibition/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immunomagnetic Separation , Lymphocyte Activation , Osteoclasts/immunology , Osteoclasts/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism
16.
Cancer Res ; 67(11): 5418-24, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545623

ABSTRACT

Targeting the ubiquitin-proteasome pathway has emerged as a potent anticancer strategy. Bortezomib, a specific proteasome inhibitor, has been approved for the treatment of relapsed or refractory multiple myeloma. Multiple myeloma cell survival is highly dependent on Mcl-1 antiapoptotic molecules. In a recent study, proteasome inhibitors induced Mcl-1 accumulation that slowed down their proapoptotic effects. Consequently, we investigated the role of Bcl-2 family members in bortezomib-induced apoptosis. We found that bortezomib induced apoptosis in five of seven human myeloma cell lines (HMCL). Bortezomib-induced apoptosis was associated with Mcl-1 cleavage regardless of Mcl-1L accumulation. Furthermore, RNA interference mediated Mcl-1 decrease and sensitized RPMI-8226 HMCL to bortezomib, highlighting the contribution of Mcl-1 in bortezomib-induced apoptosis. Interestingly, an important induction of Noxa was found in all sensitive HMCL both at protein and mRNA level. Concomitant to Mcl-1 cleavage and Noxa induction, we also found caspase-3, caspase-8, and caspase-9 activation. Under bortezomib treatment, Mcl-1L/Noxa complexes were highly increased, Mcl-1/Bak complexes were disrupted, and there was an accumulation of free Noxa. Finally, we observed a dissociation of Mcl-1/Bim complexes that may be due to a displacement of Bim induced by Noxa. Thus, in myeloma cells, the mechanistic basis for bortezomib sensitivity can be explained mainly by the model in which the sensitizer Noxa can displace Bim, a BH3-only activator, from Mcl-1, thus leading to Bax/Bak activation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Multiple Myeloma/drug therapy , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrazines/pharmacology , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Bortezomib , Cell Line, Tumor , Down-Regulation , Humans , Membrane Proteins/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , RNA, Small Interfering/genetics , Up-Regulation/drug effects , bcl-2 Homologous Antagonist-Killer Protein/metabolism
17.
Eur J Intern Med ; 68: 51-54, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31387843

ABSTRACT

The International Staging System (ISS) -calculated from serum albumin and beta-2 microglobulin (ß2m)- is an established prognostic marker in multiple myeloma (MM), which has also been suggested to account for survival among general senior population. Our objective was to examine long-term survival of older women free of MM according to baseline ISS. The study included 230 community-dwelling healthy older women without known MM from the EPIDOS cohort (mean ±â€¯SD, 80.4 ±â€¯3.4 years). Serum albumin and ß2m were measured at baseline, and used to calculate the ISS a posteriori. Abnormal ISS was defined as ISS = 2 or ISS = 3, although ISS = 1 was considered normal. The vital status was sought after a mean follow-up of 17.6 ±â€¯0.2 years (range, 16.8-18.3). Age, body mass index, mean arterial pressure, diabetes mellitus, hypertension, coronary heart disease, stroke, use corticosteroids, number of drugs daily taken, smoking, physical activity, fall history, bone mineral density, and creatinine clearance were used as potential confounders. All participants died during the 17-year follow-up. Compared to women with normal ISS, those with abnormal ISS (n = 24) had shorter survival time (4.9 ±â€¯4.3 versus 8.7 ±â€¯5.2 years, P = .001) and died earlier (85.6 ±â€¯4.8 versus 89.1 ±â€¯5.6 years old, P = .003). Survival time after blood test correlated with both serum albumin (r = 0.16, P = .015) and ß2m (r = -0.27, P < .001). Cox regression revealed that abnormal ISS was associated with mortality (adjusted HR = 3.03, P < .001). Kaplan-Meier distributions showed that participants with abnormal ISS died earlier than those with normal ISS (log-rank P < .001). In conclusion, community-dwelling older women with abnormal ISS had shorter survival time than the others, suggesting that ISS could be considered as a universal prognostic "aging system" rather than a specific MM "staging system".


Subject(s)
Geriatric Assessment/methods , Serum Albumin/analysis , Aged , Biomarkers/blood , Female , Humans , Kaplan-Meier Estimate , Prognosis , Survival Analysis , beta 2-Microglobulin/blood
18.
Leuk Res ; 32(3): 379-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17767956

ABSTRACT

CD117 (c-kit) was evaluated on normal plasma cells (PC) (n=10), PC of individuals with monoclonal gammopathy of undetermined significance (MGUS, n=12), malignant PC from patients with multiple myeloma (MM) either at diagnosis (n=83) or relapse (n=38) and on 23 human myeloma cell lines (HMCL). Whereas CD117 is never expressed in normal PC, it is expressed in 50% of MGUS (p=0.015). Furthermore, 33% of MM at diagnosis do express CD117, as opposed to 8% of those in relapse (p=0.003). Finally, CD117 was never found in HMCL. CD117 expression was associated with a better prognosis: overall survival was 93% at 4 years in CD117+ MM versus 64% in CD117- MM (p=0.05). Conversely, lack of CD117, but also high beta-2 microglobulin, t(4;14) and CD221 (IGF-1R) expression were associated with a poorer prognosis. Multivariate analysis revealed that CD117- patients were those with CD221 and t(4;14) and had the poorest prognosis. In conclusion, CD117 (c-kit) is aberrantly expressed on a subset of MGUS and MM with a more indolent presentation and is functionally antinomic to CD221 (IGF-1R). CD117 expression could be related to a specific oncogenic pathway in MM.


Subject(s)
Multiple Myeloma/immunology , Paraproteinemias/immunology , Proto-Oncogene Proteins c-kit/metabolism , Aged , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Prognosis , Survival Analysis
19.
Clin Cancer Res ; 13(20): 6010-8, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17947462

ABSTRACT

PURPOSE: Intrinsic activation of nuclear factor kappaB (NF-kappaB) characterizes various hematologic malignancies. In this study, we specifically address the role of NF-kappaB blockade in mediated antimyeloma activity using the IkappaB kinase-2 pharmacologic inhibitor, AS602868. EXPERIMENTAL DESIGN: Human myeloma cell lines (n = 16) and primary myeloma cells (n = 10) were tested for their sensitivity to AS602868 in terms of proliferation and apoptosis. Both in vitro and in vivo experiments were conducted. Functional mechanisms regarding the apoptotic pathways triggered by AS602868 were studied. The potential proapoptotic synergy between AS602868 and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was also evaluated. RESULTS: Our results show that AS602868 efficiently targeted the canonical NF-kappaB pathway in myeloma cells and potently inhibited their growth in inducing apoptosis through Bax and caspase-3 activation. AS602868 also induced apoptosis in primary myeloma cells even in the presence of bone marrow mononuclear cells. Moreover, the IkappaB kinase-2 inhibitor targeted the paracrine effect on the bone marrow environment. Indeed, it decreased the intrinsic and myeloma-induced secretion of interleukin-6 from bone marrow stromal cells. In addition, AS602868 inhibited myeloma cell growth in the MM.1S xenograft myeloma model. Of particular interest, AS602868 strongly increased myeloma sensitivity to TRAIL in blocking TRAIL-induced NF-kappaB activation and in decreasing the expression of antiapoptotic proteins such as cFLIP and cIAP-1/2. CONCLUSIONS: Taken together, our data point out the interest to inhibit the canonical NF-kappaB pathway in myeloma and clearly encourage clinical evaluation of novel therapies based on targeting NF-kappaB, especially in combination with TRAIL.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Multiple Myeloma/pathology , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Interleukin-6/metabolism , Mice , Models, Biological , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neoplasm Transplantation , Pyrimidines/pharmacology , Receptors, Immunologic/metabolism , bcl-2-Associated X Protein/metabolism
20.
Med Sci (Paris) ; 24(10): 821-7, 2008 Oct.
Article in French | MEDLINE | ID: mdl-18950577

ABSTRACT

Discovered 10 years ago, survivin has a dual role in the smooth progress of mitosis and in apoptosis resistance. Survivin plays an important physiological role in development, but is absent in differentiated adult tissues. In contrast, aberrant survivin expression is found in most human cancers because of the activation of various signalling pathways. A complex survivin network appears to intersect multiple pathways in cell biology, related to several molecular partners and fine subcellular localizations. Based on its pro-oncogenic properties, basic and translational studies have shown a growing interest in survivin that has led to consider survivin as a prognostic marker and a promising target for anti-tumoral therapies.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Microtubule-Associated Proteins/physiology , Neoplasm Proteins/physiology , Animals , Animals, Genetically Modified , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Apoptosis Regulatory Proteins/therapeutic use , Biomarkers, Tumor , Cancer Vaccines/therapeutic use , Cell Cycle/physiology , Clinical Trials, Phase I as Topic , Drug Delivery Systems , Drug Screening Assays, Antitumor , Embryonic Development/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Inhibitor of Apoptosis Proteins , Models, Biological , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Fusion Proteins/therapeutic use , Subcellular Fractions/metabolism , Survivin , tat Gene Products, Human Immunodeficiency Virus/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL