Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Oral Oncol ; 149: 106680, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38218022

ABSTRACT

OBJECTIVES: In head and neck squamous cell carcinoma (HNSCC), PD-1/PD-L1 inhibitors remain inefficient in most patients, which points to the need for better characterization of immune checkpoint (ICP) molecule expression. MATERIAL AND METHODS: We evaluated the expression of 22 ICP ligands (ICPL) in 2,176 malignant cells from 10 patients in a public single-cell RNA-sequencing dataset and in two cohorts of HNSCC patients for which gene expression data are available. RESULTS: Based on ICPL expression, malignant cells formed three distinct clusters characterized either by a strong expression of ICPL together with an immune phenotype linked to IFN-γ response (cluster 1) or by a weak ICPL expression and little response to IFN-γ (clusters 2 and 3). Malignant cells from cluster 3 showed a high PD-L1 expression associated with NRF2 signature. The relevance of 3 groups of patients, i.e "high ICPL/high IFN-γ", "low ICPL/low IFN-γ" or "low ICPL/low IFN-γ/high PD-L1" was confirmed in a cohort of 259 OSCC whole tumor samples from TCGA and in the CLB-IHN cohort including patients treated with PD1/PD-L1 inhibitors. The heterogeneous expression of ICPL among patients' malignant cells was associated with immunologically distinct microenvironments, evaluated with the "hot/cold" and the Tumor microenvironment (TME) classification. Finally, the "low ICPL/low IFN-γ/high PD-L1" group 3 displayed a poor prognosis in the TCGA cohort. CONCLUSION: Hence, the global picture of ICPL gene expression in malignant cells from HNSCC patients may contribute to the broader issue of improving immunotherapy strategies though a better stratification of patients and the design of new treatment combinations.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Carcinoma, Squamous Cell/pathology , B7-H1 Antigen/metabolism , Mouth Neoplasms/genetics , Immune Checkpoint Inhibitors , Phenotype , Tumor Microenvironment/genetics , RNA
2.
Cancer Lett ; 472: 29-39, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31838086

ABSTRACT

Toll-like receptor 3 (TLR3) is an immune receptor that behaves like a death receptor in tumor cells, thereby providing an original target for cancer therapy. The therapeutic potential of TLR3 targeting in malignant mesothelioma, an aggressive and incurable neoplasia of the pleura and peritoneum, has so far not been addressed. We investigated TLR3 expression and sensitivity of human mesothelioma cell lines to the synthetic dsRNA Poly(I:C), alone or in combination with cisplatin, the gold standard chemotherapy in mesothelioma. Activation of TLR3 by Poly(I:C) induced apoptosis of 4/8 TLR3-positive cell lines but not of TLR3-negative cell lines. The combined cisplatin/Poly(I:C) treatment enhanced apoptosis of 3/4 Poly(I:C)-sensitive cell lines and overcame resistance to Poly(I:C) or cisplatin alone in 2/4 cell lines. Efficacy of the combined treatment relied on cisplatin-induced downregulation of c-FLIP, the main regulator of the extrinsic apoptotic pathway, leading to an enhanced caspase-8-mediated pathway. Of note, 6/6 primary cell samples isolated from patients with peritoneal mesothelioma expressed TLR3. Patient-derived cells were sensitive to Poly(I:C) alone while the combined cisplatin/Poly(I:C) treatment induced dramatic cell death. Our findings demonstrate that TLR3 targeting in combination with cisplatin presents an innovative therapeutic strategy in mesothelioma.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Toll-Like Receptor 3/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Caspase 8/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Mesothelioma/genetics , Mesothelioma/physiopathology , Mesothelioma, Malignant , Poly I-C/pharmacology , Signal Transduction/drug effects
3.
J Lipid Res ; 56(6): 1110-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25833686

ABSTRACT

Interleukin 17A (IL-17A) is a proinflammatory cytokine involved in the pathogenesis of chronic inflammatory diseases. In the field of immunometabolism, we have studied the impact of IL-17A on the lipid metabolism of human in vitro-generated monocyte-derived dendritic cells (DCs). Microarrays and lipidomic analysis revealed an intense remodeling of lipid metabolism induced by IL-17A in DCs. IL-17A increased 2-12 times the amounts of phospholipids, cholesterol, triglycerides, and cholesteryl esters in DCs. Palmitic (16:0), stearic (18:0), and oleic (18:ln-9c) acid were the main fatty acid chains present in DCs. They were strongly increased in response to IL-17A while their relative proportion remained unchanged. Capture of extracellular lipids was the major mechanism of lipid droplet accumulation, visualized by electron microscopy and Oil Red O staining. Besides this foamy phenotype, IL-17A induced a mixed macrophage-DC phenotype and expression of the nuclear receptor NR1H3/liver X receptor-α, previously identified in the context of atherosclerosis as the master regulator of cholesterol homeostasis in macrophages. These IL-17A-treated DCs were as competent as untreated DCs to stimulate allogeneic naive T-cell proliferation. Following this first characterization of lipid-rich DCs, we propose to call these IL-17A-dependent cells "foamy DCs" and discuss the possible existence of foamy DCs in atherosclerosis, a metabolic and inflammatory disorder involving IL-17A.


Subject(s)
Atherosclerosis/metabolism , Dendritic Cells/metabolism , Fatty Acids/metabolism , Interleukin-17/metabolism , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Dendritic Cells/immunology , Foam Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Lipid Droplets/immunology , Lipid Droplets/metabolism , Lipid Metabolism/immunology , Liver X Receptors , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Orphan Nuclear Receptors/biosynthesis , Orphan Nuclear Receptors/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Cancer Res ; 74(22): 6554-64, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25371412

ABSTRACT

The classic model of tumor suppression implies that malignant transformation requires full "two-hit" inactivation of a tumor-suppressor gene. However, more recent work in mice has led to the proposal of a "continuum" model that involves more fluid concepts such as gene dosage-sensitivity and tissue specificity. Mutations in the tumor-suppressor gene von Hippel-Lindau (VHL) are associated with a complex spectrum of conditions. Homozygotes or compound heterozygotes for the R200W germline mutation in VHL have Chuvash polycythemia, whereas heterozygous carriers are free of disease. Individuals with classic, heterozygous VHL mutations have VHL disease and are at high risk of multiple tumors (e.g., CNS hemangioblastomas, pheochromocytoma, and renal cell carcinoma). We report here an atypical family bearing two VHL gene mutations in cis (R200W and R161Q), together with phenotypic analysis, structural modeling, functional, and transcriptomic studies of these mutants in comparison with classical mutants involved in the different VHL phenotypes. We demonstrate that the complex pattern of disease manifestations observed in VHL syndrome is perfectly correlated with a gradient of VHL protein (pVHL) dysfunction in hypoxia signaling pathways. Thus, by studying naturally occurring familial mutations, our work validates in humans the "continuum" model of tumor suppression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Carcinogenesis/metabolism , Mutation , Signal Transduction/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Carcinoma, Renal Cell/genetics , Humans , Kidney Neoplasms/genetics , Molecular Dynamics Simulation , Pheochromocytoma/genetics , Polymorphism, Single Nucleotide
5.
PLoS One ; 7(2): e30359, 2012.
Article in English | MEDLINE | ID: mdl-22363428

ABSTRACT

The Wnt/ß-catenin pathway plays a crucial role in the pathogenesis of various human cancers. In multiple myeloma (MM), aberrant auto-and/or paracrine activation of canonical Wnt signaling promotes proliferation and dissemination, while overexpression of the Wnt inhibitor Dickkopf1 (DKK1) by MM cells contributes to osteolytic bone disease by inhibiting osteoblast differentiation. Since DKK1 itself is a target of TCF/ß-catenin mediated transcription, these findings suggest that DKK1 is part of a negative feedback loop in MM and may act as a tumor suppressor. In line with this hypothesis, we show here that DKK1 expression is low or undetectable in a subset of patients with advanced MM as well as in MM cell lines. This absence of DKK1 is correlated with enhanced Wnt pathway activation, evidenced by nuclear accumulation of ß-catenin, which in turn can be antagonized by restoring DKK1 expression. Analysis of the DKK1 promoter revealed CpG island methylation in several MM cell lines as well as in MM cells from patients with advanced MM. Moreover, demethylation of the DKK1 promoter restores DKK1 expression, which results in inhibition of ß-catenin/TCF-mediated gene transcription in MM lines. Taken together, our data identify aberrant methylation of the DKK1 promoter as a cause of DKK1 silencing in advanced stage MM, which may play an important role in the progression of MM by unleashing Wnt signaling.


Subject(s)
DNA Methylation/genetics , Gene Silencing , Intercellular Signaling Peptides and Proteins/genetics , Multiple Myeloma/genetics , Promoter Regions, Genetic , Transcription, Genetic , Wnt Proteins/antagonists & inhibitors , Azacitidine/pharmacology , Base Sequence , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , DNA Methylation/drug effects , Disease Progression , Gene Silencing/drug effects , Humans , Molecular Sequence Data , Multiple Myeloma/pathology , Neoplasm Staging , Transcription, Genetic/drug effects , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
6.
Biochim Biophys Acta ; 1806(2): 208-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20655987

ABSTRACT

Hepatocyte growth factor (HGF) and its receptor MET are essential during embryonic development and throughout postnatal life. However, aberrant MET activation, due to overexpression, mutations, or autocrine ligand production, contributes to the development and progression of a variety of human cancers, often being associated with poor clinical outcome and drug resistance. B cell malignancies arise from B cells that are clonally expanded at different stages of differentiation. Despite major therapeutic advances, most mature B cell malignancies remain incurable and biologically-oriented therapeutic strategies are urgently needed. This review addresses the role of the HGF/MET pathway during B cell development and discusses how its aberrant activation contributes to the development of B cell lymphoproliferative disorders, with particular emphasis on multiple myeloma and diffuse large B cell lymphoma. These insights, combined with the recent development of clinical-grade agents targeting the MET pathway, provide the rationale to envision the HGF/MET pathway as a new promising target for the treatment of B cell malignancies.


Subject(s)
Hepatocyte Growth Factor/physiology , Lymphoma, B-Cell/etiology , Multiple Myeloma/etiology , Proto-Oncogene Proteins c-met/physiology , Receptors, Growth Factor/physiology , Signal Transduction/physiology , Cell Differentiation , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/chemistry , Humans , Lymphoma, B-Cell/drug therapy , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/chemistry , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/chemistry
7.
BMC Cancer ; 10: 198, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20465808

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is characterized by a strong dependence of the tumor cells on their microenvironment, which produces growth factors supporting survival and proliferation of myeloma cells (MMC). In the past few years, many myeloma growth factors (MGF) have been described in the literature. However, their relative importance and the nature of the cells producing MGF remain unidentified for many of them. METHODS: We have analysed the expression of 51 MGF and 36 MGF receptors (MGFR) using Affymetrix microarrays throughout normal plasma cell differentiation, in MMC and in cells from the bone marrow (BM) microenvironment (CD14, CD3, polymorphonuclear neutrophils, stromal cells and osteoclasts). RESULTS: 4/51 MGF and 9/36 MGF-receptors genes were significantly overexpressed in plasmablasts (PPC) and BM plasma cell (BMPC) compared to B cells whereas 11 MGF and 11 MGFR genes were overexpressed in BMPC compared to PPC. 3 MGF genes (AREG, NRG3, Wnt5A) and none of the receptors were significantly overexpressed in MMC versus BMPC. Furthermore, 3/51 MGF genes were overexpressed in MMC compared to the the BM microenvironment whereas 22/51 MGF genes were overexpressed in one environment subpopulation compared to MMC. CONCLUSIONS: Two major messages arise from this analysis 1) The majority of MGF genes is expressed by the bone marrow environment. 2) Several MGF and their receptors are overexpressed throughout normal plasma cell differentiation. This study provides an extensive and comparative analysis of MGF expression in plasma cell differentiation and in MM and gives new insights in the understanding of intercellular communication signals in MM.


Subject(s)
Bone Marrow Cells/chemistry , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Plasma Cells/chemistry , RNA, Messenger/analysis , Bone Marrow Cells/pathology , Case-Control Studies , Cell Differentiation/genetics , Humans , Middle Aged , Multiple Myeloma/pathology , Neoplasm Staging , Plasma Cells/pathology , Receptors, Growth Factor/genetics , Up-Regulation
8.
Eur J Haematol ; 83(2): 119-29, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19456850

ABSTRACT

BLyS and APRIL share two receptors - transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) and B-cell maturation antigen (BCMA) - and BLyS binds to a third receptor, BAFF-R. We previously reported that TACI gene expression is a good indicator of a BLyS-binding receptor in human multiple myeloma cell lines (HMCLs), unlike BCMA, which is expressed by all HMCLs or BAFF-R which is typically not expressed by late-stage B cells. We hypothesised a link between APRIL and TACI through syndecan-1, similar to the situation reported for FGF and FGFR. We observed very strong binding of APRIL, but not BLyS, at the surface of all syndecan-1(+) HMCLs and primary multiple myeloma cells (MMC). All syndecan-1(+) HMCLs and MMC could also bind TACI-Fc, but not BCMA-Fc or BAFF-R-Fc molecules. Binding of APRIL or TACI-Fc was abrogated by heparin or cell pretreatment with heparitinase, which cleaves heparan sulfate chains. The growth factor activity of APRIL on MMC was also inhibited by heparin. Our data identify syndecan-1 as a co-receptor for APRIL and TACI at the cell surface of MMC, promoting the activation of an APRIL/TACI pathway that induces survival and proliferation in MMC.


Subject(s)
Cell Membrane/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Syndecan-1/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Flow Cytometry , Heparin/pharmacology , Humans , Multiple Myeloma/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transmembrane Activator and CAML Interactor Protein/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
9.
Br J Haematol ; 145(3): 350-68, 2009 May.
Article in English | MEDLINE | ID: mdl-19298595

ABSTRACT

Syndecan-1 is a proteoglycan that concentrates heparin-binding factors on the surface of multiple myeloma cells, and probably plays a major role in multiple myeloma biology. As heparan sulphate and chondroitin sulphate are the bioactive components of syndecan-1, we analysed the signature of genes encoding 100 proteins involved in synthesis of these chains, i.e. from precursor uptake to post-translational modifications, using Affymetrix microarrays. The expression of enzymes required for heparan sulphate and chondroitin sulphate biosynthesis was shown to increase in parallel with syndecan-1 expression, throughout the differentiation of memory B cells into plasmablasts and normal bone marrow plasma cells. Sixteen genes were significantly different between normal and malignant plasma cells, nine of these genes -EXT2, CHSY3, CSGALNACT1, HS3ST2, HS2ST1, CHST11, CSGALNACT2, HPSE, SULF2 - encode proteins involved in glycosaminoglycan chain synthesis or modifications. Kaplan-Meier analysis was performed in two independent series of patients: B4GALT7, CSGALNACT1, HS2ST1 were associated with a good prognosis whereas EXT1 was linked to a bad prognosis. This study provides an overall picture of the major genes encoding for proteins involved in heparan sulphate and chondroitin sulphate synthesis and modifications that can be implicated in normal and malignant plasma cells.


Subject(s)
Chondroitin Sulfates/biosynthesis , Gene Expression Profiling , Heparitin Sulfate/biosynthesis , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Plasma Cells/metabolism , B-Lymphocytes/metabolism , Case-Control Studies , Cell Line, Tumor , Gene Expression , Gene Expression Profiling/methods , Humans , Immunologic Memory , Kaplan-Meier Estimate , Multiple Myeloma/enzymology , Multiple Myeloma/immunology , Principal Component Analysis , Prognosis , Protein Processing, Post-Translational , Syndecan-1/metabolism
10.
Blood ; 114(1): 128-43, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19299335

ABSTRACT

Abundant bone marrow angiogenesis is present in almost all myeloma patients requiring therapy and correlated to treatment response and survival. We assessed the expression of 402 angiogenesis-associated genes by Affymetrix DNA microarrays in 466 samples, including CD138-purified myeloma cells (MMCs) from 300 previously untreated patients, in vivo microcirculation by dynamic contrast-enhanced magnetic resonance imaging, and in vitro angiogenesis (AngioKit-assay). Normal bone marrow plasma cells (BMPCs) express a median of 39 proangiogenic (eg, VEGFA, ADM, IGF-1) and 28 antiangiogenic genes (eg, TIMP1, TIMP2). Supernatants of BMPCs unlike those of memory B cells induce angiogenesis in vitro. MMCs do not show a significantly higher median number of expressed proangiogenic (45) or antiangiogenic (31) genes, but 97% of MMC samples aberrantly express at least one of the angiogenic factors HGF, IL-15, ANG, APRIL, CTGF, or TGFA. Supernatants of MMCs and human myeloma cell lines induce significantly higher in vitro angiogenesis compared with BMPCs. In conclusion, BMPCs express a surplus of proangiogenic over antiangiogenic genes transmitting to the ability to induce in vitro angiogenesis. Aberrant expression of proangiogenic and down-regulation of antiangiogenic genes by MMCs further increases the angiogenic stimulus, together leading to bone marrow angiogenesis at various degrees in all myeloma patients.


Subject(s)
Multiple Myeloma/blood supply , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Plasma Cells/pathology , Plasma Cells/physiology , Angiogenic Proteins/genetics , Angiostatic Proteins/genetics , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Bone Marrow/blood supply , Case-Control Studies , Cell Line, Tumor , Gene Expression , Gene Expression Profiling , Humans , In Vitro Techniques , Models, Biological , Multiple Myeloma/genetics , Neovascularization, Physiologic/genetics , Oligonucleotide Array Sequence Analysis
11.
Blood ; 113(19): 4614-26, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19228610

ABSTRACT

A plethora of myeloma growth factors (MGFs) has been identified, but their relative importance and cooperation have not been determined. We investigated 5 MGFs (interleukin-6 [IL-6], insulin-like growth factor type 1 [IGF-1], hepatocyte growth factor [HGF], HB-epidermal growth factor [HB-EGF], and a proliferation-inducing ligand [APRIL]) in serum-free cultures of human myeloma cell lines (HMCLs). In CD45(-) HMCLs, an autocrine IGF-1 loop promoted autonomous survival whereas CD45(+) HMCLs could not survive without addition of MGFs, mainly IGF-1 and IL-6. IGF-1 was the major one: its activity was abrogated by an IGF-1R inhibitor only, whereas IL-6, HGF, or HB-EGF activity was inhibited by both IGF-1R- and receptor-specific inhibition. APRIL activity was inhibited by its specific inhibitor only. Of the investigated MGFs and their receptors, only expressions of IGF-1R and IL-6R in multiple myeloma cells (MMCs) of patients delineate a group with adverse prognosis. This is mainly explained by a strong association of IGF-1R and IL-6R expression and t(4;14) translocation, but IGF-1R expression without t(4;14) can also have a poor prognosis. Thus, IGF-1-targeted therapy, eventually in combination with anti-IL-6 therapy, could be promising in a subset of patients with MMCs expressing IGF-1R.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Multiple Myeloma/pathology , Receptor, IGF Type 1/metabolism , Blotting, Western , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Culture Media, Serum-Free , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Heparin-binding EGF-like Growth Factor , Hepatocyte Growth Factor/pharmacology , Humans , Immunoblotting , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Interleukin-6/pharmacology , Leukocyte Common Antigens/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic , Tumor Necrosis Factor Ligand Superfamily Member 13/pharmacology
12.
Clin Cancer Res ; 13(24): 7289-95, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18094409

ABSTRACT

Multiple myeloma is a B-cell neoplasia characterized by the proliferation of a clone of malignant plasma cells in the bone marrow. We review here the input of gene expression profiling of myeloma cells and of their tumor microenvironment to develop new tumor classifiers, to better understand the biology of myeloma cells, to identify some mechanisms of drug sensitivity and resistance, to identify new myeloma growth factors, and to depict the complex interactions between tumor cells and their microenvironment. We discuss how these findings may improve the clinical outcome of this still incurable disease.


Subject(s)
Gene Expression Profiling/methods , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Gene Expression , Humans , Multiple Myeloma/classification , Multiple Myeloma/metabolism
13.
Br J Haematol ; 138(2): 160-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17542984

ABSTRACT

The pathophysiologic basis for multiple myeloma (MM) has been attributed to the dysregulation of various paracrine or autocrine growth factor loops and to perturbations in several signal transduction pathways including IkappaB kinase/nuclear factor-kappaB (IKK/NF-kappaB). The present study aimed at investigating the effect of a pharmaceutical IKK2 inhibitor, the anilinopyrimidine derivative AS602868, on the in vitro growth of 14 human MM cell lines (HMCL) and primary cells from 13 patients. AS602868 induced a clear dose-dependent inhibition of MM cell growth on both HMCL and primary MM cells, which was the result of a simultaneous induction of apoptosis and inhibition of the cell cycle progression. Combination of AS602868 with suboptimal doses of melphalan or Velcade showed an additive effect in growth inhibition of HMCL. AS602868 also induced apoptosis of primary myeloma cells. Importantly, AS602868 did not alter the survival of other bone marrow mononuclear cells (CD138(-)) co-cultured with primary MM (CD138(+)) cells, except for CD34(+) haematopoietic stem cells. The results demonstrate the important role of NF-kappaB in maintaining the survival of MM cells and suggest that a pharmacological inhibition of the NF-kappaB pathway by the IKK2 inhibitor AS602868 can efficiently kill HMCL and primary myeloma cells and therefore might represent an innovative approach for treating MM patients.


Subject(s)
Antineoplastic Agents/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Multiple Myeloma/physiopathology , Pyrimidines/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Humans , Melphalan/pharmacology , NF-kappa B/drug effects , Pyrazines/pharmacology
14.
Blood ; 109(11): 4914-23, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17339423

ABSTRACT

The heparan sulfate (HS) proteoglycan, syndecan-1, plays a major role in multiple myeloma (MM) by concentrating heparin-binding growth factors on the surface of MM cells (MMCs). Using Affymetrix microarrays and real-time reverse transcriptase-polymerase chain reaction (RT-PCR), we show that the gene encoding heparanase (HPSE), an enzyme that cleaves HS chains, is expressed by 11 of 19 myeloma cell lines (HMCLs). In HSPE(pos) HMCLs, syndecan-1 gene expression and production of soluble syndecan-1, unlike expression of membrane syndecan-1, were significantly increased. Knockdown of HPSE by siRNA resulted in a decrease of syndecan-1 gene expression and soluble syndecan-1 production without affecting membrane syndecan-1 expression. Thus, HPSE influences expression and shedding of syndecan-1. Contrary to HMCLs, HPSE is expressed in only 4 of 39 primary MMC samples, whereas it is expressed in 36 of 39 bone marrow (BM) microenvironment samples. In the latter, HPSE is expressed at a median level in polymorphonuclear cells and T cells; it is highly expressed in monocytes and osteoclasts. Affymetrix data were validated at the protein level, both on HMCLs and patient samples. We report for the first time that a gene's expression mainly in the BM environment (ie, HSPE) is associated with a shorter event-free survival of patients with newly diagnosed myeloma treated with high-dose chemotherapy and stem cell transplantation. Our study suggests that clinical inhibitors of HPSE could be beneficial for patients with MM.


Subject(s)
Bone Marrow/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glucuronidase/physiology , Multiple Myeloma/enzymology , Syndecan-1/biosynthesis , Aged , Bone Marrow Cells/metabolism , Cell Line, Tumor , Humans , Middle Aged , Monocytes/metabolism , Multiple Myeloma/diagnosis , Multiple Myeloma/pathology , Neutrophils/metabolism , Osteoclasts/metabolism , Prognosis
15.
Immunol Rev ; 210: 86-104, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16623766

ABSTRACT

Plasma cells (PCs) develop from B lymphocytes following stimulation by antigen and express a genetic program aimed at the synthesis of immunoglobulins. This program includes the induction of genes coding for transcription factors such as PRDM1, X-box-binding protein 1 and BHLHB3, cell-surface molecules such as CD138/syndecan-1, and for the unfolded protein response. We review how the microarray technology has recently contributed to the understanding of the biology of this rare but essential cell population and its transformation into premalignant and malignant PCs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Paraproteinemias/genetics , Plasma Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/genetics , Humans , Plasma Cells/cytology , Plasma Cells/metabolism
16.
Eur Cytokine Netw ; 16(1): 57-64, 2005.
Article in English | MEDLINE | ID: mdl-15809207

ABSTRACT

Primary myeloma cells rapidly apoptose as soon as they are removed from their bone-marrow environment. A likely explanation is that the tumor environment produces survival factors that may counteract a spontaneous activation of pro-apoptotic program. Additional factors may trigger cell cycling in surviving myeloma cells. Interleukin-6 (IL-6) is a well recognized myeloma cell growth factor produced mainly by the tumor environment. However, myeloma cells themselves may produce low levels of autocrine IL-6. The respective roles of paracrine versus autocrine IL-6 are a matter of debate. We investigated these roles using the XG-6 myeloma cell line whose growth is dependent on addition of exogenous IL-6, despite its weak IL-6 mRNA and protein expression. The apoptosis induced by exogenous IL-6 deprivation was blocked by transferring the Mcl-1 gene coding for an anti-apoptotic protein in XG-6 cells. An XG-6Mcl-1 cell line which can survive and grow without adding IL-6 was obtained. We show that anti-IL-6 or anti-gp130 antibodies abrogated cell cycling whereas they did not affect cell survival. These data indicate that the weak autocrine IL-6 produced by myeloma cells is sufficient to trigger cell cycling whereas their survival requires large exogenous IL-6 concentrations. This important role of autocrine IL-6 has to be considered when evaluating the mechanism of action of myeloma cell growth factors. These factors may actually block an activated pro-apoptotic program, making possible a weak production of autocrine IL-6 to promote cell cycling.


Subject(s)
Cell Cycle , Cell Survival , Interleukin-6/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Antibodies, Monoclonal/pharmacology , Apoptosis , Autocrine Communication , Cell Line, Tumor , Humans , Interleukin-6/genetics , Interleukin-6/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Paracrine Communication , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
17.
Blood ; 106(3): 1021-30, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15827134

ABSTRACT

B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) have been shown to promote multiple myeloma (MM) cell growth. We show that the main site of production for BAFF and APRIL is the bone marrow (BM) environment, and that production is mainly by monocytes and neutrophils. In addition, osteoclasts produce very high levels of APRIL, unlike BM stromal cells. Myeloma cells (MMCs) express TACI (transmembrane activator and calcium modulator and cyclophilin ligand interactor), the receptor of BAFF/APRIL, at varying levels. TACI expression is a good indicator of a BAFF-binding receptor. Expression data of purified MMCs from 65 newly diagnosed patients have been generated using Affymetrix microarrays and were analyzed by supervised clustering of groups with higher (TACI(hi)) versus lower (TACI(lo)) TACI expression levels. Patients in the TACI(lo) group had clinical parameters associated with bad prognosis. A set of 659 genes was differentially expressed between TACI(hi) and TACI(lo) MMCs. This set makes it possible to efficiently classify TACI(hi) and TACI(lo) MMCs in an independent cohort of 40 patients. TACI(hi) MMCs displayed a mature plasma cell gene signature, indicating dependence on the BM environment. In contrast, the TACI(lo) group had a gene signature of plasmablasts, suggesting an attenuated dependence on the BM environment. Taken together, our findings suggest using gene expression profiling to identify the group of patients who might benefit most from treatment with BAFF/APRIL inhibitors.


Subject(s)
Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Multiple Myeloma/pathology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/genetics , B-Cell Activating Factor , Bone Marrow/metabolism , Cluster Analysis , Gene Expression Profiling , Humans , Membrane Proteins/analysis , Middle Aged , Multiple Myeloma/genetics , Osteoclasts/metabolism , Plasma Cells/pathology , Prognosis , Receptors, Tumor Necrosis Factor/analysis , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/analysis
18.
Oncogene ; 24(21): 3512-24, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15735670

ABSTRACT

A hallmark of plasma cells is the expression of syndecan-1, which has major functions in epithelial cells, in particular as the coreceptor of heparin-binding growth factors. We previously found that heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a growth factor for malignant plasma cells. As amphiregulin (AREG) is another heparin-binding factor of the EGF family, we investigated its role in multiple myeloma (MM). Using Affymetrix DNA microarrays, we show here that the AREG gene was expressed by purified primary myeloma cells from 65 patients and that the expression was higher than in normal bone marrow (BM) plasma cells or plasmablastic cells. AREG stimulated IL-6 production and growth of BM stromal cells. Using real-time reverse transcriptase-polymerase chain reaction, we found that MM cells expressed ErbB receptors and that AREG promoted their growth. Furthermore, PD169540 (a pan-ErbB inhibitor) and IRESSA (an ErbB1-specific inhibitor) induced apoptosis of primary myeloma cells from 10/14 and 4/14 patients, respectively, and there was a synergistic effect with dexamethasone. Altogether, our data provide strong evidence that AREG plays an important role in the biology of MM and emphasize the advantages of using ErbB inhibitors, which might target myeloma cells as well as the tumor environment.


Subject(s)
ErbB Receptors/physiology , Gene Expression Profiling , Glycoproteins/biosynthesis , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Multiple Myeloma/genetics , Multiple Myeloma/physiopathology , Amphiregulin , Antineoplastic Agents/pharmacology , Apoptosis , Bone Marrow Cells , Cell Proliferation , EGF Family of Proteins , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Oligonucleotide Array Sequence Analysis , Plasma Cells/physiology , Quinazolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
19.
Blood ; 103(5): 1829-37, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14576062

ABSTRACT

We previously found that some myeloma cell lines express the heparin-binding epidermal growth factor-like growth factor (HB-EGF) gene. As the proteoglycan syndecan-1 is an HB-EGF coreceptor as well as a hallmark of plasma cell differentiation and a marker of myeloma cells, we studied the role of HB-EGF on myeloma cell growth. The HB-EGF gene was expressed by bone marrow mononuclear cells in 8 of 8 patients with myeloma, particularly by monocytes and stromal cells, but not by purified primary myeloma cells. Six of 9 myeloma cell lines and 9 of 9 purified primary myeloma cells expressed ErbB1 or ErbB4 genes coding for HB-EGF receptor. In the presence of a low interleukin-6 (IL-6) concentration, HB-EGF stimulated the proliferation of the 6 ErbB1+ or ErbB4+ cell lines, through the phosphatidylinositol 3-kinase/AKT (PI-3K/AKT) pathway. A pan-ErbB inhibitor blocked the myeloma cell growth factor activity and the signaling induced by HB-EGF. This inhibitor induced apoptosis of patients'myeloma cells cultured with their tumor environment. It also increased patients' myeloma cell apoptosis induced by an anti-IL-6 antibody or dexamethasone. The ErbB inhibitor had no effect on the interaction between multiple myeloma cells and stromal cells. It was not toxic for nonmyeloma cells present in patients' bone marrow cultures or for the growth of hematopoietic progenitors. Altogether, these data identify ErbB receptors as putative therapeutic targets in multiple myeloma.


Subject(s)
Dexamethasone/pharmacology , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/physiology , ErbB Receptors/antagonists & inhibitors , Interleukin-6/immunology , Multiple Myeloma/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis , Blotting, Western , Bone Marrow Cells/cytology , Cell Adhesion , Cell Differentiation , Cell Division , Cell Line, Tumor , Cell Separation , Cells, Cultured , Drug Synergism , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , ErbB Receptors/biosynthesis , Hematopoietic Stem Cells/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharide Receptors/biosynthesis , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, ErbB-4 , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...