Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Blood ; 140(12): 1408-1418, 2022 09 22.
Article in English | MEDLINE | ID: mdl-35667047

ABSTRACT

To determine the survival benefit of allogeneic hematopoietic cell transplantation (allo-HCT) in chronic myelomonocytic leukemias (CMML), we assembled a retrospective cohort of CMML patients 18-70 years old diagnosed between 2000 and 2014 from an international CMML dataset (n = 730) and the EBMT registry (n = 384). The prognostic impact of allo-HCT was analyzed through univariable and multivariable time-dependent models and with a multistate model, accounting for age, sex, CMML prognostic scoring system (low or intermediate-1 grouped as lower-risk, intermediate-2 or high as higher-risk) at diagnosis, and AML transformation. In univariable analysis, lower-risk CMMLs had a 5-year overall survival (OS) of 20% with allo-HCT vs 42% without allo-HCT (P < .001). In higher-risk patients, 5-year OS was 27% with allo-HCT vs 15% without allo-HCT (P = .13). With multistate models, performing allo-HCT before AML transformation reduced OS in patients with lower-risk CMML, and a survival benefit was predicted for men with higher-risk CMML. In a multivariable analysis of lower-risk patients, performing allo-HCT before transformation to AML significantly increased the risk of death within 2 years of transplantation (hazard ratio [HR], 3.19; P < .001), with no significant change in long-term survival beyond this time point (HR, 0.98; P = .92). In higher-risk patients, allo-HCT significantly increased the risk of death in the first 2 years after transplant (HR 1.46; P = .01) but not beyond (HR, 0.60; P = .09). Performing allo-HCT before AML transformation decreases life expectancy in lower-risk patients but may be considered in higher-risk patients.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Chronic , Leukemia, Myelomonocytic, Juvenile , Adolescent , Adult , Aged , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/therapy , Male , Middle Aged , Retrospective Studies , Transplantation, Homologous , Young Adult
2.
Exp Hematol ; 41(4): 387-97.e1, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23178378

ABSTRACT

Multiple myeloma (MM)-induced osteoclast (OC) formation is mainly due to an imbalance of the receptor activator NF-κB ligand (RANKL)-osteoprotegerin (OPG) ratio in favor of RANKL in the bone microenvironment and to the CCL3 production by MM cells. The purpose of the study was to investigate the effect of the immunomodulatory drugs on RANKL/OPG ratio, the production of pro-osteoclastogenic cytokines, and MM-induced OC formation. We found that in vivo concentrations of both lenalidomide (LEN) and pomalidomide (POM) significantly blunted RANKL upregulation normalizing the RANKL/OPG ratio in human osteoprogenitor cells (PreOBs) when co-cultured with MM cells and also inhibited CCL3 production by MM cells. A reduction in CD49d expression, a molecule critically involved in RANKL upregulation in the MM microenvironment, accompanied this effect. Consistently, the pro-osteoclastogenic property of MM cells co-cultured with PreOBs was reduced by both LEN and POM. We further investigated the effect of these drugs on the transcriptional profile of both MM cells and PreOBs by microarray analysis, which showed that adhesion molecules, such as ITGA8 and ICAM2, are significantly downregulated in MM cells. Our data suggest that LEN and POM inhibit MM-induced OC formation through normalization of the RANKL/OPG ratio targeting the expression of adhesion molecules by MM cells.


Subject(s)
Osteoblasts/metabolism , Osteoprotegerin/genetics , RANK Ligand/genetics , Thalidomide/analogs & derivatives , Tumor Microenvironment/drug effects , Antineoplastic Agents/pharmacology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Coculture Techniques , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunologic Factors/pharmacology , Integrin alpha4/genetics , Integrin alpha4/metabolism , Lenalidomide , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteoblasts/cytology , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thalidomide/pharmacology , Tumor Cells, Cultured , Tumor Microenvironment/genetics
3.
Sci Transl Med ; 3(104): 104ra101, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21998408

ABSTRACT

Success with transplantation of autologous hematopoietic stem and progenitor cells (HSPCs) in patients depends on adequate collection of these cells after mobilization from the bone marrow niche by the cytokine granulocyte colony-stimulating factor (G-CSF). However, some patients fail to achieve sufficient HSPC mobilization. Retrospective analysis of bone marrow transplant patient records revealed that diabetes correlated with poor mobilization of CD34+ HSPCs. In mouse models of type 1 and type 2 diabetes (streptozotocin-induced and db/db mice, respectively), we found impaired egress of murine HSPCs from the bone marrow after G-CSF treatment. Furthermore, HSPCs were aberrantly localized in the marrow niche of the diabetic mice, and abnormalities in the number and function of sympathetic nerve termini were associated with this mislocalization. Aberrant responses to ß-adrenergic stimulation of the bone marrow included an inability of marrow mesenchymal stem cells expressing the marker nestin to down-modulate the chemokine CXCL12 in response to G-CSF treatment (mesenchymal stem cells are reported to be critical for HSPC mobilization). The HSPC mobilization defect was rescued by direct pharmacological inhibition of the interaction of CXCL12 with its receptor CXCR4 using the drug AMD3100. These data suggest that there are diabetes-induced changes in bone marrow physiology and microanatomy and point to a potential intervention to overcome poor HSPC mobilization in diabetic patients.


Subject(s)
Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/biosynthesis , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Cell Movement , Cell Separation/methods , Chemokine CXCL12/metabolism , Flow Cytometry/methods , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Mobilization , Humans , Intermediate Filament Proteins/metabolism , Male , Mice , Nerve Tissue Proteins/metabolism , Nestin , Stem Cell Transplantation/methods
4.
Exp Hematol ; 39(1): 55-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20977926

ABSTRACT

OBJECTIVE: Multiple myeloma (MM) cells are extremely resistant to drug-induced apoptosis due to both intrinsic- and bone marrow (BM) microenvironment-dependent drug resistance particularly supported by bone cells. Growing evidence suggest that the osteoclast inhibitor zoledronic acid (ZOL) exerts both indirect and direct anti-tumoral effects, including an in vitro proapoptotic effect on MM cells, although this property has not yet been clearly observed in MM patients. MATERIALS AND METHODS: In this study, we attempt to better define the cytotoxic effect of ZOL on MM cells in order to identify novel drug combinations able to potentiate its proapoptotic effect. RESULTS: Our data shows that ZOL at concentrations ranging from 10 to 100 µM was able to induce MM cell apoptosis overcoming the prosurvival effect of both stromal cells and osteoclasts and independent of the intrinsic bortezomib resistance of MM cells. Interestingly, we found that the capacity of ZOL to induce apoptosis in bortezomib-resistant cells was associated with a downregulation of the proapoptotic molecule myeloid cell leukemia-1. A transcriptional analysis by microarray was also performed to identify genes specifically modulated by ZOL in bortezomib-resistant MM cells. Finally, we show an additive effect of arsenic trioxide on apoptosis when used in combination with ZOL. CONCLUSIONS: Our in vitro data suggest that the use of ZOL at appropriate doses could be explored clinically in bortezomib-resistant MM patients and combined with arsenic trioxide to increase its proapoptotic effect.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Boronic Acids/pharmacology , Diphosphonates/pharmacology , Imidazoles/pharmacology , Multiple Myeloma/pathology , Oxides/pharmacology , Pyrazines/pharmacology , Aged , Arsenic Trioxide , Bortezomib , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Humans , Middle Aged , Multiple Myeloma/genetics , Polymerase Chain Reaction , Zoledronic Acid
5.
Br J Haematol ; 149(3): 376-82, 2010 May.
Article in English | MEDLINE | ID: mdl-20230405

ABSTRACT

This study compared two schedules of low-dose gemtuzumab ozogamicin (GO) as induction monotherapy for untreated acute myeloid leukaemia in older patients unfit for intensive chemotherapy, to identify the more promising regimen for further study. Patients were randomized to receive either best supportive care or a course of GO according to one of two schedules: 3 mg/m(2) on days 1, 3 and 5 (arm A), or GO 6 mg/m(2) on day 1 and 3 mg/m(2) on day 8 (arm B). Primary endpoint was the rate of disease non-progression (DnP), defined as the proportion of patients either achieving a response or maintaining a stable disease following GO induction in each arm. Fifty-six patients were randomized in the two GO arms (A, n = 29; B, n = 27). The rate of DnP was 38% [90% confidence interval (CI), 23-55] in arm A, and 63% (90% CI, 45-78) in arm B. Peripheral cytopenias were the most common adverse events for both regimens. The all-cause early mortality rate was 14% in arm A and 11% in arm B. The day 1 + 8 schedule, which was associated with the highest rate of DnP, met the statistical criteria to be selected as the preferred regimen for phase III comparison with best supportive care.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Aged , Aminoglycosides/adverse effects , Aminoglycosides/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Disease Progression , Drug Administration Schedule , Feasibility Studies , Female , Gemtuzumab , Humans , Male , Middle Aged , Treatment Outcome
6.
Exp Hematol ; 38(2): 141-53, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19963035

ABSTRACT

OBJECTIVE: Multiple myeloma (MM) is characterized by a high incidence of osteolytic bone lesions, which have been previously correlated with the gene expression profiles of MM cells. The aim of this study was to investigate the transcriptional patterns of cells in the bone microenvironment and their relationships with the presence of osteolysis in MM patients. MATERIALS AND METHODS: Both mesenchymal (MSC) and osteoblastic (OB) cells were isolated directly from bone biopsies of MM patients and controls to perform gene expression profiling by microarrays and real-time polymerase chain reaction on selected bone-related genes. RESULTS: We identified a series of upregulated and downregulated genes that were differentially expressed in the MSC cells of osteolytic and nonosteolytic patients. Comparison of the osteolytic and nonosteolytic samples also showed that the MSC cells and OB had distinct transcriptional patterns. No significantly modulated genes were found in the OBs of the osteolytic and nonosteolytic patients. CONCLUSIONS: Our data suggest that the gene expression profiles of cells of the bone microenvironment are different in MM patients and controls, and that MSC cells, but not OBs, have a distinct transcriptional pattern associated with the occurrence of bone lesions in MM patients. These data support the idea that alterations in MSC cells may be involved in MM bone disease.


Subject(s)
Bone and Bones/pathology , Gene Expression Profiling , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/complications , Osteoblasts/metabolism , Osteolysis/etiology , Bone Remodeling/genetics , Cell Division , Gene Expression Regulation , Humans , Mesenchymal Stem Cells/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Oligonucleotide Array Sequence Analysis , Osteoblasts/pathology , Osteolysis/metabolism , Osteolysis/pathology , Polymerase Chain Reaction , RNA, Messenger/analysis
7.
Clin Cancer Res ; 15(21): 6495-502, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19861456

ABSTRACT

The p53 tumor suppressor is part of a small family of related proteins that includes two other members, p73 and p63. Interest in the p53 family members, their functions and their complex interactions and regulation, has steadily grown over recent years and does not show signs of waning. p73 is a major determinant of chemosensitivity in humans, and mutant p53 proteins carrying specific polymorphisms can induce drug resistance by inhibiting TAp73. Cooperation between TA (transactivating, proapoptotic, antiproliferative) and Delta N (truncated, antiapoptotic, pro-proliferative) p73 isoforms and among the three family members guarantees equilibrium between proliferation, differentiation, and cell death, thus creating a harmony that is lost in several human cancers. In this article, we review our current knowledge of the role of p73 in cancer chemosensitivity and the real prospect of therapy targeting this molecule. We also draw attention to the crucial role of specific phosphorylation and acetylation events for p73-induced apoptosis and drug chemosensitivity.


Subject(s)
DNA-Binding Proteins/metabolism , Drug Delivery Systems , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis , DNA-Binding Proteins/antagonists & inhibitors , Drug Design , Humans , Models, Biological , Mutation , Nuclear Proteins/antagonists & inhibitors , Protein Isoforms/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors
8.
Exp Hematol ; 37(8): 879-86, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19446662

ABSTRACT

OBJECTIVE: Osteogenic differentiation of mesenchymal cells toward osteoprogenitor and osteoblastic cells is tightly regulated by several growth and transcription factors at the molecular level. In this article, we focus on the biological mechanisms involved in the osteoblast inhibition induced by myeloma cells. MATERIALS AND METHODS: Current research on the mechanisms regulating myeloma cell and osteoprogenitor cells interactions and on potential therapeutic targets to treat multiple myeloma bone disease is reviewed. RESULTS: Runt-related transcription factor 2 is critically involved in this process along with a large number of nuclear coregulators. Wnt signaling has been recently identified as a critical pathway involved in the regulation of osteoblastogenesis. The impairment of osteogenic differentiation in mesenchymal stem cells occurs in multiple myeloma due to the capacity of malignant plasma cells to suppress the osteogenic differentiation of mesenchymal cells either through the cell contact or the release of soluble factors as interleukin-7, hepatocyte growth factor, interleukin-3, and Wnt inhibitors. CONCLUSION: Runt-related transcription factor 2 and Wnt pathways could be therapeutic targets in the treatment of multiple myeloma bone disease to counterbalance the block of osteogenic differentiation induced by multiple myeloma cells.


Subject(s)
Mesenchymal Stem Cells/pathology , Multiple Myeloma/pathology , Osteogenesis , Animals , Bone Diseases/drug therapy , Bone Diseases/etiology , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Osteoblasts/pathology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism
9.
J Cell Physiol ; 220(2): 401-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19365806

ABSTRACT

Bone marrow stromal cells (MSCs) and osteoblasts are the two main non-haematopoietic cellular components of human bone tissue. To identify novel osteoblast-related molecules, we performed a gene expression profiling analysis comparing MSCs and osteoblasts isolated from the same donors. Genes differentially overexpressed in osteoblasts were mainly related to the negative control of cell proliferation, pro-apoptotic processes, protein metabolism and bone remodelling. Notably, we also identified the collagen XV (COL15A1) gene as the most up-regulated gene in osteoblasts compared with MSCs, previously described as being expressed in the basement membrane in other cell types. The expression of collagen type XV was confirmed at the protein level on isolated osteoblasts and we demonstrated that it significantly increases during the osteogenic differentiation of MSCs in vitro and that free ionised extracellular calcium significantly down-modulates its expression. Moreover, light and electron microscopy showed that collagen type XV is expressed in bone tissue biopsies mainly by working osteoblasts forming new bone tissue or lining bone trabeculae. To our knowledge, these data represent the first evidence of the expression of collagen type XV in human osteoblasts, a calcium-regulated protein which correlates to a specific functional state of these cells.


Subject(s)
Collagen/metabolism , Extracellular Matrix/metabolism , Gene Expression Profiling , Microarray Analysis , Osteoblasts/metabolism , Aged , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Collagen/genetics , Extracellular Matrix/chemistry , Humans , Middle Aged , Osteoblasts/cytology , Osteogenesis/physiology , Phenotype , Stromal Cells/cytology , Stromal Cells/metabolism
10.
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
11.
Cancer Res ; 68(16): 6840-50, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18703490

ABSTRACT

The expression of the chemokine CC-chemokine ligand 20 (CCL20)/macrophage inflammatory protein (MIP)-3alpha and its receptor CC-chemokine receptor 6 (CCR6) by multiple myeloma (MM) and microenvironment cells and their potential relationship with osteoclast (OC) formation and osteolytic bone lesions in MM patients was investigated in this study. First, we found that MM cells rarely produce CCL20/MIP-3alpha but up-regulate its production by bone marrow (BM) osteoprogenitor cells and osteoblasts in coculture with the involvement of soluble factors as interleukin-1beta and tumor necrosis factor alpha. MM cells also stimulate both CCL20/MIP-3alpha and CCR6 expression by OCs in coculture. Thereafter, we showed that CCL20/MIP-3alpha significantly increases both the number of multinucleated tartrate-resistant acid phosphatase-positive OCs and receptor activator of nuclear factor-kappaB-positive OC progenitor cells similar to CCL3/MIP-1alpha. Finally, we found that blocking anti-CCL20/MIP-3alpha and anti-CCR6 antibodies significantly inhibits MM-induced OC formation. In vitro data were further expanded in vivo analyzing a total number of 64 MM patients. Significantly higher CCL20/MIP-3alpha levels were detected in MM patients versus monoclonal gammopathy of uncertain significance (MGUS) subjects and in MM osteolytic patients versus nonosteolytic ones. Moreover, a significant increase of CCL20/MIP-3alpha-positive osteoblasts in osteolytic MM patients compared with nonosteolytic ones was observed. Interestingly, no significant difference in BM CCL20/MIP-3alpha expression and level was observed between MGUS and nonosteolytic MM patients. Our data indicate that CCL20/MIP-3alpha and its receptor CCR6 are up-regulated in the bone microenvironment by MM cells and contribute to OC formation and osteolytic bone lesions in MM patients.


Subject(s)
Bone Diseases/metabolism , Chemokine CCL20/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteoclasts/metabolism , Receptors, CCR6/biosynthesis , Bone Diseases/pathology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Multiple Myeloma/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteogenesis/physiology , Paraproteinemias/genetics , Paraproteinemias/metabolism , Paraproteinemias/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Cells, Cultured
12.
Blood ; 112(6): 2450-62, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18583568

ABSTRACT

We demonstrate that blockade of the MEK/ERK signaling module, using the small-molecule inhibitors PD184352 or PD325901 (PD), strikingly enhances arsenic trioxide (ATO)-induced cytotoxicity in human myeloma cell lines (HMCLs) and in tumor cells from patients with multiple myeloma (MM) through a caspase-dependent mechanism. In HMCLs retaining a functional p53, PD treatment greatly enhances the ATO-induced p53 accumulation and p73, a p53 paralog, cooperates with p53 in caspase activation and apoptosis induction. In HMCLs carrying a nonfunctional p53, cotreatment with PD strikingly elevates the (DR4 + DR5)/(DcR1 + DcR2) tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors ratio and caspase-8 activation of ATO-treated cells. In MM cells, irrespective of p53 status, the combined PD/ATO treatment increases the level of the proapoptotic protein Bim (PD-mediated) and decreases antiapoptotic protein Mcl-1 (ATO-mediated). Moreover, Bim physically interacts with both DR4 and DR5 TRAIL receptors in PD/ATO-treated cells, and loss of Bim interferes with the activation of both extrinsic and intrinsic apoptotic pathways in response to PD/ATO. Finally, PD/ATO treatment induces tumor regression, prolongs survival, and is well tolerated in vivo in a human plasmacytoma xenograft model. These preclinical studies provide the framework for testing PD325901 and ATO combination therapy in clinical trials aimed to improve patient outcome in MM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , MAP Kinase Signaling System/drug effects , Multiple Myeloma/drug therapy , Oxides/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide , Arsenicals/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Humans , Mice , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/pathology , Oxides/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Rev. bras. hematol. hemoter ; 30(supl.2): 3-5, jun. 2008.
Article in English | LILACS | ID: lil-496436

ABSTRACT

Multiple myeloma (MM) is a plasma cell malignancy characterized by the accumulation of malignant plasma cells within the bone marrow (BM). MM cells interact with the microenvironment and induce pathological modifications that in turn support the growth and survival of MM cells. The BM microenvironment consists of various extracellular matrix proteins, and cell components as haematopoietic stem cells, progenitor and precursor cells, immune cells, erythrocytes, BM stromal cells (BMSCs), BM endothelial cells, as well as osteoclasts and osteoblasts that are able to secret several growth factors for MM cells. The direct interactions of MM cells with the microenvironment and the secreted cytokines activate signalling pathways mediating growth, survival, drug resistance and the migration of MM cells as well as osteoclastogenesis and angiogenesis. In this article we underline in particular the new evidences at the basis of the interaction between MM cell and bone cells and the potential role of osteoclast and osteoblast in MM pathophysiology.


O mieloma múltiplo (MM) é uma doença maligna das células plasmáticas caracterizada pelo acúmulo de células plasmáticas na medula óssea (MO). As células do MM interagem com o microambiente e induzem modificações patológicas que, por seu turno, propiciam o crescimento e a sobrevida das células do MM. O microambiente da MO consiste de várias proteínas da matriz extracelular e de componentes hematopoéticos: células-tronco, progenitoras e precursoras, células imunes, eritrocitárias, estromais, endoteliais. Possuem também osteoclastos e osteoblastos capazes de secreção de fatores de crescimento das células do MM. A direta interação das células mielomatosas com o microambiente e a secreção de citocinas ativam cascatas sinalizadoras que mediam o crescimento, sobrevida, resistência a drogas e a migração destas células assim como a osteoclastogênese e a angiogênese. Neste artigo explicitamos novas evidências e as bases da interação das células mielomatosas e as células medulares e o provável papel dos osteoclastos e dos osteoblastos na fisiopatologia do MM.


Subject(s)
Humans , Bone Marrow Cells/pathology , Multiple Myeloma , Multiple Myeloma/physiopathology
14.
Blood ; 112(3): 750-9, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18474725

ABSTRACT

The interleukin-12 (IL-12) receptor (R) B2 gene acts as tumor suppressor in human acute and chronic B-cell leukemias/lymphomas and IL-12rb2-deficient mice develop spontaneously localized plasmacytomas. With this background, we investigated the role of IL-12R beta 2 in multiple myeloma (MM) pathogenesis. Here we show the following: (1) IL-12R beta 2 was expressed in primary MM cells but down-regulated compared with normal polyclonal plasmablastic cells and plasma cells (PCs). IL-6 dampened IL-12R beta 2 expression on polyclonal plasmablastic cells and MM cells. (2) IL-12 reduced the proangiogenic activity of primary MM cells in vitro and decreased significantly (P = .001) the tumorigenicity of the NCI-H929 cell line in SCID/NOD mice by inhibiting cell proliferation and angiogenesis. The latter phenomenon was found to depend on abolished expression of a wide panel of proangiogenic genes and up-regulated expression of the antiangiogenic genes IFN-gamma, IFN-alpha, platelet factor-4, and TIMP-2. Inhibition of the angiogenic potential of primary MM cells was related to down-regulated expression of the proangiogenic genes CCL11, vascular endothelial-cadherin, CD13, and AKT and to up-regulation of an IFN-gamma-related antiangiogenic pathway. Thus, IL-12R beta 2 directly restrains MM cell growth, and targeting of IL-12 to tumor cells holds promise as new therapeutic strategy.


Subject(s)
Gene Expression Regulation, Neoplastic , Multiple Myeloma/chemistry , Receptors, Interleukin-12/analysis , Aged , Aged, 80 and over , Animals , Cell Proliferation/drug effects , Humans , Interleukin-12/pharmacology , Mice , Mice, SCID , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Receptors, Interleukin-12/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Leuk Res ; 32(1): 49-53, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17512053

ABSTRACT

This study examines the response to dexamethasone-doxorubicin-vincristine (DAV) therapy, followed by conditioning regimen and autologous stem cells transplantation (ASCT) in patients with multiple myeloma in relation with the presence of polymorphisms in genes involved in drug metabolism (GSTP1) and DNA synthesis (TYMS). GSTP1 G313G genotype (OR=5.49; 95% CI, 1.3-22.5, p=0.02) and TYMS A227A genotype (OR=3.41; 95% CI, 1.3-8.9, p=0.01) resulted significantly associated with a poor response following chemotherapy and the risk increased for the combined genotype (OR=13.54; 95% CI, 2.0-91.3, p=0.01). TYMS T157T genotype was significantly associated with a poor response after ASCT (OR=4.60; 95% CI, 1.2-16.9, p=0.02). Pre-therapeutic individual determination of the GSTP1 and TYMS polymorphisms could help in choosing the most appropriate protocol.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glutathione S-Transferase pi/genetics , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Polymorphism, Single Nucleotide , Stem Cell Transplantation , Thymidylate Synthase/genetics , Adult , Aged , Aged, 80 and over , Cyclophosphamide/therapeutic use , Dacarbazine/therapeutic use , Female , Humans , Male , Melphalan/therapeutic use , Middle Aged , Nimustine/therapeutic use , Survival Analysis , Transplantation Conditioning , Transplantation, Autologous , Treatment Outcome , Vincristine/therapeutic use
16.
Leuk Lymphoma ; 48(12): 2323-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18067006

ABSTRACT

Bone destruction is the hallmark of multiple myeloma (MM) due to the high capacity of malignant plasma cells to induce a severe imbalance of bone remodeling. Growing evidences suggest that MM cell interactions with bone marrow (BM) osteoblast have a critical role in the pathophysiology of osteolytic lesions. Indeed histomorphometric studies have demonstrated that MM patients with osteolytic bone lesions have lower numbers of osteoblasts and decreased bone formation together with osteoclast activation. Recently, the biological mechanisms involved in the osteoblast inhibition induced by MM cells have begun to be elucidated, underlying the main role of the block of osteoblast differentiation in the development of bone lesions. In this article, we summarize the main mechanisms regulating MM cell and osteoblast interactions.


Subject(s)
Bone Marrow Cells/physiology , Cell Communication , Multiple Myeloma/pathology , Osteoblasts/physiology , Osteolysis/etiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-3/physiology , Interleukin-7/physiology , Proteasome Endopeptidase Complex/physiology , Ubiquitin/metabolism , Wnt Proteins/physiology
17.
Blood ; 110(13): 4464-75, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17848618

ABSTRACT

Angiogenesis has a critical role in the pathophysiology of multiple myeloma (MM); however, the molecular mechanisms underlying this process are not completely elucidated. The new tumor-suppressor gene inhibitor of growth family member 4 (ING4) has been recently implicated in solid tumors as a repressor of angiogenesis. In this study, we found that ING4 expression in MM cells was correlated with the expression of the proangiogenic molecules interleukin-8 (IL-8) and osteopontin (OPN). Moreover, we demonstrate that ING4 suppression in MM cells up-regulated IL-8 and OPN, increasing the hypoxia inducible factor-1alpha (HIF-1alpha) activity and its target gene NIP-3 expression in hypoxic condition. In turn, we show that the inhibition of HIF-1alpha by siRNA suppressed IL-8 and OPN production by MM cells under hypoxia. A direct interaction between ING4 and the HIF prolyl hydroxylase 2 (HPH-2) was also demonstrated. Finally, we show that ING4 suppression in MM cells significantly increased vessel formation in vitro, blunted by blocking IL-8 or OPN. These in vitro observations were confirmed in vivo by finding that MM patients with high IL-8 production and microvascular density (MVD) have significantly lower ING4 levels compared with those with low IL-8 and MVD. Our data indicate that ING4 exerts an inhibitory effect on the production of proangiogenic molecules and consequently on MM-induced angiogenesis.


Subject(s)
Angiogenic Proteins/biosynthesis , Cell Cycle Proteins/physiology , Homeodomain Proteins/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Multiple Myeloma/pathology , Neovascularization, Pathologic/etiology , Tumor Suppressor Proteins/physiology , Aged , Angiogenic Proteins/genetics , Bone Marrow Examination , Cell Line, Tumor , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Middle Aged , Multiple Myeloma/blood supply , Multiple Myeloma/metabolism , Osteopontin/biosynthesis , Osteopontin/genetics
18.
Cancer Res ; 67(16): 7665-74, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17702698

ABSTRACT

Osteoblast impairment occurs within multiple myeloma cell infiltration into the bone marrow. Canonical Wnt signaling activation in osteoprogenitor cells is involved in osteoblast formation through the stabilization of dephosphorylated beta-catenin and its nuclear translocation. The effects of multiple myeloma cells on Wnt signaling in human mesenchymal/osteoprogenitor cells are unclear. In 60 multiple myeloma patients checked, we found that among the Wnt inhibitors, Dickkopf-1 and secreted frizzled-related protein-3 were produced by multiple myeloma cells. However, although multiple myeloma cells or multiple myeloma bone marrow plasma affected expression of genes in the canonical Wnt signaling and inhibited beta-catenin stabilization in murine osteoprogenitor cells, they failed to block the canonical Wnt pathway in human mesenchymal or osteoprogenitor cells. Consistently, Wnt3a stimulation in human osteoprogenitor cells did not blunt the inhibitory effect of multiple myeloma cells on osteoblast formation. Consequently, despite the higher Wnt antagonist bone marrow levels in osteolytic multiple myeloma patients compared with nonosteolytic ones, beta-catenin immunostaining was not significantly different. Our results support the link between the production of Wnt antagonists by multiple myeloma cells and the presence of bone lesions in multiple myeloma patients but show that myeloma cells do not inhibit canonical Wnt signaling in human bone microenvironment.


Subject(s)
Bone Marrow Cells/metabolism , Glycoproteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Multiple Myeloma/metabolism , Wnt Proteins/antagonists & inhibitors , Animals , Bone Marrow Cells/pathology , Cell Line, Tumor , Coculture Techniques , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Osteoblasts/metabolism , Osteoblasts/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Wnt Proteins/metabolism
19.
Br J Haematol ; 137(5): 454-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17488488

ABSTRACT

The multidrug resistance gene (MDR1) has been reported to be an additional prognostic factor in acute myeloid leukaemia patients. This study evaluated the prognostic role of MDR1 in the outcome of 115 multiple myeloma patients treated with DAV (dexamethasone, doxorubicin [adryamicin] and vincristine) regimen followed by autologous transplantation. In particular, when investigating the C3435T polymorphism, a prognostic value of MDR1 genotypes for overall survival (OS) was observed. Our data suggested a longer OS for patients with C/T and T/T genotypes (log-rank test, P = 0.02) compared with patients with C/C genotype.


Subject(s)
Genes, MDR , Multiple Myeloma/genetics , Polymorphism, Single Nucleotide , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adult , Aged , Aged, 80 and over , Alleles , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Odds Ratio , Prognosis , Survival Analysis , Vincristine/administration & dosage
20.
Tumori ; 93(1): 97-9, 2007.
Article in English | MEDLINE | ID: mdl-17455879

ABSTRACT

Most of the information about the genetic composition of parathyroid tumors has been obtained by comparative genomic hybridization (CGH) and loss of heterozygosity (LOH) studies, whereas only few conventional cytogenetic investigation results are available. We have performed cytogenetic analysis of short-term cultures from 3 parathyroid adenoma tissue samples. Two cases showed a normal karyotype in all the metaphases obtained from independent primary cultures. In one case 5 metaphases (in a total of 25) from 2 independent cultures showed a nonrandom translocation t(4;13)(q21;q14), which was therefore accepted as clonal. To our knowledge this is the second clonal translocation described in this tumor type. Further conventional cytogenetic analysis of more parathyroid tumor specimens would be necessary to identify other specific abnormalities and the involved genes with a potential important role in the diagnosis, prognosis and pathogenesis of parathyroid tumors.


Subject(s)
Adenoma/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 4/genetics , Parathyroid Neoplasms/genetics , Translocation, Genetic , Cytogenetic Analysis , Humans , Karyotyping
SELECTION OF CITATIONS
SEARCH DETAIL
...