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










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731936

ABSTRACT

Multiple myeloma is a malignancy characterized by the accumulation of malignant plasma cells in bone marrow and the production of monoclonal immunoglobulin. A hallmark of cancer is the evasion of immune surveillance. Histone deacetylase inhibitors have been shown to promote the expression of silenced molecules and hold potential to increase the anti-MM efficacy of immunotherapy. The aim of the present work was to assess the potential effect of tinostamustine (EDO-S101), a first-in-class alkylating deacetylase inhibitor, in combination with daratumumab, an anti-CD38 monoclonal antibody (mAb), through different preclinical studies. Tinostamustine increases CD38 expression in myeloma cell lines, an effect that occurs in parallel with an increment in CD38 histone H3 acetylation levels. Also, the expression of MICA and MICB, ligands for the NK cell activating receptor NKG2D, augments after tinostamustine treatment in myeloma cell lines and primary myeloma cells. Pretreatment of myeloma cell lines with tinostamustine increased the sensitivity of these cells to daratumumab through its different cytotoxic mechanisms, and the combination of these two drugs showed a higher anti-myeloma effect than individual treatments in ex vivo cultures of myeloma patients' samples. In vivo data confirmed that tinostamustine pretreatment followed by daratumumab administration significantly delayed tumor growth and improved the survival of mice compared to individual treatments. In summary, our results suggest that tinostamustine could be a potential candidate to improve the efficacy of anti-CD38 mAbs.


Subject(s)
ADP-ribosyl Cyclase 1 , Antibodies, Monoclonal , Multiple Myeloma , NK Cell Lectin-Like Receptor Subfamily K , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Humans , ADP-ribosyl Cyclase 1/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Mice , Cell Line, Tumor , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Xenograft Model Antitumor Assays , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Membrane Glycoproteins/metabolism , Drug Synergism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Up-Regulation/drug effects
2.
Cells ; 10(3)2021 03 04.
Article in English | MEDLINE | ID: mdl-33806619

ABSTRACT

BH3-mimetics targeting anti-apoptotic proteins such as MCL-1 (S63845) or BCL-2 (venetoclax) are currently being evaluated as effective therapies for the treatment of multiple myeloma (MM). Interleukin 6, produced by mesenchymal stromal cells (MSCs), has been shown to modify the expression of anti-apoptotic proteins and their interaction with the pro-apoptotic BIM protein in MM cells. In this study, we assess the efficacy of S63845 and venetoclax in MM cells in direct co-culture with MSCs derived from MM patients (pMSCs) to identify additional mechanisms involved in the stroma-induced resistance to these agents. MicroRNAs miR-193b-3p and miR-21-5p emerged among the top deregulated miRNAs in myeloma cells when directly co-cultured with pMSCs, and we show their contribution to changes in MCL-1 and BCL-2 protein expression and in the activity of S63845 and venetoclax. Additionally, direct contact with pMSCs under S63845 and/or venetoclax treatment modifies myeloma cell dependence on different BCL-2 family anti-apoptotic proteins in relation to BIM, making myeloma cells more dependent on the non-targeted anti-apoptotic protein or BCL-XL. Finally, we show a potent effect of the combination of S63845 and venetoclax even in the presence of pMSCs, which supports this combinatorial approach for the treatment of MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Thiophenes/therapeutic use , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Multiple Myeloma/pathology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology
3.
Cancers (Basel) ; 13(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802806

ABSTRACT

Immunosuppression is a common feature of multiple myeloma (MM) patients and has been associated with disease evolution from its precursor stages. MM cells promote immunosuppressive effects due to both the secretion of soluble factors, which inhibit the function of immune effector cells, and the recruitment of immunosuppressive populations. Alterations in the expression of surface molecules are also responsible for immunosuppression. In this scenario, immunotherapy, as is the case of immunotherapeutic monoclonal antibodies (mAbs), aims to boost the immune system against tumor cells. In fact, mAbs exert part of their cytotoxic effects through different cellular and soluble immune components and, therefore, patients' immunosuppressive status could reduce their efficacy. Here, we will expose the alterations observed in symptomatic MM, as compared to its precursor stages and healthy subjects, in the main immune populations, especially the inhibition of effector cells and the activation of immunosuppressive populations. Additionally, we will revise the mechanisms responsible for all these alterations, including the interplay between MM cells and immune cells and the interactions among immune cells themselves. We will also summarize the main mechanisms of action of the four mAbs approved so far for the treatment of MM. Finally, we will discuss the potential immune-stimulating effects of non-immunotherapeutic drugs, which could enhance the efficacy of immunotherapeutic treatments.

4.
Cancers (Basel) ; 12(12)2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33287189

ABSTRACT

PD1 expression in CD4+ and CD8+ T cells is increased after treatment in multiple myeloma patients with persistent disease. The GEM-Pembresid trial analyzed the efficacy and safety of pembrolizumab as consolidation in patients achieving at least very good partial response but with persistent measurable disease after first- or second-line treatment. Moreover, the characteristics of the immune system were investigated to identify potential biomarkers of response to pembrolizumab. One out of the 17 evaluable patients showed a decrease in the amount of M-protein, although a potential late effect of high-dose melphalan could not be ruled out. Fourteen adverse events were considered related to pembrolizumab, two of which (G3 diarrhea and G2 pneumonitis) prompted treatment discontinuation and all resolving without sequelae. Interestingly, pembrolizumab induced a decrease in the percentage of NK cells at cycle 3, due to the reduction of the circulating and adaptive subsets (0.615 vs. 0.43, p = 0.007; 1.12 vs. 0.86, p = 0.02). In the early progressors, a significantly lower expression of PD1 in CD8+ effector memory T cells (MFI 1327 vs. 926, p = 0.03) was observed. In conclusion, pembrolizumab used as consolidation monotherapy shows an acceptable toxicity profile but did not improve responses in this MM patient population. The trial was registered at clinicaltrials.gov with identifier NCT02636010 and with EUDRACT number 2015-003359-23.

5.
Cancers (Basel) ; 12(10)2020 Sep 24.
Article in English | MEDLINE | ID: mdl-32987735

ABSTRACT

BACKGROUND: Proviral Insertion site for Moloney murine leukemia virus (PIM) kinases are overexpressed in hematologic malignancies, including multiple myeloma. Previous preclinical data from our group demonstrated the anti-myeloma effect of the pan-PIM kinase inhibitor PIM447. METHODS: Based on those data, we evaluate here, by in vitro and in vivo studies, the activity of the triple combination of PIM447 + pomalidomide + dexamethasone (PIM-Pd) in multiple myeloma. RESULTS: Our results show that the PIM-Pd combination exerts a potent anti-myeloma effect in vitro and in vivo, where it markedly delays tumor growth and prolongs survival of treated mice. Mechanism of action studies performed in vitro and on mice tumor samples suggest that the combination PIM-Pd inhibits protein translation processes through the convergent inhibition of c-Myc and mTORC1, which subsequently disrupts the function of eIF4E. Interestingly the MM pro-survival factor IRF4 is also downregulated after PIM-Pd treatment. As a whole, all these molecular changes would promote cell cycle arrest and deregulation of metabolic pathways, including glycolysis and lipid biosynthesis, leading to inhibition of myeloma cell proliferation. CONCLUSIONS: Altogether, our data support the clinical evaluation of the triple combination PIM-Pd for the treatment of patients with multiple myeloma.

7.
Cells ; 8(11)2019 11 13.
Article in English | MEDLINE | ID: mdl-31766279

ABSTRACT

A high priority problem in multiple myeloma (MM) management is the development of resistance to administered therapies, with most myeloma patients facing successively shorter periods of response and relapse. Herewith, we review the current knowledge on the mechanisms of resistance to the standard backbones in MM treatment: proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies (mAbs). In some cases, strategies to overcome resistance have been discerned, and an effort should be made to evaluate whether resensitization to these agents is feasible in the clinical setting. Additionally, at a time in which we are moving towards precision medicine in MM, it is equally important to identify reliable and accurate biomarkers of sensitivity/refractoriness to these main therapeutic agents with the goal of having more efficacious treatments and, if possible, prevent the development of relapse.


Subject(s)
Drug Resistance, Neoplasm , Genetic Background , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Standard of Care , Drug Resistance, Neoplasm/genetics , Humans , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/standards , Multiple Myeloma/metabolism , Precision Medicine/methods , Precision Medicine/standards , Standard of Care/standards , Treatment Outcome
8.
Haematologica ; 102(12): 2113-2124, 2017 12.
Article in English | MEDLINE | ID: mdl-28860344

ABSTRACT

Kinesin spindle protein inhibition is known to be an effective therapeutic approach in several malignancies. Filanesib (ARRY-520), an inhibitor of this protein, has demonstrated activity in heavily pre-treated multiple myeloma patients. The aim of the work herein was to investigate the activity of filanesib in combination with pomalidomide plus dexamethasone backbone, and the mechanisms underlying the potential synergistic effect. The ability of filanesib to enhance the activity of pomalidomide plus dexamethasone was studied in several in vitro and in vivo models. Mechanisms of this synergistic combination were dissected by gene expression profiling, immunostaining, cell cycle and short interfering ribonucleic acid studies. Filanesib showed in vitro, ex vivo, and in vivo synergy with pomalidomide plus dexamethasone treatment. Importantly, the in vivo synergy observed in this combination was more evident in large, highly proliferative tumors, and was shown to be mediated by the impairment of mitosis transcriptional control, an increase in monopolar spindles, cell cycle arrest and the induction of apoptosis in cells in proliferative phases. In addition, the triple combination increased the activation of the proapoptotic protein BAX, which has previously been associated with sensitivity to filanesib, and could potentially be used as a predictive biomarker of response to this combination. Our results provide preclinical evidence for the potential benefit of the combination of filanesib with pomalidomide and dexamethasone, and supported the initiation of a recently activated trial being conducted by the Spanish Myeloma group which is investigating this combination in relapsed myeloma patients.


Subject(s)
Dexamethasone/therapeutic use , Kinesins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Thalidomide/analogs & derivatives , Thiadiazoles/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Drug Synergism , Humans , Mice , Thalidomide/therapeutic use , Treatment Outcome
9.
Clin Cancer Res ; 23(21): 6602-6615, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28790111

ABSTRACT

Purpose: The search for new drugs that control the continuous relapses of multiple myeloma is still required. Here, we report for the first time the potent antimyeloma activity of amiloride, an old potassium-sparing diuretic approved for the treatment of hypertension and edema due to heart failure.Experimental Design: Myeloma cell lines and primary samples were used to evaluate cytotoxicity of amiloride. In vivo studies were carried out in a xenograft mouse model. The mechanisms of action were investigated using RNA-Seq experiments, qRT-PCR, immunoblotting, and immunofluorescence assays.Results: Amiloride-induced apoptosis was observed in a broad panel of multiple myeloma cell lines and in a xenograft mouse model. Moreover, amiloride also had a synergistic effect when combined with dexamethasone, melphalan, lenalidomide, and pomalidomide. RNA-Seq experiments showed that amiloride not only significantly altered the level of transcript isoforms and alternative splicing events, but also deregulated the spliceosomal machinery. In addition, disruption of the splicing machinery in immunofluorescence studies was associated with the inhibition of myeloma cell viability after amiloride exposure. Although amiloride was able to induce apoptosis in myeloma cells lacking p53 expression, activation of p53 signaling was observed in wild-type and mutated TP53 cells after amiloride exposure. On the other hand, we did not find a significant systemic toxicity in mice treated with amiloride.Conclusions: Overall, our results demonstrate the antimyeloma activity of amiloride and provide a mechanistic rationale for its use as an alternative treatment option for relapsed multiple myeloma patients, especially those with 17p deletion or TP53 mutations that are resistant to current therapies. Clin Cancer Res; 23(21); 6602-15. ©2017 AACR.


Subject(s)
Amiloride/administration & dosage , Diuretics/administration & dosage , Drug Synergism , Multiple Myeloma/drug therapy , Tumor Suppressor Protein p53/genetics , Amiloride/adverse effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Diuretics/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
J Hematol Oncol ; 10(1): 127, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28633670

ABSTRACT

BACKGROUND: Despite recent advances in the treatment of multiple myeloma (MM), the prognosis of most patients remains poor, and resistance to traditional and new drugs frequently occurs. EDO-S101 is a novel therapeutic agent conceived as the fusion of a histone deacetylase inhibitor radical to bendamustine, with the aim of potentiating its alkylating activity. METHODS: The efficacy of EDO-S101 was evaluated in vitro, ex vivo and in vivo, alone, and in combination with standard anti-myeloma agents. The underlying mechanisms of action were also evaluated on MM cell lines, patient samples, and different murine models. RESULTS: EDO-S101 displayed potent activity in vitro in MM cell lines (IC50 1.6-4.8 µM) and ex vivo in cells isolated from MM patients, which was higher than that of bendamustine and independent of the p53 status and previous melphalan resistance. This activity was confirmed in vivo, in a CB17-SCID murine plasmacytoma model and in de novo Vk*MYC mice, leading to a significant survival improvement in both models. In addition, EDO-S101 was the only drug with single-agent activity in the multidrug resistant Vk12653 murine model. Attending to its mechanism of action, the molecule showed both, a HDACi effect (demonstrated by α-tubulin and histone hyperacetylation) and a DNA-damaging effect (shown by an increase in γH2AX); the latter being again clearly more potent than that of bendamustine. Using a reporter plasmid integrated into the genome of some MM cell lines, we demonstrate that, apart from inducing a potent DNA damage, EDO-S101 specifically inhibited the double strand break repair by the homologous recombination pathway. Moreover, EDO-S101 treatment reduced the recruitment of repair proteins such as RAD51 to DNA-damage sites identified as γH2AX foci. Finally, EDO-S101 preclinically synergized with bortezomib, both in vitro and in vivo. CONCLUSION: These findings provide rationale for the clinical investigation of EDO-S101 in MM, either as a single agent or in combination with other anti-MM drugs, particularly proteasome inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , DNA Damage/drug effects , DNA Repair/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bendamustine Hydrochloride/analogs & derivatives , Bendamustine Hydrochloride/pharmacology , Bendamustine Hydrochloride/therapeutic use , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, SCID , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology
11.
Haematologica ; 102(1): 168-175, 2017 01.
Article in English | MEDLINE | ID: mdl-27540138

ABSTRACT

Despite new advances in multiple myeloma treatment and the consequent improvement in overall survival, most patients relapse or become refractory to treatment. This suggests that new molecules and combinations that may further inhibit important survival pathways for these tumor cells are needed. In this context, zalypsis is a novel compound, derived from marine organisms, with a powerful preclinical anti-myeloma effect based on the sensitivity of malignant plasma cells to DNA-damage induction; and it has already been tested in a phase I/II clinical trial in multiple myeloma. We hypothesized that the addition of this compound to the combination of bortezomib plus dexamethasone may improve efficacy with acceptable toxicity. The triple combination demonstrated strong synergy and higher efficacy compared with double combinations; not only in vitro, but also ex vivo and, especially, in in vivo experiments. The triple combination triggers cell death, mainly through a synergistic induction of DNA damage and a decrease in the nuclear localization of nuclear factor kappa B. Our findings support the clinical evaluation of this combination for relapsed and refractory myeloma patients.


Subject(s)
Bortezomib/pharmacology , DNA Damage/drug effects , Dexamethasone/pharmacology , Multiple Myeloma/genetics , Tetrahydroisoquinolines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Drug Synergism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , NF-kappa B/metabolism , Protein Transport/drug effects , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 23(1): 225-238, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27440267

ABSTRACT

PURPOSE: PIM kinases are a family of serine/threonine kinases recently proposed as therapeutic targets in oncology. In the present work, we have investigated the effects of the novel pan-PIM kinase inhibitor, PIM447, on myeloma cells and myeloma-associated bone disease using different preclinical models. EXPERIMENTAL DESIGN: In vitro/ex vivo cytotoxicity of PIM447 was evaluated on myeloma cell lines and patient samples. Synergistic combinations with standard treatments were analyzed with Calcusyn Software. PIM447 effects on bone cells were assessed on osteogenic and osteoclastogenic cultures. The mechanisms of PIM447 were explored by immunoblotting, qPCR, and immunofluorescence. A murine model of disseminated multiple myeloma was employed for in vivo studies. RESULTS: PIM447 is cytotoxic for myeloma cells due to cell-cycle disruption and induction of apoptosis mediated by a decrease in phospho-Bad (Ser112) and c-Myc levels and the inhibition of mTORC1 pathway. Importantly, PIM447 demonstrates a very strong synergy with different standard treatments such as bortezomib + dexamethasone (combination index, CI = 0.002), lenalidomide + dexamethasone (CI = 0.065), and pomalidomide + dexamethasone (CI = 0.077). PIM447 also inhibits in vitro osteoclast formation and resorption, downregulates key molecules involved in these processes, and partially disrupts the F-actin ring, while increasing osteoblast activity and mineralization. Finally, PIM447 significantly reduced the tumor burden and prevented tumor-associated bone loss in a disseminated murine model of human myeloma. CONCLUSIONS: Our results demonstrate dual antitumoral and bone-protective effects of PIM447. This fact, together with the very strong synergy exhibited with standard-of-care treatments, supports the future clinical development of this drug in multiple myeloma. Clin Cancer Res; 23(1); 225-38. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Bone and Bones/drug effects , Bone and Bones/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Protective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bone Resorption/drug therapy , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Gene Expression , Humans , Mice , Multiple Myeloma/drug therapy , Osteoclasts/drug effects , Osteoclasts/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Standard of Care , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
PLoS One ; 9(3): e92378, 2014.
Article in English | MEDLINE | ID: mdl-24658332

ABSTRACT

Despite recent advances in the treatment of multiple myeloma (MM), it remains an incurable disease potentially due to the presence of resistant myeloma cancer stem cells (MM-CSC). Although the presence of clonogenic cells in MM was described three decades ago, the phenotype of MM-CSC is still controversial, especially with respect to the expression of syndecan-1 (CD138). Here, we demonstrate the presence of two subpopulations--CD138++ (95-99%) and CD138low (1-5%)--in eight MM cell lines. To find out possible stem-cell-like features, we have phenotypically, genomic and functionally characterized the two subpopulations. Our results show that the minor CD138low subpopulation is morphologically identical to the CD138++ fraction and does not represent a more immature B-cell compartment (with lack of CD19, CD20 and CD27 expression). Moreover, both subpopulations have similar gene expression and genomic profiles. Importantly, both CD138++ and CD138low subpopulations have similar sensitivity to bortezomib, melphalan and doxorubicin. Finally, serial engraftment in CB17-SCID mice shows that CD138++ as well as CD138low cells have self-renewal potential and they are phenotypically interconvertible. Overall, our results differ from previously published data in MM cell lines which attribute a B-cell phenotype to MM-CSC. Future characterization of clonal plasma cell subpopulations in MM patients' samples will guarantee the discovery of more reliable markers able to discriminate true clonogenic myeloma cells.


Subject(s)
Multiple Myeloma/physiopathology , Syndecan-1/genetics , Syndecan-1/physiology , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , DNA Copy Number Variations , Doxorubicin/pharmacology , Heterografts , Humans , Immunophenotyping , Melphalan/pharmacology , Mice, SCID , Multiple Myeloma/immunology , Neoplastic Stem Cells , Phenotype , Plasma Cells/immunology , Precursor Cells, B-Lymphoid , Pyrazines/pharmacology , Syndecan-1/biosynthesis
14.
Blood ; 122(22): 3591-8, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24072855

ABSTRACT

Circulating myeloma tumor cells (CTCs) as defined by the presence of peripheral blood (PB) clonal plasma cells (PCs) are a powerful prognostic marker in multiple myeloma (MM). However, the biological features of CTCs and their pathophysiological role in MM remains unexplored. Here, we investigate the phenotypic, cytogenetic, and functional characteristics as well as the circadian distribution of CTCs vs paired bone marrow (BM) clonal PCs from MM patients. Our results show that CTCs typically represent a unique subpopulation of all BM clonal PCs, characterized by downregulation (P < .05) of integrins (CD11a/CD11c/CD29/CD49d/CD49e), adhesion (CD33/CD56/CD117/CD138), and activation molecules (CD28/CD38/CD81). Fluorescence in situ hybridization analysis of fluorescence-activated cell sorter-sorted CTCs also unraveled different cytogenetic profiles vs paired BM clonal PCs. Moreover, CTCs were mostly quiescent and associated with higher clonogenic potential when cocultured with BM stromal cells. Most interestingly, CTCs showed a circadian distribution which fluctuates in a similar pattern to that of CD34(+) cells, and opposite to stromal cell-derived factor 1 plasma levels and corresponding surface expression of CXC chemokine receptor 4 on clonal PCs, suggesting that in MM, CTCs may egress to PB to colonize/metastasize other sites in the BM during the patients' resting period.


Subject(s)
Multiple Myeloma/blood , Neoplastic Cells, Circulating/pathology , Antigens, CD/blood , Cell Cycle , Circadian Rhythm , Cytogenetic Analysis , Humans , Immunophenotyping , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Neoplastic Cells, Circulating/classification , Neoplastic Cells, Circulating/immunology , Plasma Cells/classification , Plasma Cells/immunology , Plasma Cells/pathology , Prognosis , Prospective Studies , Tumor Stem Cell Assay
16.
Haematologica ; 98(4): 640-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23100276

ABSTRACT

MicroRNA have been demonstrated to be deregulated in multiple myeloma. We have previously reported that miR-214 is down-regulated in multiple myeloma compared to in normal plasma cells. The functional role of miR-214 in myeloma pathogenesis was explored by transfecting myeloma cell lines with synthetic microRNA followed by gene expression profiling. Putative miR-214 targets were validated by luciferase reporter assay. Ectopic expression of miR-214 reduced cell growth and induced apoptosis of myeloma cells. In order to identify the potential direct target genes of miR-214 which could be involved in the biological pathways regulated by this microRNA, gene expression profiling of the H929 myeloma cell line transfected with precursor miR-214 was carried out. Functional analysis revealed significant enrichment for DNA replication, cell cycle phase and DNA binding. miR-214 directly down-regulated the expression of PSMD10, which encodes the oncoprotein gankyrin, and ASF1B, a histone chaperone required for DNA replication, by binding to their 3'-untranslated regions. In addition, gankyrin inhibition induced an increase of P53 mRNA levels and subsequent up-regulation of CDKN1A (p21Waf1/Cip1) and BAX transcripts, which are direct transcriptional targets of p53. In conclusion, MiR-214 functions as a tumor suppressor in myeloma by positive regulation of p53 and inhibition of DNA replication.


Subject(s)
Cell Proliferation , DNA Replication , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Tumor Suppressor Protein p53/genetics , 5' Untranslated Regions/genetics , Apoptosis/genetics , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Methylation , Gene Expression Profiling , Humans , Immunoblotting , MicroRNAs/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Oligonucleotide Array Sequence Analysis , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
17.
Haematologica ; 97(7): 1110-4, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22315496

ABSTRACT

Although new therapies have doubled the survival of multiple myeloma patients, this remains an incurable disease. It has been postulated that the so-called myeloma cancer stem cells would be responsible for tumor initiation and relapse but their unequivocal identification remains unclear. Here, we investigated in a panel of myeloma cell lines the presence of CD20(+) cells harboring a stem-cell phenotype. Thus, only a small population of CD20(dim+) cells (0.3%) in the RPMI-8226 cell line was found. CD20(dim+) RPMI-8226 cells expressed the plasma cell markers CD38 and CD138 and were CD19(-)CD27(-). Additionally, CD20(dim+) RPMI-8226 cells did not exhibit stem-cell markers as shown by gene expression profiling and the aldehyde dehydrogenase assay. Furthermore, we demonstrated that CD20(dim+) RPMI-8226 cells are not essential for CB17-SCID mice engraftment and show lower self-renewal potential than the CD20(-) RPMI-8226 cells. These results do not support CD20 expression for the identification of myeloma cancer stem cells.


Subject(s)
Antigens, CD20/genetics , Biomarkers, Tumor/genetics , Multiple Myeloma/metabolism , Neoplastic Stem Cells/metabolism , Aldehyde Dehydrogenase/analysis , Animals , Antigens, CD/genetics , Cell Line, Tumor , Flow Cytometry , Gene Expression , Gene Expression Profiling , Humans , Immunophenotyping , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , Transplantation, Heterologous
18.
Haematologica ; 96(5): 687-95, 2011 May.
Article in English | MEDLINE | ID: mdl-21330323

ABSTRACT

BACKGROUND: Although the majority of patients with acute myeloid leukemia initially respond to conventional chemotherapy, relapse is still the leading cause of death, probably because of the presence of leukemic stem cells that are insensitive to current therapies. We investigated the antileukemic activity and mechanism of action of zalypsis, a novel alkaloid of marine origin. DESIGN AND METHODS: The activity of zalypsis was studied in four acute myeloid leukemia cell lines and in freshly isolated blasts taken from patients with acute myeloid leukemia before they started therapy. Zalypsis-induced apoptosis of both malignant and normal cells was measured using flow cytometry techniques. Gene expression profiling and western blot studies were performed to assess the mechanism of action of the alkaloid. RESULTS: Zalypsis showed a very potent antileukemic activity in all the cell lines tested and potentiated the effect of conventional antileukemic drugs such as cytarabine, fludarabine and daunorubicin. Interestingly, zalypsis showed remarkable ex vivo potency, including activity against the most immature blast cells (CD34(+) CD38(-) Lin(-)) which include leukemic stem cells. Zalypsis-induced apoptosis was the result of an important deregulation of genes involved in the recognition of double-strand DNA breaks, such as Fanconi anemia genes and BRCA1, but also genes implicated in the repair of double-strand DNA breaks, such as RAD51 and RAD54. These gene findings were confirmed by an increase in several proteins involved in the pathway (pCHK1, pCHK2 and pH2AX). CONCLUSIONS: The potent and selective antileukemic effect of zalypsis on DNA damage response mechanisms observed in acute myeloid leukemia cell lines and in patients' samples provides the rationale for the investigation of this compound in clinical trials.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Damage , Stem Cells/drug effects , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Cells, Cultured
19.
Neuropharmacology ; 59(6): 480-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20603136

ABSTRACT

It is well known that the efficiency of Herpes simplex virus thymidine kinase gene/ganciclovir (HSV-tk/GCV) therapy is improved by the bystander effect, which mainly relies on gap junctional intercellular communication (GJIC). Malignant gliomas communicate poorly through gap junctions, consequently, agents with the ability to increase GJIC are good candidates to improve the efficiency of this therapy. Since we previously showed that the inhibition of ATP-sensitive potassium (KATP) channels promoted by tolbutamide increased GJIC in rat C6 glioma cells, we have investigated whether tolbutamide could increase the bystander effect in HSV-tk/GCV therapy against human glioma cells. We found that tolbutamide increased GJIC in U373 human glioma cells, an effect that was due to the up-regulation of connexin43, a protein that forms gap junctions channels. More interestingly, our results show that tolbutamide increased the efficiency of HSV-tk/GCV in co-cultures containing U373 cells and U373 cells transfected with HSV-tk. This effect was impaired in the presence of carbenoxolone, an inhibitor of GJIC. Furthermore, tolbutamide did not enhance the bystander effect in connexin43-silenced co-cultures. Together our results reveal that the inhibition of KATP channels promoted by tolbutamide enhances the bystander effect in HSV-tk/GCV therapy by increasing connexin43-mediated gap junctional intercellular communication in U373 human glioma cells.


Subject(s)
Bystander Effect/genetics , Gap Junctions/metabolism , KATP Channels/metabolism , Simplexvirus/metabolism , Thymidine Kinase/metabolism , Viral Proteins/metabolism , Analysis of Variance , Blotting, Western , Bystander Effect/drug effects , Cell Line, Tumor , Cells, Cultured , Flow Cytometry , Ganciclovir/pharmacology , Gap Junctions/drug effects , Gap Junctions/genetics , Humans , In Situ Nick-End Labeling , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Simplexvirus/genetics , Thymidine Kinase/genetics , Transfection , Viral Proteins/genetics
20.
Glia ; 54(2): 125-34, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16718685

ABSTRACT

Our previous work has shown that tolbutamide increases gap junctional permeability in poorly coupled C6 glioma cells and that this effect is similar and additive to that found with dbcAMP, a well-known activator of gap junctional communication. Furthermore, the increase in gap junctional communication promoted by tolbutamide or dbcAMP is concurrent with the inhibition of proliferation of C6 glioma cells. In the present work, we show that tolbutamide and dbcAMP increase the synthesis of the tumor suppressor protein Cx43 and that they decrease the level of Ki-67, a protein expressed when cells are proliferating. These effects were accompanied by a reduction in the phosphorylation of pRb, mainly on Ser-795, a residue critical for the control of cell proliferation. The decrease in the phosphorylation of pRb is not likely to be mediated by a reduction in the levels of D-type cyclins, since instead of decreasing the expression of cyclins, D1 and D3 increased slightly after treatment with tolbutamide or dbcAMP. However, the Cdk inhibitors p21 and p27 were up-regulated after treatment with tolbutamide and dbcAMP, suggesting that they would be involved in the decrease in pRb phosphorylation. When Cx43 was silenced by siRNA, neither tolbutamide nor dbcAMP were able to up-regulate p21 and consequently to reduce glioma cell proliferation, as judged by Ki-67 expression. In conclusion, tolbutamide and dbcAMP inhibit C6-glioma cell proliferation by increasing Cx43, which correlates with a reduction in pRb phosphorylation due to the up-regulation of the Cdk inhibitors p21 and p27.


Subject(s)
Connexin 43/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Glioma/metabolism , Retinoblastoma/metabolism , Tolbutamide/pharmacology , Animals , Cell Proliferation/drug effects , Connexin 43/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Glioma/genetics , Glioma/pathology , Phosphorylation/drug effects , Rats , Retinoblastoma/genetics , Retinoblastoma/pathology , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...