GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Experimental Hematology, Elsevier BV, Vol. 38, No. 2 ( 2010-02), p. 141-153
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2005403-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1585-1585
    Abstract: Although estrogens have been shown to regulate normal B cell proliferation and differentiation, the impact of estrogen receptor (ER) signaling on B cell malignancies, including Waldenström Macroglobulinemia (WM), remains unexplored. To address this issue, we have analyzed, through preclinical WM models, the druggability either of the classical estrogen receptor ERβ, an emerging target for acute leukemias and certain types of lymphomas, and of the novel, more recently deorphanized G-protein coupled ER GPER, whose therapeutic potential in solid tumors has recently emerged. Both ERβ and GPER were found expressed at mRNA and protein level in a panel of WM and IgM-secreting lymphoma cell lines. However, while treatment with ERβ agonists did not elicit any effect, selective GPER activation significantly reduced WM cell viability. On this basis, we investigated GPER expression pattern and therapeutic potential in WM. By interrogating public microarray datasets, we found GPER transcript significantly upregulated in WM as compared to normal B cells; moreover, immunohistochemical analysis showed elevated GPER expression in lymph node biopsies from newly-diagnosed WM patients as compared to healthy lymph nodes. Treatment with the selective GPER agonist G-1 ( (±) -1-[(3aR*,4S*,9bS*)-4-(6-Bromo-1,3-benzodioxol-5-yl) -3a,4,5,9b-tetrahydro-3H cyclopenta [c]quinolin-8-yl] ethanone) led to reduced viability, clonogenicity and migration of WM and IgM-secreting lymphoma cell lines (IC50 at 48h ranging between 2.5 and 5.0 mM), including MYD88 and CXCR4 mutated cell lines, and even in the presence of bone marrow-derived stromal cells, while it did not affect healthy CD19+ B cell viability; conversely, GPER antagonists G-36 and G-15 slightly enhanced cell proliferation. The growth inhibitory activity of G-1 was associated with accumulation in G2/M cell cycle phase and induction of apoptosis, the latter phenomenon assessed by Annexin V/7AAD staining, analysis of mitochondrial membrane depolarization and cleavage of caspase 3, 7 and 9; hybridization of an antibody-array also highlighted G-1-induced down-regulation of anti-apoptotic proteins, including survivin and Bcl2. A significant effect of G-1 was also observed in primary tumor cells from refractory WM patients. Moreover, G-1 synergistically enhanced bortezomib and ibrutinib cytotoxicity in vitro. Importantly, intraperitoneal injection of G-1 (2mg/kg) significantly reduced the growth of BCWM-1 xenografts in NOD/SCID mice. To gain further insight into the consequences of selective GPER activation, we performed gene expression profile and GSEA analysis of WM cell lines treated with G-1. Of note, the p53 signaling pathway was strongly induced upon GPER activation, and upregulation of p53 and of its target genes or microRNAs (p21CIP1, Bax, Bad, PUMA and miR-34a) was confirmed both in G-1 treated WM cell lines and primary WM cells; moreover, G-1 combination with bortezomib or ibrutinib led to stronger increase in p53 and p21CIP1 levels as compared to single agent treatment. Finally, p53 knock-down partially reversed G-1-dependent anti-WM effects, thus suggesting p53 as downstream mediator of GPER. In summary, we report a significant anti-tumor activity upon selective GPER activation in WM cells, with apoptosis induction and a potent synergistic anti-WM activity in combination with ibrutinib and bortezomib. Altogether, these preclinical data suggest GPER agonists as a potential therapeutic option in WM. Disclosures Munshi: OncoPep: Other: Board of director.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3107-3107
    Abstract: Background- Multiple Myeloma (MM) is a hematologic malignancy strongly characterized by genomic instability, which promotes disease progression and drug resistance. We previously demonstrated that LIG3-dependent Alt-NHEJ repair is involved in genomic instability, drug resistance and survival of MM cells. On these premises, we investigated PARP1 as driver component of Alt-NHEJ pathway and new therapeutic target in MM. Materials and methods- Cell proliferation and apoptosis were evaluated with CellTiter-Glo assay and Annexin V staining. Alt-NHEJ repair was evaluated using EJ2-GFP. PARP1, Caspase 3, MYC and DNA Damage Response protein levels were analyzed by Western blot of whole protein extracts. In vivo anti-MM activity was evaluated in NOD-SCID mice bearing subcutaneous H929 and AMO-1 Bortezomib resistant (ABZB) xenografts, daily treated with Olaparib (Selleckchem) via oral gavage. Results - By interrogating public available datasets, we found significant correlation between higher mRNA expression of PARP1 and shorter survival of MM patients. On these findings, we investigated the effect of available PARP inhibitors (PARPi) on MM cell survival. We found that Olaparib, a clinically available PARPi induced a significant reduction of proliferation and clonogenic growth of MM cell lines at low micromolar concentrations. Importantly, Olaparib impaired viability of MM cell lines or primary malignant plasmacells co-cultured with stromal cells, thus overcoming the bone marrow microenvironment supportive effect for MM survival. As result of PARP-mediated Alt-NHEJ repair inhibition, anti-proliferative effects were associated to increase of DNA double-strand breaks (DSBs), activation of DNA damage response, cell cycle arrest and finally apoptosis. To identify predictive biomarkers for PARPi in MM, a published sensitivity gene expression signature was applied to our MM gene expression profiling (GEP) data. Interestingly, this signature was particularly enriched in TC2 MM and secondary plasma cell leukemia (PCL). Therefore, in order to evaluate concordantly modulated sets of genes that were possibly associated to PARPi signature in MM, PARPi-positive and PARPi-negative MM-TC2 cases were compared by GSEA analysis. Interestingly, groups of genes regulated by MYC or involved in DNA repair resulted among the most significantly up-regulated in PARPi-positive versus PARPi-negative MM-TC2 cases. Accordingly, MYC transcript reached the highest median expression levels in sPCL and HMCLs across PC dyscrasia groups, and in MM-TC2 class. Consistently U266 cells, which was quiet insensitive to PARP knockdown or PARP inhibitor Olaparib, were null for c-MYC as compared to multiple myeloma cell lines evaluated in this study. Conversely, as formal proof of our hypothesis, over-expression of c-MYC in U266 cells (MYC-OE) induced cell death upon PARP silencing or PARP inhibitor treatment. Notably, we found that c-MYC-PARP1 loop was also hyper-activated in Bortezomib resistant cells, thus confirming pivotal role of Alt-NHEJ repair in drug resistance development. Remarkably, to demonstrate the in vivo relevance of our findings, we showed that clinically available Parp-inhibitor Olaparib exerted a significant anti-MM activity on both Bortezomib sensitive (H929) and resistant (ABZB) MM cells injected in immunocompromised mice. Conclusion Taken together, our findings indicate that MM cells are dependent on PARP-mediated Alt-NHEJ repair pathway, which therefore represents a novel druggable target pathway in MYC-driven MM cells. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2740-2740
    Abstract: Multiple myeloma (MM) is a plasma cell malignancy with the high capacity to induce osteolytic bone lesions. Whereas previous studies identified genes overexpressed by MM cells related to the bone status, the occurrence of transcriptional alterations in the bone microenvironment cells in the relationship with the bone involvement has not yet investigated. To clarify this issue, in this study we have analyzed the gene expression profiling of mesenchymal (MSC) and osteoblastic (OB) cells obtained from MM patients (n=24; osteolytic n=10; non-osteolytic n=14) in relationship with the presence or absence of osteolytic bone lesions. MGUS subjects (n=7) and healthy donors (n=8) were also included in the study as controls. Both MSC and OB were isolated from trabecular bone biopsies without in vitro differentiation. The presence of potential contaminating cells was excluded by FACS analysis in both MCS and OB, testing CD3, CD14, CD20 and CD138 antigens, as well as the expression of CD105 and CD146; the osteoblast-related markers Osteocalcin, Alkaline Phosphatase, Collagen I and Runx2 were evaluated in OB in comparison with MSC. Thereafter a gene expression profiling analysis of isolated MSC and OB cells was performed using GeneChip® HG-U133A oligonucleotide arrays. The obtained data were validated by real time PCR. Using conventional hierarchical clustering, the unsupervised analyses performed of the whole dataset generated a dendrogram clearly distinguishing MSC and OB cellular types. When considering MSC and OB dataset separately, a preferential clustering in relation to the presence of osteolytic bone lesions was observed for MSC but not OB samples. A supervised multi class analysis identified a total of 84 probe sets differentially expressed in MSC with an intermediate transcriptional profile in MGUS-MSC between osteolytic and non osteolytic MM patients. A supervised analysis performed on MSC MM samples revealed a total of 49 probe-sets (36 up-regulated and 9 down-regulated genes) as differentially expressed in osteolytic vs. non-osteolytic patients. Specifically, genes belonging to Wnt signaling as WNT6 and extracellular matrix structure as decorin (DCN) were found to be down-regulated in osteolytic as compared to and non-osteolytic MSC. Interestingly, no significantly modulated genes were found by comparing osteolytic and non-osteolytic OB samples. Finally, we performed two distinct supervised analyses by comparing the two cellular types (MSC and OB) in the two groups of MM patients in relationship with the bone status. A distinct transcriptional pattern was observed in MSC versus OB between osteolytic and non-osteolytic MM patients (52 vs. 21 differentially expressed probe-sets, respectively), mainly involving cell-cycle realted genes. Our results highlight that in MM bone microenvironment MSC rather than OB show transcriptional alterations in relationship with the presence of osteolytic bone lesions in MM patients.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2925-2925
    Abstract: Abstract 2925 Primary plasma cell leukemia (PPCL) is an aggressive variant of multiple myeloma, accounting for 0.5–4% of all newly diagnosed myeloma cases and characterized by a short survival (generally less than 1 year), which is only moderately improved by transplant procedures. Novel agents seem to be able to ameliorate the poor clinical outcome of both primary and secondary leukemic phases of myeloma; however, no data are currently available on the use of lenalidomide as first line therapy in PPCL. On March, 2009, we started a multicenter, phase II trial aiming to evaluate safety and antitumor activity of lenalidomide in combination with dexamethasone (LD) in previously untreated PPCL. Here we report the final results of this study. Newly diagnosed PPCL patients received lenalidomide at a dose of 25 mg/d for 21 days and oral dexamethasone at a dose of 40 mg on days 1, 8, 15, and 22 for each 28-day cycle. After 4 cycles, responding patients not eligible for stem cell transplantation (SCT) continued until 8 cycles of full-dose LD, if tolerated, followed by a maintenance dose of single agent lenalidomide equal to 10 mg/d on days 1–21 of each 28-day cycle. Patients responding after 4 cycles and eligible for SCT proceeded according to single Centre transplant policy. Patients not responding after 4 cycles or progressing during this treatment were considered off-study. The primary endpoint was early response rate according to International Uniform Criteria. The secondary endpoints were PFS, OS, safety and percentage of eligible patients able to undergo autologous or allogeneic SCT. Appropriate dose reductions, contraception methods and anti-thrombotic prophylaxis were applied. Twenty-three patients, as requested by the Simon Optimal Two-Stage Adaptive Design adopted, were enrolled. The trial was therefore closed on May, 31, 2011. M/F ratio was 0.7, mean age was 62 years (range 44–80). Circulating plasma cells ranged from 2.1 to 115 × 10e9/l. Moderate renal failure, increased LDH and extramedullary disease occurred in 39.1%, 43.5% and 13 % of patients, respectively. Hb was 〈 10 g/dl in 19 patients (82.6%), while platelet count was 〈 50 × 10e9/l in 5 patients (21.7%). Karyotype abnormalities were detected by FISH in 21 out of 22 tested patients; in particular, 1p loss was found in 9 patients, 1q gain in 10 patients, del(13q) in 16 patients, del(17p13) in 7 patients, t(11;14) in 7 patients, t(4;14) in 3 patients and MAF translocations, including t(14;20) and t(14;16), in 8 patients. Seventeen patients had a combination of two (n. 5) or more (n. 12) cytogenetic lesions. On intention-to-treat analysis, 14 patients completed the initial four planned cycles and all of them responded. In particular, 6 PR (26.1%), 4 VGPR (17.4%), 1 near-CR (4.3%) and 3 CR (13%) were achieved (ORR 60.8%, VGPR or better 34.7%). Causes of early treatment discontinuation were: a) progressive disease (4 patients, after an initial, brief response in 2 cases); b) severe adverse events (4 patients: one acute renal failure, one Stevens-Johnson's syndrome, one pneumonia suspected for Pneumocystis carinii etiology, one multi-organ failure); c) death in PR due to causes unrelated to treatment or disease (one patient). Other relevant non-hematological toxicities included four episodes of pneumonia and one case of DVT. Grade 3–4 hematological toxicities occurred in about half of cases, requiring Lenalidomide dose adjustments. So far, among subjects achieving a response after 4 LD cycles, 8 eligible patients have successfully collected peripheral blood stem cells: 5 of them have completed single or double autologous SCT, one patient received tandem autologous-allogeneic non myeloablative SCT from a MUD donor. All patients transplanted after LD are currently alive and in remission phase. The maintenance phase has been reached in 3 responding patients not eligible for SCT, 2 of whom have relapsed after 2 and 8 months, respectively. With a mean follow-up of 15 months, OS and PFS are 65.2% and 52.1%, respectively. LD is a possible initial therapeutic option for PPCL, particularly in patients who receive SCT after a short course of induction treatment. Caution is required to prevent and to manage renal and hematological toxicities, as well as infectious complications. Considering some previous results obtained with other novel agents, the combination of lenalidomide and bortezomib might be an appealing approach to investigate prospectively in PPCL patients. Disclosures: Musto: Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Petrucci:Celgene: Honoraria. Cascavilla:Celgene: Honoraria. Di Raimondo:Celgene: Honoraria. Caravita:Celgene: Honoraria. Morabito:Celgene: Honoraria. Offidani:Celgene: Honoraria. Bringhen:Celgene: Honoraria. Boccadoro:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Palumbo:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4040-4040
    Abstract: Abstract 4040 Multiple myeloma (MM) is a clonal proliferation of malignant plasma cells (PCs) characterized by a marked genomic instability. Primary plasma-cell leukemia (pPCL) is an aggressive, rare variant of plasma cell dyscrasia characterized by extra-medullary proliferation of PCs, high genomic instability and very poor prognosis. To date, global genomics studies in pPCL are still limited. Highly purified PCs were obtained from 17 previously untreated pPCL patients, recruited in an open-label, exploratory, single-arm, two-stage study from the GIMEMA myeloma network aimed at the evaluation of safety and antitumor activity of the immunomodulatory agent lenalidomide in combination with low dose dexamethasone in previously untreated pPCL. All the samples were characterized for the main chromosomal aberrations by Fluorescence in-situ hybridization (FISH). Specifically, 13q and 17p deletions have been identified in 12/17 (70.6%) and 8/17 (47.1%) cases, respectively; the presence of t(11;14) was found in 4/17 patients (23.5%), t(4;14) in only 1/17 cases (5.9%) and MAF translocations in 8/17 (47.1%). To further investigate the genomic complexity of pPCL, we analyzed a subset of 13 samples by means of an integrative approach using different high-throughput microarray technologies: Human Mapping 250K Nsp SNP-array (Affymetrix) for the detection of copy number alterations (CNA), Gene 1.0 ST array (Affymetrix) for transcriptional profiling and miRNA Microarray v2 (Agilent) for the global miRNA expression. SNP-array data were concordant with FISH results for the detection of the main chromosomal aberrations, i.e. 13q (10/13=77%) and 17p (7/13=58.3%) deletions. The most recurrent CNA specifically identified by SNP-array was represented by 1q gain (8/13=61.5%); in addition. losses involving chromosomes 1p (5/13=38.5%), 8p (4/13=30.8%), 14q (5/13=38.5%), 16q (5/13=38.5%), gains affected 7q (4/13=30.8%) and 19p (4/13=30.8%) and one amplification at 17q21 in 6/13 pPCL (46.2%) were detected. One case displayed a near tetraploid karyotype and, interestingly, another one showed a hyperdiploid pattern. Mapping information was integrated with the gene expression and miRNA profiles of the tumor samples. A non-parametric analysis (Kendall's tau correlation at a p-value 〈 0.005) identifying 199 probes whose expression levels strongly correlated with the occurrence of allelic imbalances. Most of those probes (181/199=91%) were localized in the previously described altered regions; specifically, chromosomes 1p (12.6%), 1q (40.7%), 7q (3.5%), 8p (6.0%), 13q (4.5%), 14q (9.5%), 16q (6.5%) and 17p (7.5%). The same integrative approach has been applied to investigate the correlation between miRNA expression levels and the CNAs: 23 miRNAs had been detected at a p 〈 0.05, most of them (16/23=69.5%) mapped to chromosomes 1p (21.7%), 13q (26.1%) and 19 (21.7%). These results highlight a wide gene-dosage effect suggesting that genomic structural abnormalities in pPCL closely reflect in expression imbalances. Our integrative approach data provide insights into the characterization of novel genetic lesion in pPCL, and suggest that a wide gene- and microRNA-dosage effect is a common characteristic of plasma cell dyscrasias, as previously described by our group in multiple myeloma PCs. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1259-1259
    Abstract: Abstract 1259 Poster Board I-282 We describe the clinical-biological features of 63 cases of variant B chronic lymphocytic leukemia (v-B-CLL), characterized by a mantle cell lymphoma-like immunophenotype, atypical cytomorphology in absence of t(11;14)(q13;q32) in FISH analysis. A historical series of 130 B-CLL was used as comparison. The v-B-CLL were significantly different from the B-CLL in terms of the following clinico-hematological variables: age 〈 70 yrs (p 〈 .001), lymphocytosis 〈 20 × 109/ (p 〈 .001), lymphocyte doubling time 〈 12 months (p = .02), high serum β2-microglobulin levels (p 〈 .001), and splenomegaly (p = .002). Considering immunophenotipic features, CD38 and CD49d expression were significantly more expressed in v-B-CLL than B-CLL (p 〈 .001); whereas, no statistical difference was observed for ZAP-70 reactivity. Considering the IgVH mutation status, there were more patients mutated in the v-B-CLL group than in the B-CLL group (p = .001). Other significant differences were found about the frequency of the recurrent chromosome alterations, evaluated by means of FISH analysis: trisomy 12 was more frequent in v-B-CLL ( p 〈 .001), while del13q14, considered as a single alteration, was more frequent in B-CLL (p=.008). Gene expression profiling of a panel of 9 v-B-CLL compared with 60 B-CLL samples indicated that the variant group is characterized by a specific molecular pattern of gene expression. In particular we observed the upregulation of tumor protein D52 (TPD52), and that of 6 genes (AFF1, GMPS, PICALM, JUN, REL, RAC2) known to be protooncogenes. Furthermore, we found that upregulated genes with apoptosis related functions (IL-7, HSP90B1, NOTCH2, BECN1, ANXA4, MCL1) are all negative regulators of apoptosis. Microarray analysis revealed that various genes (TRIM38, EEF1D, CASP1, MALT1, RHOH0) involved in the I-kB kinase/NF-kB cascade of the canonical NF-kB signaling pathway, are furtherly upregulated in v-B-CLL. Furthermore, among the genes found differentially expressed in SAM analysis, we observed also the upregulation of CD1c (according to the surface expression of this antigen), OSBPL3 and ITGA4. After a median follow-up of 55 months (range 4-196) and 60 months (range 6-180), 25/42 (59%) v-CLL and 55/93 (59 %) CLL pts were treated. TTT was significant different between 2 groups when the IgVH mutational status was considered (p= .006). Median OS of v-CLL subset was 112 months vs 171 months of CLL subset. When the IgVH mutational status was considered, mutated cases showed a worse OS even if a statistical difference was not observed (p= 0.062). In conclusion, our study identifies, on the basis of a defined CFM-FISH diagnostic approach, a variant form of B-CLL that shows peculiar biological and clinical features that should be considered in the future clinical and prognostic studies. The inclusion of this form in B-CLL study could alter the interpretation of results, especially related to biological markers. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1822-1822
    Abstract: The human fibroblast growth factor receptor (FGF-R) family plays an essential role in a wide range of cellular processes, such as cell growth, proliferation, differentiation, migration and survival. It has been reported that FGF-Rs are expressed in hematopoietic cells; and FGF/FGFR signaling deregulation is largely involved in hematologic malignancies, including Waldenström macroglobulinemia (WM). WM is still an incurable disease, and patients succumb due to disease progression. Therefore, novel therapeutics designed to specifically target deregulated signaling pathways in WM are required. We aimed to investigate the role of FGF/FGF-R system in FGF-dependent WM cell lines by using an anti-pan FGF trap molecule (NSC12), responsible for FGF/FGF-R blocking. We first interrogated the GSE9656 dataset in order to confirm the expression of FGFs and FGF-Rs in WM cells, demonstrating an enrichment of several FGF- and FGF-R-isoforms in primary WM patients' derived tumor cells compared to the normal cellular counterpart (P 〈 0.05); and demonstrated the ability of NSC12 to inhibit FGF-secretion within the conditioned media of NCS12-treated WM cells, as shown by ELISA. Wide-transcriptome profiling of NSC12-treated WM cells (BCWM.1; MWCL1) revealed a significant inhibition of Myc-target related genes, coupled with silencing of genes involved in cell cycle progression, cell proliferation, PI3K-AKT-mTOR signaling, oxidative phosphorylation (Hallmark; FDR 〈 0.25; P 〈 0.05). This prompted us to evaluate the anti-tumor functional sequelae exerted by NSC12 in WM cells: NSC12 induced significant inhibition of WM cell growth (BCWM1 and WMCL1) in a dose-dependent fashion (0.1-10μM; IC50 ~3μM), even in the presence of bone marrow microenvironment. In addition, a significant effect was also observed in primary tumor cells from WM patients; while no effect was observed on healthy donor-derived peripheral blood mononuclear cells. The growth inhibitory effect was associated with induction of apoptotic cell death, caspase activation and PARP cleavage, as demonstrated by flow cytometry and western blot, respectively. Moreover, we also observed a NSC12 dose-dependent increase of mitochondrial reactive oxigen species (ROS), at protein level. Cell cycle analysis revealed a reduction of the S-phase and increase of G0/G1 phase. Mechanistically, NSC12 targeted WM cells by inhibiting MAPK, JAK/STAT3 and PI3K-Akt pathways known to be FGFRs-activated signaling cascades. Importantly, the same effect was maintained in WM cells even in the presence of the supporting BM microenvironment. Functional studies demonstrated the ability of NSC12 to impair the adhesion of both cell lines to the supportive primary bone marrow stromal cells, in vitro. NCS12-dependnet anti-WM activity was also tested in combination with bortezomib, carfilzomib, everolimus and ibrutinib: the combinatory treatment (48h) resulted in a more significant dose-dependent inhibition of WM cell survival and proliferation (P 〈 0.05), thus suggesting the rational for combining FGF-blockade with proteasome-, mTOR-, or BTK-inhibitors. In vivo studies are being performed, in order to further corroborate the anti-WM activity of NSC12 using WM animal models. Disclosures Ronca: Associazione Italiana per la Ricerca sul Canctro (AIRC): Research Funding. Rossi:Astellas: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria; Mundipharma: Honoraria; BMS: Honoraria; Sandoz: Honoraria; Amgen: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Jazz: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Consultancy; Roche: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Roccaro:AstraZeneca: Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Associazione Italiana per al Ricerca sul Cancro (AIRC): Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees; European Hematology Association: Research Funding; Associazione Italiana per al Ricerca sul Cancro (AIRC): Research Funding; Transcan2-ERANET: Research Funding; AstraZeneca: Research Funding; European Hematology Association: Research Funding; Transcan2-ERANET: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1814-1814
    Abstract: Abstract 1814 Poster Board I-840 Background The growth and survival of multiple myeloma (MM) cells in the bone marrow microenvironment is regulated by functional complex interactions between the tumor cells and the surrounding bone marrow stromal cells mediated by adhesion molecules and the production of several cytokines of which interleukin-6 (IL-6) has been identified as the most important. Major advances in the investigation of MM biology were made possible by the availability of human myeloma cell lines (HMCLs). The IL-6-dependent CMA-03 cell line was established in our laboratory from a peritoneal effusion of a refractory relapsed MM patient. By gradually decreasing the IL-6 added to the culture, an IL-6-independent variant, CMA-03/06, could be obtained. Aims. To perform a biological and molecular characterization of this novel cell line, and to provide insights into the signaling pathways and target genes involved in the growth and survival of CMA-03/06. Methods. The growth, immunophenotypic, cytogenetic and fluorescence in situ hybridization (FISH) characterization of CMA-03/06 cell line was performed by means of standard procedures. IL-6 production into the culture media was determined using a high sensitivity IL-6 specific ELISA. Genome-wide profiling data were generated by means of Affymetrix GeneChip® Human Mapping 250K Nsp arrays; copy number (CN) alterations were calculated using the DNAcopy Bioconductor package, based on circular binary segmentation method. Global gene expression profiling (GEP) was performed by means of the GeneChip® Human Gene 1.0 ST Arrays (Affymetrix); the supervised analyses were done using the SAM software version 3.0. Results Unlike CMA-03, the addition of IL-6 to the culture medium of CMA-03/06 cells or co-culture with multipotent mesenchymal stromal cells did not induce an increase in CMA-03/06 proliferation. IL-6 was not detected in the supernatants from either CMA-03 or CMA-03/06 cell lines within 48 h, suggesting that the IL-6 independence of CMA03/06 cells is not a result of the development of an autocrine IL-6 loop. Nevertheless, IL-6 induced the activation of STAT3 and STAT1 in both cell lines, even if a slight constitutive STAT3 phosphorylation was found in CMA-03/06. The immunophenotypic analysis showed a significant difference in the expression of three antigens in the 2 cell lines: CD45 was considerably reduced in CMA-03/06 cells, whereas they were found positive for both chains of IL-6 receptor, CD126 and CD130, almost undetectable in CMA-03. Conventional cytogenetic and FISH analyses did not reveal differences between the 2 HMCLs. The genome-wide analysis allowed the identification of about 100 altered chromosomal regions common to both HMCLs, mostly DNA gains. Comparison of CMA-03/06 and CMA-03 cells evidenced a different CN in only 15 small chromosomal regions, 8 of which did not contain any transcript, whereas few genes were located on the other ones. GEP analysis of CMA-03/06 compared with CMA-03 identified 21 upregulated and 47 downregulated genes, many of which particularly relevant for MM biology, mainly involved in cellular signaling, cell cycle, cell adhesion, cell development, regulation of transcription, immunologic, inflammatory or defense activity, apoptosis. None of the genes differentially expressed in CMA-03/06 compared with CMA-03 except 1 were positioned on the chromosomal regions showing a different CN. Finally, CMA-03/06 cell line showed a lower susceptibility to camptothecin-induced apoptosis compared to CMA-03 cells. Conclusions Our data show the IL-6 independence of CMA-03/06 cell line in the absence of an autocrine IL-6 loop; the cells, however, maintain the IL-6 signaling pathway responsiveness. A consistent number of genes particularly relevant for MM biology were found deregulated in CMA-03/06 cell line compared with CMA-03. Furthermore, CMA-03/06 cell line shows an increased resistance to apoptosis. The novel CMA03/06 cell line may thus represent a suitable model for studies investigating molecular mechanisms involved in clonal evolution towards IL-6 and/or stroma-independent growth and survival of myeloma cells. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 2824-2824
    Abstract: Abstract 2824 Poster Board II-800 Multiple myeloma (MM) is a malignant proliferation of bone marrow (BM) plasma cells (PCs), characterized by a profound genomic instability involving both numerical and structural chromosomal aberrations of potential prognostic relevance. The discovery of microRNA (miRNA) genes, encoding for a class of small non-coding RNAs involved in the regulation of cell cycle, survival and differentiation programs, has added a further level of complexity to normal and cancer cell biology; it has been suggested that chromosomal abnormalities and other types of genetic or epigenetic alterations might contribute to miRNA deregulation in cancer. To date, only little evidence of miRNA expression/deregulation in multiple myeloma (MM) has been reported. To characterize miRNA expression profiling of MM plasma cells (PCs) and integrate miRNA expression data with other molecular features of MM patients, global miRNA expression profiles of PCs isolated from BM biopsies of 38 newly diagnosed MM, 2 plasma cell leukemia patients, and 3 healthy donors were generated using the Agilent Human miRNA microarray V2 (representing 723 human mature miRNAs from the Sanger miRBase v10.1). All of the patients had previously been characterized by FISH for the main IGH translocations and other genetic abnormalities; they were also profiled for global gene expression by means of Affymetrix U133A arrays; nineteen of the patients profiled for miRNA expression underwent genome-wide DNA analysis using Affymetrix GeneChip® Human Mapping 50K XbaI microarrays. An unsupervised analysis of the samples, using conventional hierarchical clustering algorithm and based on the most variably expressed miRNAs across the dataset, grouped the PCs from healthy donors separately from MM PCs; among the pathological samples, the most striking finding was that the seven patients with t(4;14) (TC4) were tightly clustered, as were four out of the five samples with translocated MAF genes (TC5). A partial grouping of the TC2 cases (mostly hyperdiploid) was also observed, whereas the TC1, showing t(11;14), and TC3 (mostly expressing Cyclin D2), samples were dispersed along the dendrogram. A multiclass supervised analysis of the miRNA expression between the members of the 5 TC groups highlighted specific miRNA signatures, in particular characterizing the TC4 and TC5 groups. A slight miRNA signature was observed in t(11;14) cases and TC2 group, whereas we could not identify any TC3-specific miRNA signature. None of the differentially expressed miRNAs in patients with specific IGH translocations maps to the rearranged chromosomal regions. The levels of some differentially expressed miRNAs were quantified by means of Q-RT-PCR in all of the 43 samples, using specific TaqMan® microRNA assays (Applied Biosystems); a linear correlation analysis indicated very good correspondence between microarrays and Q-RT-PCR data. Furthermore, the expression of specific miRNAs was also evaluated in 14 additional cases, four carrying t(11;14), seven t(4;14), and three t(14;16) or t(14;20): as well, in this broaden panel the selected miRNAs confirmed their TC-specific over-expression. The occurrence of other lesions, such as 1q gain, 13q and 17p deletions, and hyperdiploidy, was slightly characterized by specific miRNA signatures. Furthermore, the integrated analysis with the genomic profiles revealed the occurrence of several allelic imbalances significantly associated with the altered expression of miRNAs located in the involved regions, such as let-7b at 22q13.31 (loss) and miR-142-3p at 17q22 (gain), or miR-140-3p at 16q22 (loss-of-heterozygosity). Finally, in an attempt to define the consequences of deregulated miRNA expression, we performed an integrative analysis based on computational target prediction, miRNA and mRNA profiling. Specifically, we searched for putative functional targeting relationships in MM cells supported by expression data, i.e. anti-correlations between miRNA and target mRNA expression profiles, and thus defined a global miRNAs/mRNAs regulatory network. Our data provide the first evidence of miRNA deregulation in the context of the molecular subtypes of MM, and represent the first attempt to define the complex of miRNAs/mRNAs regulatory relationships, aimed at deepening our understanding of the involvement of specific miRNAs and target genes in the pathology of the disease. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...