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: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2774-2774
    Abstract: Introduction: The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment is fundamental to MM pathogenesis. Cell adhesion-mediated drug resistance (CAM-DR) is regulated by adhesion receptors on MM cells such as CXCR4, CXCR7, CD49d and CD44. We and others have previously reported that CAM-DR towards drugs like bortezomib, pomalidomide or vorinostat may be dissolved by combining these novel agents with the CXCR4 inhibitor plerixafor. Different than expected, additional treatment with plerixafor in corresponding experiment however did not rescue the cytotoxic effects of the second generation proteasome inhibitor carfilzomib. We hypothesized that carfilzomib itself interferes with the CXCR4-CXCL12 axis in myeloma. Prior reports in AML and CLL indicate that PIM1-mediated CXCR4 phosphorylation at the position S339 is an essential step for CXCR4 recirculation to the cell surface and its function as CXCL12 receptor (Grundler et al. 2009, Decker et al. 2014). In this project, we therefore examined the effects of carfilzomib on the PIM1-CXCR4 axis as a not yet described, potentially class-distinct mechanism of action of this second generation proteasome inhibitor. Methods: U266, RPMI-8226, L363, MOLT-4, NCI-H929 and the stromal cell line M2-10B4 were utilized. Bortezomib (1, 10, 20, 50, 100nM), carfilzomib (20, 50, 100nM) and plerixafor (10, 50, 100μM) were used based on previous studies and are well comparable to clinically relevant doses. CXCL12 stimulation was performed with human recombinant CXCL12 (30nM). For combination studies, cells were preincubated with plerixafor (50µM). Viability was quantified by propidium iodide and annexinV-FITC using flow cytometry. For quantitative real-time PCR and Western blots, U266 monocultured cells were treated with a carfilzomib pulse (t=1h), were allowed to recover for 20 hours, starved for 4 hours and stimulated with CXCL12 for 15 minutes (n=4). PIM-1 mRNA transcript levels were assessed in U266 control vs. U266 treated with a carfilzomib pulse (100nM, t=1h) by qPCR. Data was analyzed according to the "delta-delta-CT method" based on the relative expression of PIM-1 vs. GAPDH. Results were normalized to the mean of the control samples. Results: FACS analyses determined a substantial decrease of CD138 and CXCR4 surface expression in a dose-dependent manner after 1h carfilzomib treatment of U266 cells. Further assessment of downstream signaling revealed that carfilzomib treatment significantly reduces CXCR4 phosphorylation at S339 without changing total levels of CXCR4 (Figure A) or total levels of ERK or pERK (not shown), excluding a general inhibition of phosphorylation or protein synthesis by carfilzomib. Following the hypothesis that CXCR4 is potentially phosphorylated by PIM1 kinase, we assessed the impact of carfilzomib on PIM-1 protein levels: PIM-1 kinase protein was significantly reduced in a dose-dependent manner along with the levels of pCXCR4 in response to increasing doses of carfilzomib (0-100nM, Figure B). To further investigate a possible direct interference at the mRNA level, we evaluated PIM-1 mRNA levels after 1h carfilzomib, confirming substantially reduced PIM-1 RNA transcripts (Figure C). Different from carfilzomib and in line with prior observations (Shay et al. 2005), bortezomib was shown to increase protein levels of PIM-1 (data not shown). Side-by-side comparative assays of bortezomib vs. carfilzomib in terms of reduced CXCR4 expression, decreased CXCR4 phosphorylation and PIM-1 levels on mRNA and protein level are currently ongoing and will be presented at the meeting. Conclusions: Similar to previous reports on ixazomib reducing PIM-1 on protein and mRNA levels by inhibiting the tumor-suppressive microRNA miR33b (Tian et al. 2012), this work provides a potentially distinct mechanism of action of the second generation proteasome inhibitor carfilzomib on the PIM1-CXCR4 axis and identifies PIM-1 as a valid target to overcome CAM-DR in multiple myeloma. Figure Carfilzomib overcomes stroma protection due to PIM-1 kinase inhibition. Figure. Carfilzomib overcomes stroma protection due to PIM-1 kinase inhibition. Disclosures Engelhardt: Janssen: Research Funding; Amgen: Research Funding; MSD: Research Funding; Celgene: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Oncotarget, Impact Journals, LLC, Vol. 8, No. 13 ( 2017-03-28), p. 21153-21166
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1822-1822
    Abstract: Introduction: Novel substances such as the next generation IMiD pomalidomide or the recently approved next generation proteasome inhibitor carfilzomib (Cfz) have considerably expanded our treatment options in MM, both of them influencing multiple myeloma (MM) interaction with bone marrow stroma cells (BMSCs), that provides an interesting target for anti MM therapy. More compounds directed at this disease critical crosstalk are currently under investigation, however the development of novel drugs remains inefficient, displayed by a substantial drop out rate of the 376 preclinical single agents tested since 1961 (Rongvaux, Annu Rev Immunol. 2013; Schüler, Expert Opin Biol Ther. 2013; Kortüm, CLML 2014). Our focus in the projected presented here was to develop a novel bone-derived in vitro 3D co-culture model specifically adapted to mimic the BM niche to more closely study the role of bone and BM bystander cells and to perform more reliable ex vivo compound screening in MM. Methods: Previous 2D models were compared to a novel 3D co-culture model (agarose matrix interlayer, 100 microwells/cm², 1.5mm in depth, permeable for oxygen+cytokines, but not for BMSCs utilizing U266, RPMI-8226, OPM-2 and primary BM patient (pt) cells, with and without HS-5 vs. M210B4 stroma support (Fig. A + Fig. B.a. for pt characteristics). Analyses covered Trypan Blue, Annexin/PI, MTT, FACS, cell cycle analyses and H2B-mCherry/cytochrome c-GFP assays (Udi, Br J Hematol. 2013). In a next step, primary bone-derived stroma cells were acquired from bones of C57BL/6 J mice. Bones were flushed, digested and FACS sorted in order to acquire single BM and bone bystander cell subtypes (MSPCs [mesenchymal stem and progenitor cells], endothelial cells, osteoblasts, PAS [PDFGRalphaSca1] and CaRs [CXCL12-abundant reticular cells]) which were then compared to HS-5 and M210B4 with regard to growth support, cytokine secretion and protection from anti-MM substances. Results: MMCLs and pt specimens were cultured at different concentrations (10 vs. 100 cells per microwell) with and without M210B4 demonstrating a growth advantage with vs. without M210B4 (Fig. B.b). Liquid overlay technique allowed cluster formation of pt specimens leading to more reliable propagation of pt material for up to 20d of culture. Apoptotic changes were assessed by confocal microscopy of RPMI8226 co-expressing fluorescently labelled histone 2B-mCherry (red) and cytochrome c-GFP (green) as indicators of late and early apoptosis. Comparing BMSCLs with regard to their MM growth support capacities, human HS-5 proved even more beneficent than M210B4 stroma (Fig. B.b). Phenotypic analyses of pt specimens showed decreased CXCR4 expression with vs. without BMSCs suggesting a dynamic regulation of homing molecules. The model was then used as an ex-vivo platform allowing both cytotoxicity and cell cycle analyses for the combination of bortezomib (Btz) vs. Cfz with ARRY-520 (kinesin spindle protein inhibitor). Btz (10nM) and Cfz (20nM) proved significantly cytotoxic compared to the control (U266 and pt specimens, respectively) after 48h of single agent treatment Fig.B.c). Compared to Btz (10nM, B10) and Cfz (20nM, C20) as single agents, the additional combination with ARRY-520 showed stronger additive cytotoxicity for Btz (A5+B10, median: 37.5% vs. 13.1%) than for Cfz (A5+C20, 38.6% vs. 34.1%). To note, the model could also be utilized for more profound analyses as depicted for G2/M cell cycle studies in Fig.B.d. 5nM ARRY-520 (A5) led to significant accumulation of OPM-2 cells in G2/M arrest after 48h of treatment confirming prior analyses (Hernández-Garcia Blood Suppl 4710,2014; Fig. B.d). Co-culture studies with different subsets of BM and bone-derived bystander cells are currently ongoing and will be presented at the meeting (Fig. B.e). Conclusions: More complex, 3D bone-derived high-throughput in vitro models are urgently needed to better predict the potency of preclinically tested agents and to better estimate the likelihood of their later clinical adoption into phase I-II trials. With this work, we provide an innovative model which reflects the BM microenvironment as a crucial predictor for in-vivo sensitivity as shown for ARRY-5200. This ex-vivo approach helps to better incorporate MM growth support by bone and BM-derived bystander cells and thus depicts a valid tool to better characterize the role of the BM niche in myeloma. Figure 1. Figure 1. Disclosures Engelhardt: Deutsch Krebshilfe: Other: grant. Wäsch:German Cancer Aid: Research Funding; Comprehensiv Cancer Center Freiburg: Research Funding; Janssen-Cilag: Research Funding; MSD: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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. 120, No. 21 ( 2012-11-16), p. 2450-2450
    Abstract: Abstract 2450 Introduction: The interaction between malignant plasma cells and their microenvironment is central in multiple myeloma (MM) pathogenesis. Binding of MM cells to bone marrow (BM) stroma cells triggers the expression of adhesion molecules and secretion of chemo- and cytokines, promoting MM cell growth, drug resistance and migration. Stromal-derived factor-1 (SDF-1) and its receptor CXCR4 are essential for normal hematopoietic progenitor cell movement and adherence within the BM microenvironment. In leukemia and lymphoma, oncoproteins may inhibit SDF-1-dependent cell trafficking within the BM through a mechanism that is not fully understood. For that reason, understanding SDF-1-dependent cell trafficking within the BM and targeting MM-cell - host-BM interactions display a promising approach for the development of novel therapeutic strategies. Methods: BM samples of MM patients (n=59) were analysed using flow cytometry and compared to MGUS patients (n=3) and healthy volunteers (n=7). We compared patient samples with low BM infiltration (≤5%; n=13) intermediate (5–30%; n=29) and high infiltration rates (≥30%; n=17). We also assessed expression of adhesion molecules in MM patients with long-term disease control (n=20) vs. both newly diagnosed (n=16) and symptomatic MM patients (n=23) as previously grouped by San Miguel et al. (Haematologica July 6,2012). We also sought to elucidate in vitro, whether specific anti-MM agents (bortezomib, vorinostat, pomalidomide, EGCG), with and without M210B4 stroma support, and with and without the CXCR4 inhibitor AMD3100, target the interaction of MM cells. Experiments were performed using MM cell lines (U266, RPMI8226, L363, NCI-H929), the control T-cell line MOLT-4 and MM-patient BM samples. Cell viability was assessed via Trypan Blue- and AnnexinV/PI-staining. CD138, CXCR4 (SDF1-receptor), CD49d (VLA-4), CD11a (LFA-1) and CD44 (HERMES antigen) expression was evaluated by flow cytometry and ScanR microscopy. Results: In BM samples of MM patients as compared to MGUS and healthy volunteers, the CXCR4/CD138- (p=.036), CD49d/CD138- (p=.0013) and CD44/CD138-expression (p=.0072) was significantly amplified and correlated with increasing BM infiltration rates (p=.001). Both newly diagnosed and symptomatic MM patients confirmed significantly increased CXCR4/CD138-, CD49d/CD138- (p=.0013) and CD44/CD138-expression as compared to patients with long-term disease control. Of note, in newly diagnosed patients, the expression of adhesion molecules was even more enhanced than in symptomatic myeloma patients, underlining their critical and future potential role as targets for novel therapeutics. Comparison of MM cell lines' adhesion and migration markers with that of MM-patient BM specimens revealed U266 as the cell line most closely resembling human specimens. Cytotoxic effects with use of MM cell lines and bortezomib, vorinostat and pomalidomide confirmed prior cytotoxic concentrations. Cocultivation with stroma substantially reduced apoptosis and induced tumor protective effects. Additional AMD3100 treatment restored sensitivity to bortezomib, vorinostat and pomalidomide. CXCR4 expression was substantially reduced after AMD3100 treatment, while that of CD49d, CD44 and CD11a remained widely unchanged. Toxic or therapeutic effects of AMD3100 monotherapy were excluded for used doses of 50μM. Additional use of ScanR microscopy visualized co-localisation of CXCR4 expression both on the cell surface and within the cytoplasm of MM cells. ScanR microscopy results correlated with flow cytometry-determined CXCR4 expression. Ongoing analyses of both ScanR microscopy and flow cytometry will allow the detailed assessment of treatment studies with and without anti-MM agents and AMD3100. Conclusions: Our findings underline the critical role of adhesion and migration molecules in MM and may pave the way for novel therapeutic approaches targeting these microenvironmental mediators. 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: 2012
    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. 124, No. 21 ( 2014-12-06), p. 927-927
    Abstract: Introduction: The interaction of malignant plasma cells (PC) with the bone marrow (BM) microenvironment (BMM) is fundamental for multiple myeloma (MM) pathogenesis, with BMM playing a key role in disease development, progression and patients’ symptoms. The CXCL12/CXCR4 axis is pivotal in this cross-talk. By activating CXCR4, CXCL12 drives MM cells to the protective BM and facilitates environment-mediated drug resistance. Whether or not CXCR7, the second receptor for CXCL12, is expressed by malignant PC has not been determined yet. In AML and CLL, the phosphorylation status of CXCR4 proved important: phosphorylation at Ser339 by PIM-1 kinase promoted re-externalization of CXCR4 and its reactivation to CXCL12 stimulation (Grundler et al. JEM 2009; Decker et al. Mol Cancer Therapeutics 2014). Methods: Human MM cell lines (MMCLs) U266, L363, NCI-H929 and primary BM specimens (patient samples n=9, median BM-infiltration: 30%; range: 15-90%) were used. MMCLs and primary MM patient specimens were assessed for CXCR4, CXCR7 and CD138 surface (s) expression via flow cytometry. For CXCR4 analysis, the conformation independent antibody clone 4G10 was used for both flow and image cytometry experiments. The MMCLs were cultured alone or with the stroma cell line M2-10B4, and treated with the novel proteasome inhibitor carfilzomib (20, 50, 100nM), either alone or combined with the CXCR4 antagonist AMD3100 (50µM) or anti-CXCL12 "Spiegelmer" NOX-A12 (100nM). Cell viability was assessed by PI/Annexin staining. CXCR4 downstream pathways and PIM-1 kinase expression were analyzed by Western blot. Results: All MMCLs and primary MM cells expressed sCXCR7. sCXCR7 expression was represented as mean (% of positive live cells) +/- SD: U266 (89.1 +/-7.1), L363 (82.1 +/-7.5) and NCI-H929 (82.4 +/-8.2) were largely CXCR7 positive. The sCXCR7 expression of unsorted primary BM cells was 21.7 +/- 10.5 (n=9), being slightly higher than their sCXCR4 expression: 14.6 +/- 7.5, (n=7). When analyzing only the sCD138 positively stained population, the malignant PC showed an even higher level of marker expression: sCXCR7: 78.4 +/- 26.6 (n=8) and sCXCR4: 65.5 +/- 39.1 (n=7). Within our in vitro coculture model (Udi J,..Engelhardt. Br J Haematol 2013; Zlei,...Engelhardt. Exp Hematol 2007; Schüler,..Engelhardt. Expert Opin Biol Ther 2013), CXCL12 stimulation of U266 led to CXCR4 internalization, CXCR4 phosphorylation at Ser339 and actin polymerization. The latter was abrogated by AMD3100 and NOX-A12, proving the functionality of CXCL12-CXCR4 in our MM model. In U266, CXCL12 did not induce CXCR7 internalization. Carfilzomib was rigorously tested in 3 different conditions: it proved cytotoxic for U266 and L363 with pulse (t=1h), continuous treatment (t=3d) and to a lesser extent in coculture (t=3d). Neither AMD3100 nor NOX-A12 significantly reversed the stroma protection. Apart from its cytotoxic effects, carfilzomib (t=3d) reduced sCD138 in U266 or L363 in coculture. As a pulse treatment (t=1h) in U266 monoculture, carfilzomib reduced sCD138 and sCXCR4 without affecting sCXCR7. A 1h pulse of carfilzomib had no effect on pERK/ERK (n=4), nor did it change CXCR4 protein levels (n=2). However, pCXCR4 (Ser339) and its phosphorylating kinase PIM-1 were significantly reduced by carfilzomib in a dose dependent fashion (n=4). Conclusion: CXCR7 is widely expressed on malignant PC and its contribution to the CXCL12-CXCR4 axis in MM is worthy of future study. Carfilzomib shows strong cytotoxic activity in monoculture, which cannot be completely reversed by stroma protection. Carfilzomib appears to downregulate sCXCR4 by reduction of pCXCR4 via decreased PIM-1 protein levels. These data could explain its potency and why the CXCR4/CXCL12-inhibitors AMD3100 and NOX-A12 do not induce additive cytotoxicity. Moreover, and to the best of our knowledge unknown to date, these results suggest a link between CXCR4 and PIM-1 in MM. Therefore, analyzing the (off-) target effects of anti-myeloma substances on the MM BMM interaction seems to be a promising approach to discover optimal drug combinations and new therapeutic targets. 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: 2014
    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. 128, No. 22 ( 2016-12-02), p. 2650-2650
    Abstract: Introduction: The balance between differentiation and self-renewal in hematopoietic stem and progenitor cells (HSPCs) is crucial for homeostasis and lifelong blood cell production. Differentiation is predominantly initiated in the G1 phase of the cell cycle when the E3 ligase anaphase-promoting complex or cyclosome (APC/C) is highly active. Its coactivator Cdh1 determines substrate specificity and mediates proteasomal degradation. Relevant target proteins are associated with cell fate decisions in G1/G0, and there is growing evidence that Cdh1 is an important regulator of differentiation. While this has already been demonstrated in neurons, muscle cells or osteoblasts, little is known about the role of APC/CCdh1 in hematopoiesis. Here we report on the function of Cdh1 in human and murine HSPCs in vitro and in vivo. Methods: Human CD34+ cells from the peripheral blood of G-CSF mobilized donors were exposed to different cytokine combinations and gains or losses of surface marker expression during cell division were determined. By using the established culture conditions Cdh1 expression was detected in distinct hematopoietic lineages and developmental states. CD34+ cells were transduced with a lentivirus to deplete Cdh1 by stably expressing shRNA and was then used for in vitro differentiation in liquid culture or CFU assay. In a second miR-based RNAi approach murine BM cells were depleted of Cdh1 and used for competitive transplantation assays. Complementary xenotransplantation of human Cdh1-depleted CD34+cells was carried out with NSG mice. Results: The stimulation of freshly thawed CD34+ cells with cytokines led to cell cycle entry and proliferation. Self-renewing cells preserved CD34 expression for up to 7 cell divisions with a low proliferation rate. In contrast, during granulopoiesis and erythropoiesis cells divided more frequently with rapid down-regulation of CD34. Cdh1 expression was tightly connected to differentiation status and proliferation properties. In vitro cultured CD34+ cellsand those from BM of healthy human donors showed the highest Cdh1 level compared to moderate or low expression in lymphoid and myeloid cells. Cdh1 is highly expressed at the transcriptional and translational level during both self-renewal and also when cells were directed toward erythroid differentiation. Therefore, high Cdh1 expression is characteristic of immature hematopoietic cells and differentiating precursors. The knockdown of Cdh1 (Cdh1-kd) did not affect proliferation or viability as detected by CFSE staining and measuring the cell cycle length via live-cell imaging. However, Cdh1-kd cells showed a significant maintenance of CD34+ cells under self-renewal conditions and during erythropoiesis with a lower frequency of glycophorin A+ cells. The functional relevance of Cdh1 depletion was verified in CFU assays. Cells with Cdh1-kd formed fewer primary colonies but significantly more secondary colonies, indicating a preference for self-renewal over differentiation. After competitive transplantation Cdh1-depleted murine BM cells showed a significant enhancement in the repopulation of PB, BM and spleen at week 3, while there was no change in cell cycle properties. However, after 8 weeks chimerism in each of the compartments was reduced to that of the control cells. Accordingly, higher LK and LSK frequencies supported the engraftment of Cdh1-depleted cells at week 3, but there was a significant decrease at week 8 compared to control cells, suggestive of stem cell exhaustion. The Cdh1 level also affected cell differentiation in vivo. After 8 weeks the population of B cells (B220+) was increased in transplanted Cdh1-kd cells and the frequency of mature granulocytes (CD11b+ Gr1high) was reduced. Consistently, human Cdh1-depleted CD34+ cells engrafted to a much higher degree in the murine BM 8 and 12 weeks after xenotransplantation, as shown by a higher frequency of human CD45+ cells. Moreover, the increase of human CD19+ B cells with Cdh1-kd confirmed the results of the competitive transplantation. Conclusions: Loss of the APC/C coactivator Cdh1 supports repopulation of murine HSPCs after transplantation with a lymphoid-biased differentiation, and was confirmed in xenotranplantation experiments. In the long-term, Cdh1 loss led to exhaustion of primitive LK and LSK population, highlighting the role of Cdh1 as a critical regulator of HSPC self-renewal and differentiation. Disclosures Engelhardt: Janssen: Research Funding; Amgen: Research Funding; MSD: Research Funding; Celgene: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    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. 116, No. 21 ( 2010-11-19), p. 5020-5020
    Abstract: Abstract 5020 Introduction: As the development of novel anti-MM therapies is pursued worldwide in order to further improve survival in this disease, various innovative agents have to be eagerly tested in in vitro and in vivo models; the former being applied here. Bortezomib (B) has been shown to induce cell death in MMCLs and cytotoxic synergy with sorafenib (S) on various tumor cell lines. S, an oral multikinase inhibitor, targets several cancer-specific pathways and directly affects tumor cell proliferation, cell survival and neovascularization. EGCG (E), one main green tea constituent, causes MM cell toxicity also, but seems to prevent tumor cell death induced by B in vitro and in vivo, this extend not being fully understood as yet. Methods: RPMI8226, U266 and L363 were cultured with RPMI1640/10% FCS. On day (d) 0, cells were treated with increasing concentrations of B, S and/or E. Cell viability and cytotoxicity were assessed on d3 and d6 via trypan blue and PI-staining. CD138 expression and morphologic changes were evaluated via FACS, immunocytochemistry and confocal microscopy. The effect of S on the chemotactic behaviour of L363 in response to conditioned media (CM = supernatant of M210B4 stromal cells) using 96-well chemotaxis chamber plates was also evaluated. Phosphorylation of ERK1/2 was determined by Western blot. The combined effect of S and B was determined using Calcusyn software: the resulting combination index (CI) defines additive effects (CI=1), synergism (CI 〈 1) and antagonism (CI 〉 1). Results: With 10 and 100μM S in L363, we observed increased median PI+ cells (62% and 94% on d3, respectively) as compared to the control (median PI+ d0: 11%), with similar increases on d6 (median 81% and 92%, respectively). In line with PI-observations, viable cells and CD138 expression substantially decreased in a dose- and time-dependent manner. After 3 days pre-incubation with increasing S-concentrations, MM cells were stained with Dapi, Phalloidin-Alexa-549 and CD138-FITC and analyzed by confocal microscopy: L363 cells highly expressed CD138 in the absence of S, whereas impressive CD138 downregulation, morphologic changes and reduction of F-actin content were observed with S-concentrations as low as 1μM. L363 cells exhibited a migratory response to CM, whereas after 3 days of preincubation with 10, 20 and 50μM S, L363 cells showed reduced migratory capacity in response to CM. Western blots showed a decrease in p-ERK1/2 expression levels after 24h inbubation of L363 cells with 10μM S. With 100nM B, PI in L363 increased from 11% on d0 to 84% on d3, albeit not as pronounced with 10nM B as was observed with 10μM S. E induced cytotoxicity in L363, particularly with 50 and 100μM, albeit - different to prior reports - B-induced cell death was preserved when the B-E-combination was tested: of note, however, after addition of increasing E-concentrations, no synergism or additive effect, rather than a plateau cytotoxic effect was observed. Combined B and S use showed synergism with 10nM and 10μM, respectively (CI=0.80). MMCLs stably co-expressing fluorescently labelled cytochrome C and histone H2 will allow the detection of induced apoptosis using live-cell imaging after anti-MM agent treatment. Conclusions: Our MM-based in vitro model revealed that B and S show remarkable therapeutic efficacy as single agents and synergism when combined, which confirms results in other tumor cell lines. E alone induced dose-dependent cell death and decreases in MM cell viability and when combined with B did neither synergize nor abolish B-induced cell death. Our results further enlarge the present knowledge in MM therapy and promise novel insights for innovative substances in the treatment of MM. 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 ...
  • 8
    In: Frontiers in Oncology, Frontiers Media SA, Vol. 11 ( 2021-8-13)
    Abstract: In clinical trials (CTs), the assessment of minimal residual disease (MRD) has proven to have prognostic value for multiple myeloma (MM) patients. Multiparameter flow cytometry (MFC) and next-generation sequencing are currently used in CTs as effective tools for outcome prediction. We have previously described 6- and 8-color MFC panels with and without kappa/lambda, which were equally reliable in detecting aberrant plasma cells (aPC) in myeloma bone marrow (BM) specimens. This follow-up study a) established a highly sensitive single-tube 10-color MFC panel for MRD detection in myeloma samples carrying different disease burden (monoclonal gammopathy of unknown significance (MGUS), smoldering multiple myeloma (SMM), MM), b) evaluated additional, rarely used markers included in this panel, and c) assessed MRD levels and the predictive value in apheresis vs. BM samples of MM patients undergoing autologous stem cell transplantation (ASCT). Methods + Results The 10-color MFC was performed in BM and apheresis samples of 128 MM and pre-MM (MGUS/SMM) patients. The markers CD28, CD200, CD19, and CD117 underwent closer examination. The analysis revealed distinct differences in these antigens between MM, MGUS/SMM, and patients under treatment. In apheresis samples, the 10-color panel determined MRD negativity in 44% of patients. Absence of aPC in apheresis corresponded with disease burden, cytogenetics, and response to induction. It also determined MRD negativity in BM samples after ASCT and was associated with improved progression-free survival. Conclusion These results highlight the significance of the evaluation of both BM and apheresis samples with a novel highly sensitive 10-color MFC panel.
    Type of Medium: Online Resource
    ISSN: 2234-943X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2649216-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Leukemia & Lymphoma, Informa UK Limited, Vol. 51, No. 9 ( 2010-09), p. 1632-1642
    Type of Medium: Online Resource
    ISSN: 1042-8194 , 1029-2403
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2010
    detail.hit.zdb_id: 2030637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2380-2380
    Abstract: Abstract 2380 Introduction: Sorafenib is an oral multikinase inhibitor that targets several cancer-specific pathways and directly affects tumor cell proliferation, cell survival and neovascularization. The Ras/Raf/MEK/ERK pathway is particularly known to be critical for proliferation of multiple myeloma (MM) cells. Moreover, its blockage may not only compromise MM cell survival and proliferation, but also influence cell adhesion and migration. We sought to elucidate the effects of sorafenib on proliferation, phenotype, specific signalling pathways, actin polymerization and chemotaxis, as well as cytotoxic interactions when combined with other anti-MM agents, such as bortezomib. Methods: L363, U266 and RPMI8226 were cultured with RPMI1640, 10% FCS and 0.2% penicillin/streptomycin. On day 0, cells were treated with increasing concentrations of sorafenib and/or bortezomib. Cell viability and cytotoxicity were assessed on days 3 and 6, in addition to day 1 or 2 in previous analyses. The cytotoxic effect for sorafenib and bortezomib combined was evaluated using Calcusyn Software, whereby a combination index =1, 〈 1 or 〉 1 indicated additive, synergistic and antagonistic effects, respectively. CD138 expression and morphologic changes were evaluated via flow cytometry, immunocytochemistry and confocal microscopy. The effect of sorafenib on ERK1/2 phosphorylation was investigated by western blot. Actin polymerization was studied by flow cytometry after labeling with FITC-phalloidin. Chemokine receptor expression was assessed by flow cytometry and chemotaxis of L363 cells with various chemoattractants was studied using 96-well chemotaxis chambers. Results: Our MM-in vitro model confirmed potent cytotoxicity for sorafenib single use and synergistic effects when combined with bortezomib. With 10 and 100μM sorafenib in L363, we observed increased median PI+ cells (62% and 94% on d3, respectively) compared to the control (median PI+ d0: 11%), with similar increases on d6 (median 81% and 92%, respectively). Combined sorafenib and bortezomib use showed additive effects and synergism at 10μM and 10nM bortezomib (combination index: 0.80). Similar to PI-results, viable cells and CD138 expression by flow cytometry substantially decreased with sorafenib in a dose- and time-dependent manner. Regarding the effects on the MAPK pathway, after incubating L363 cells with 1 and 10μM sorafenib for 6 and 24 hours, a dose-dependent downregulation of ERK1/2 phosphorylation was observed. After 3 days of incubation with increasing concentrations of sorafenib, MM cells were stained with DAPI, Phalloidin-Alexa594 and CD138-FITC and analyzed via confocal microscopy. L363 cells highly expressed CD138 in the absence of sorafenib. Of note, sorafenib not only affected cell proliferation, but also phenotype, morphology, actin metabolism and chemotaxis of MM cells. With sorafenib concentrations as low as 1μM, CD138 was downregulated and impressive morphologic changes with a reduction in F-actin content were observed. We could show CXCL12-stimulated actin polymerization and after treatment with sorafenib with concentrations of 10μM and 100μM its inhibition, as confirmed via flow cytometry after labeling with phalloidin-FITC. L363 cells showed high expression of the chemokine receptors CCR4 and CCR5 and underwent chemotaxis to their common ligand CCL5. Chemotaxis of L363 cells was even more evident with the use of supernatant from M210B4 bone marrow stromal cells. This M210B4-induced chemotaxis also occurred in the presence of the specific CXCR4-inhibitor AMD3100, supporting the involvement of chemokines other than CXCL12 in M210B4-induced MM cell migration. M210B4-triggered chemotaxis was substantially inhibited after 3 days of incubation with increasing concentrations of sorafenib in a dose-dependent manner. Conclusions: To the best of our knowledge this is the first analysis of the effects of sorafenib on phenotype, morphology, actin polymerization and migration of MM cells. Sorafenib induced down-regulation of phospho-ERK appeared responsible for the observed actin depolymerization and reduction in M210B4-triggered chemotaxis. Hence, further analysis of sorafenib and other novel anti-MM agents, both in MM cells and their microenvironment, should enable greater progress in this hematopoietic 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: 2011
    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...