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: Journal of Instrumentation, IOP Publishing, Vol. 17, No. 01 ( 2022-01-01), p. P01013-
    Abstract: The semiconductor tracker (SCT) is one of the tracking systems for charged particles in the ATLAS detector. It consists of 4088 silicon strip sensor modules. During Run 2 (2015–2018) the Large Hadron Collider delivered an integrated luminosity of 156 fb -1 to the ATLAS experiment at a centre-of-mass proton-proton collision energy of 13 TeV. The instantaneous luminosity and pile-up conditions were far in excess of those assumed in the original design of the SCT detector. Due to improvements to the data acquisition system, the SCT operated stably throughout Run 2. It was available for 99.9% of the integrated luminosity and achieved a data-quality efficiency of 99.85%. Detailed studies have been made of the leakage current in SCT modules and the evolution of the full depletion voltage, which are used to study the impact of radiation damage to the modules.
    Type of Medium: Online Resource
    ISSN: 1748-0221
    Language: Unknown
    Publisher: IOP Publishing
    Publication Date: 2022
    detail.hit.zdb_id: 2235672-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: The Lancet, Elsevier BV, Vol. 396, No. 10243 ( 2020-07), p. 27-38
    Type of Medium: Online Resource
    ISSN: 0140-6736
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2020
    detail.hit.zdb_id: 2067452-1
    detail.hit.zdb_id: 3306-6
    detail.hit.zdb_id: 1476593-7
    SSG: 5,21
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 730-730
    Abstract: Abstract 730 Multi-targeted kinase inhibitors, when associated with manageable toxicity, offer the therapeutically desirable option of targeting, through a single chemical entity, several pathways that may contribute to the complexity and heterogeneity of molecular lesions harbored by neoplasias such as multiple myeloma (MM). However, intractable questions often emerge while prioritizing for preclinical studies different multi-targeted agents with extensive and/or only partially overlapping of sets of known targets. We have hypothesized that the potential therapeutic relevance of a multi-targeted inhibitor may be reflected on the prognostic relevance of its targets' transcriptional signature. We applied this concept in the case of the orally bioavailable multi-targeted kinase inhibitor AS703569, which targets (with IC50 in low nM range) all 3 Aurora kinase (AK) isoforms as well as various other kinases (e.g. cSRC, FGFR1, Flt3, Fyn, Lyn, Rsk1-3, Yes, Axl, et.c.) and evaluated the transcriptional signature of AS703569 kinase targets (with IC50 〈 10 nM) in MM cells of patients receiving Bortezomib as part of Phase II/III trials (specifically SUMMIT/APEX). We observed that patients with high transcriptional signature of AS703569 targets had inferior progression-free and overall survival (p=0.005 and p=0.012, log-rank test) and also validated that, in a study of tandem autologous transplant, a subset of patients with high levels of this AS703569 target transcriptional signature also have inferior overall survival (p=0.032, log-rank test) compared to cases with low levels of the signature. These observations supported the notion that the kinome space targeted by AS703569 is enriched for targets associated with adverse clinical outcome in MM. In preclinical assays, we observed that AS703569 decreased the viability of MM cell lines and primary CD138+ MM tumor cells in a time- and dose-dependent manner, with IC50 values 〈 50 nM for the majority of cell lines tested; and without evidence of cross-resistance with established anti-MM agents. Combinations of AS703569 with dexamethasone, doxorubicin, or bortezomib did not exhibit antagonism, suggesting that AS703569 can be incorporated in regimens with these established anti-MM drug classes. Interestingly, in vitro compartment-specific bioluminescence imaging (CS-BLI) assays showed that against MM cells which respond to stromal cells with increased proliferation and survival, the anti-MM activity of AS703569 is more pronounced when these MM cells are co-cultured with bone marrow stromal cells than in conventional cultures in isolation. This indicated that AS703569 is capable of overcoming the protective effects that BMSCs confer to MM tumor cells and prompted in vivo validation studies in our orthotopic SCID/NOD model of diffuse MM bone lesions established by i.v. injection of MM-1S-GFP/Luc cells monitored by whole body bioluminescence imaging. AS703569 (50 mg/kg p.o. once weekly)-treated mice had longer overall survival than vehicle-treated mice (median 50.0 days, 95% C.I. 40.3-59.7 days vs. 39.0 days, 95% C.I., 35.4-42.6 days, p=0.019, log-rank test). An alternative schedule of AS703569 at 16.7 mg/kg 3 times/week also resulted in longer overall survival (median 54.0 days, 95% C.I. 33.2-74.8 days, p=0.023, log-rank test). These data indicate that AS703569 exhibits anti-MM activity in vitro and in orthotopic in vivo MM models, and suggests that this multi-targeted inhibitor merits considerations for further preclinical studies, as well as potential clinical studies in MM, especially given the otherwise adverse outcome associated with the inhibitor's target transcriptional signature. Disclosures: Laubach: Novartis: Consultancy, Honoraria. Rastelli:EMD Serono: Employment. Clark:EMD Serono: Employment. Sarno:EMD Serono: Employment. Richardson:Millenium: (Speakers' Bureau up to 7/1/09), Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: (Speakers' Bureau up to 7/1/09), Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Anderson:Millennium: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Mitsiades:Millennium: Consultancy, Honoraria; Novartis : Consultancy, Honoraria; Bristol-Myers Squibb : Consultancy, Honoraria; Merck & Co: Consultancy, Honoraria; Kosan Pharmaceuticals: Consultancy, Honoraria; Pharmion: Consultancy, Honoraria; PharmaMar: Patents & Royalties; Amgen: Research Funding; AVEO Pharma: Research Funding; EMD Serono : Research Funding; Sunesis Pharmaceuticals: Research Funding.
    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 ...
  • 4
    In: Nature Chemical Biology, Springer Science and Business Media LLC, Vol. 9, No. 12 ( 2013-12), p. 840-848
    Type of Medium: Online Resource
    ISSN: 1552-4450 , 1552-4469
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2013
    detail.hit.zdb_id: 2190276-8
    SSG: 12
    SSG: 15,3
    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. 276-276
    Abstract: Most conventional methods to sensitively quantify tumor cell proliferation and viability in vitro involve processing of cells in ways that preclude continuation of the respective experiment or prevent the longitudinal collection of data. This common technical feature of conventional assays limits their ability to provide detailed insight into the kinetics of tumor cell responses to treatment(s). Additionally, these limitations hinder the use of these assays to monitor how the kinetics of treatment response can be altered by nonmalignant "accessory" cells of the tumor microenvironment (e.g. bone marrow stromal cells [BMSCs] for hematologic malignancies or bone metastases of solid tumors). To address these obstacles, we modified our previously developed tumor cell compartment-specific bioluminescence imaging (CS-BLI) platform (McMillin et al. Nat Med. 2010), to enable longitudinal assessment of tumor cell response to diverse experimental conditions; we cultured luciferase-expressing tumor cells, with or without stromal cells, in the presence of bioluminescent substrates, using optimized conditions which provide detectable bioluminescent signal even after several days of culture, while having no adverse effect on the viability of tumor or non-malignant cells in this system. This modified approach (time-lapse CSBLI, [TL-CSBLI] ) preserved the linear correlation of bioluminescent signal with tumor cell viability. Furthermore, results obtained at the end of the experiment and during interim time-points are consistent with those generated using either non-time-lapse applications of CS-BLI or conventional techniques. We applied TL-CSBLI to delineate, in high-throughput manner, the temporal dynamics of the responses of tumor cells (e.g. multiple myeloma (MM) and other hematologic malignancies) to diverse treatments (e.g. conventional chemotherapeutics, glucocorticoids; proteasome inhibitors (PIs, bortezomib or carfilzomib), and kinase inhibitors). Using the time-lapse capabilities of this assay, we evaluated tumor cell responses in the presence vs. absence of stromal cells. We observed that the kinetics of tumor cell response to diverse therapeutic classes are heterogeneous, even within the same tumor type: for instance, tumor cells with pronounced responses at the end of drug incubation (e.g. 24, 48, 72, hrs after initiation of treatment with PIs, DNA-damaging chemotherapeutics, or dexamethasone respectively), can have different magnitude of responses at intermediate time points. This suggests that TL-CSBLI data can further stratify treatment-responsive tumor cells into those with early vs. late kinetics of response. We also observed that the kinetics of the proliferative / anti-apoptotic effect conferred by stromal cells on tumor cells are highly variable between different cell lines, even within the same tumor type. For instance, the time between initiation of coculture and maximum stimulation of tumor cell viability by stromal cells was variable between cell lines and did not correlate with the magnitude of stimulation by stromal cells. Importantly, TL-CSBLI identified that the response of diverse types of tumor cells to treatments can be delayed in the presence of stromal cells, compared to conventional tumor cell monocultures: this initial delay in treatment response of tumor cells in stromal co-cultures may be observed even in cases where similar cytoreductive responses are eventually observed at later time-points in both the presence and absence of stromal cells. This observation suggests that a more expansive definition of stroma-induced resistance to a given treatment may be warranted, to specifically incorporate the ability of stromal cells to delay the tumor cell response to such treatment. In summary, TL-CSBLI enables detailed characterization of the kinetics of tumor cell responses to diverse experimental conditions. Its use can provide insight into the underappreciated impact that cell-autonomous variations or stroma-induced changes in the kinetics of tumor cell response to a given anti-tumor therapy can have on determining its efficacy. This is particularly consequential for agents (e.g. PIs) which have clinical pharmacokinetic profiles associated with transient peak exposure. Disclosures McMillin: Axios Biosciences: Equity Ownership; DFCI: patent submission on stromal co-culture technologies Patents & Royalties. Negri:DFCI: patent submission on stromal co-culture technologies Patents & Royalties. Mitsiades:Johnson & Johnson: Research Funding; Amgen: Research Funding; Celgene: Consultancy, Honoraria; Millennium Pharmaceuticals: Consultancy, Honoraria; DFCI: patent submission on stromal co-culture technologies Patents & Royalties.
    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: Stem Cell Reports, Elsevier BV, Vol. 9, No. 6 ( 2017-12), p. 1868-1884
    Type of Medium: Online Resource
    ISSN: 2213-6711
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 2720528-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 8, No. 12_Supplement ( 2009-12-10), p. A237-A237
    Abstract: Multi-targeted kinase inhibitors, when associated with manageable toxicity, offer the therapeutically desirable option of targeting, through a single chemical entity, several pathways contributing to the molecular complexity and heterogeneity of neoplasias, such as multiple myeloma (MM). However, when multi-targeted agents have extensive and/or only partially overlapping known targets, it is difficult to prioritize them for further development. We hypothesized that the potential therapeutic relevance of a multi-targeted inhibitor may be reflected on the prognostic relevance of its targets' transcriptional signature. We applied this concept in the case of the orally bioavailable multi-targeted kinase inhibitor AS703569, which targets (IC50 in low nM range) all 3 Aurora kinase isoforms and diverse other kinases (e.g. cSRC, FGFR1, Flt3, Fyn, Lyn, Rsk1-3, Axl, et.c.) and evaluated the transcriptional signature of AS703569 kinase targets (with IC50 & lt;10 nM) in MM cells of patients receiving Bortezomib as part of Phase II/III trials. We observed that patients with high transcriptional signature of AS703569 targets had inferior progression-free and overall survival (p=0.005 and p=0.012, log-rank test) and also validated that, in a study of tandem autologous transplant, a subset of patients with high levels of this AS703569 target transcriptional signature also have inferior overall survival (p=0.032, log-rank test) compared to cases with low levels of the signature. These observations indicate that the kinome space targeted by AS703569 is enriched for targets associated with adverse clinical outcome in MM. In preclinical assays, we observed that AS703569 decreased the viability of MM cell lines and primary CD138+ MM tumor cells in time- and dose-dependent manner (IC50 values & lt;50 nM for the majority of cell lines tested), without evidence of cross-resistance with established anti-MM agents. Combinations of AS703569 with dexamethasone, doxorubicin, or bortezomib did not exhibit antagonism, suggesting that AS703569 can be incorporated in regimens with these established anti-MM drug classes. Interestingly, in vitro compartment-specific bioluminescence imaging (CS-BLI) assays showed that against MM cells which respond to stromal cells with increased proliferation and survival, the anti-MM activity of AS703569 is more pronounced when these MM cells are co-cultured with bone marrow stromal cells than in conventional cultures in isolation. This in vitro result that AS703569 can overcome protective effects of BSMCs on MM tumor cells was validated in vivo in orthotopic SCID/NOD model of diffuse MM bone lesions established by i.v. injection of MM-1S-GFP/Luc cells monitored by whole body bioluminescence imaging. AS703569 (50 mg/kg p.o. once weekly)-treated mice had longer overall survival than vehicle-treated mice (median 50.0 vs. 39.0 days, p=0.019, log-rank test). An alternative schedule of AS703569 at 16.7 mg/kg 3 times/week also resulted in longer overall survival (median 54.0 days, p=0.023). These data suggest that AS703569 merits further studies towards potential clinical trials in MM, especially given the adverse outcome associated with the molecular signature of its targets. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A237.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 119, No. 15 ( 2012-04-12), p. e131-e138
    Abstract: Conventional assays evaluating antitumor activity of immune effector cells have limitations that preclude their high-throughput application. We adapted the recently developed Compartment-Specific Bioluminescence Imaging (CS-BLI) technique to perform high-throughput quantification of innate antitumor activity and to show how pharmacologic agents (eg, lenalidomide, pomalidomide, bortezomib, and dexamethasone) and autologous BM stromal cells modulate that activity. CS-BLI–based screening allowed us to identify agents that enhance or inhibit innate antitumor cytotoxicity. Specifically, we identified compounds that stimulate immune effector cells against some tumor targets but suppressed their activity against other tumor cells. CS-BLI offers rapid, simplified, and specific evaluation of multiple conditions, including drug treatments and/or cocultures with stromal cells and highlights that immunomodulatory pharmacologic responses can be heterogeneous across different types of tumor cells. This study provides a framework to identify novel immunomodulatory agents and to prioritize compounds for clinical development on the basis of their effect on antitumor immunity.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2652-2652
    Abstract: Obesity and insulin resistance are associated with increased risk for several malignancies, including multiple myeloma (MM), possibly due to hyperinsulinemia and the resulting increased circulating free IGF-I levels that can have a growth-promoting and anti-apoptotic effect on MM cells. It has been proposed that improving insulin sensitivity, with a low-fat diet or with the use of the biguanide metformin, could suppress circulating insulin and free IGF-I levels and lower cancer risk. Metformin has been widely used as an antidiabetic agent since the 1950s and has an excellent safety profile. Interestingly, epidemiologic studies have suggested that patients with type 2 diabetes mellitus who are treated with metformin have lower cancer incidence and lower cancer-related mortality than diabetics who receive sulfonylurea monotherapy or insulin. Even more intriguing is the recent finding that metformin can have direct anticancer activity in vitro against prostate and breast carcinoma cell lines. In breast carcinoma cells, metformin inhibits mammalian target of rapamycin (mTOR) activity and suppresses translation initiation and global protein synthesis, via an AMP-activated protein kinase (AMPK)-dependent pathway. We investigated the direct effects of metformin in vitro in a panel of 17 MM cell lines and sublines using MTT, flow cytometry, annexin V-PI, as well as compartment-specific bioluminescence (CS-BLI) assays in the absence or presence of bone marrow stromal cells (BMSCs). We found that metformin suppressed the growth of all MM cell lines and sublines, including those that are resistant to conventional and several investigational anti-MM agents. The anti-MM activity of metformin was achieved with clinically relevant sub-mM or low mM levels, and was in the same order of magnitude or stronger than its activity reported in solid tumor cell line models. The anti-MM activity of metformin did not involve apoptotic/necrotic cell death, but inhibition of proliferation (G0/G1 arrest). Western blot analyses showed that metformin treatment of RPMI-8226/S MM cells led to increased phosphorylation of AMPK; suppression of p70S6K phosphorylation; and decreased cyclin D1 levels. These results are consistent with the known effect of metformin in triggering AMPK activity and thereby suppressing the function of its downstream targets mTOR and p70S6K. Co-culture with BMSCs did not abrogate the anti-MM effect of metformin, but further sensitized MM cell to this agent, at concentrations which did not affect BMSC viability. This sensitization was more prominent in MM cell lines that respond to BMSCs with increased proliferation/survival (e.g. MM-1S, MM-1R, KMS-11, L363) than in cell lines independent/unresponsive to stromal stimulation (e.g. OPM-2). To further probe the mechanistic basis for this stroma-induced sensitization to metformin, we studied the transcriptional signature of genes suppressed by mTOR inhibition, a downstream effect of metformin, in MM cells cultured alone vs. co-cultured with BMSCs. We observed that stroma triggers a significant increase in the average expression of mTOR-responsive genes in stroma responsive MM cell lines, but not in stroma unresponsive lines, in which this transcriptional signature is constitutively upregulated. This observation suggests that the increased metformin sensitivity of MM cells co-cultured with BMSCs may reflect the ability of metformin to counteract the upregulation of p70S6K/mTOR pathway activity triggered by stroma. Our studies demonstrate a direct, growth-suppressive effect of metformin in MM cells in vitro, which can be potentiated by interactions with the bone marrow microenvironment. Taken together with the beneficial effect of this antidiabetic agent on circulating insulin and free IGF-I levels and its favorable safety profile, these data support the clinical evaluation of metformin in patients with MM.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 219-219
    Abstract: Introduction: Osteoclasts (OCLs) have a central role in the pathophysiology of multiple myeloma (MM), because their excessive and unopposed activation leads to the characteristic bone lesions of this disease. However, unlike the well documented role of bone marrow stromal cells (BMSCs) in conferring resistance to diverse conventional anti- MM agents, the impact of OCLs in drug responsiveness of MM cells has not been studied extensively. This is due in part to limitations of conventional assays which are either not amenable to co-culture studies or not conducive to comprehensive high-throughput studies. Methods/Results: To address this void in the MM field, we applied compartment specific bioluminescence imaging (CS-BLI) in vitro assay in the context of MM-osteoclast co-cultures. Specifically, in these co-cultures the MM cell compartment is engineered to stably express luciferase (Luc) and is co-cultured with Luc-negative primary mature osteoclasts generated after ex vivo differentiation of peripheral blood monocytes with MCSF and RANKL. In these co-cultures, the activity of luciferase in response to addition of substrate results in bioluminescence signal which is directly proportional to number of viable Luc+ cells only, thus allowing for selective and sensitive quantification of the viable MM tumor cell compartment. By applying the CS-BLI platform in the context of MM-OCL co-cultures and comparing them with results of MM-BMSC co-cultures, we observed that mature OCLs stimulate increase in the number of viable MM cells from stroma-responsive, but not in stroma-unresponsive, cell lines. Furthermore, stroma/OCL-responsive MM cell lines that are sensitive to glucocorticoids and anthracyclines (e.g. MM-1S) exhibit significant decrease in their response to these conventional anti-MM agents when co-cultured with OCLs, similar to the resistance conferred to these agents by BMSCs. In addition, novel anti-MM agents, such as the proteasome inhibitor bortezomib and hsp90 inhibitors, were equally active against MM cells in the presence vs. absence of OCLs at concentrations and durations of drug treatment that did not significantly affect the viability of OCLs, consistent with results obtained with similar co-cultures of MM cells with BMSCs. These results confirm that BMSCs are not the only cellular compartment of the BM microenvironment that can function to support MM cell proliferation, survival and drug resistance, and that OCLs can also function in that accessory role as well. Conclusions: CS-BLI addresses key limitations that have precluded the highthroughput testing of novel agents in tumor-OCL co-cultures. Its comparative application in co-cultures of MM cells with OCL vs. BMSCs provides an opportunity to compare qualitatively and quantitatively the similarities or differences in patterns of drug resistance conferred by these accessory cell compartments. It also offers a powerful tool to identify new and, hopefully, more effective classes of drugs which are active in MM despite the effects of the BM microenvironment.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...