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
    Online Resource
    Online Resource
    Wiley ; 2023
    In:  British Journal of Educational Psychology Vol. 93, No. S2 ( 2023-08), p. 339-352
    In: British Journal of Educational Psychology, Wiley, Vol. 93, No. S2 ( 2023-08), p. 339-352
    Abstract: What is redundancy? While most studies confirm that redundancy is harmful to learning, there are two theoretical approaches to redundancy. The first understands redundancy as a contentual overlap that puts demand on the limited cognitive capacities of the learner. The second understands redundancy as an ineffective combination of sources leading to an overload of the limited working memory modalities. Aims Since these theoretical differences are rarely acknowledged in operation, this study proposes a classification of two distinct types of redundancy to compare these experimentally to investigate their possible main and interaction effects. The first type, content redundancy, is concerned with the contentual overlap of information. The second type, modal redundancy, is concerned with the modalities in which the information is displayed. Methods We used these two types of redundancy as factors in a 2 × 2 within‐subject design, in which we experimentally compared their effects. Sample University students ( N  = 46) learned from specifically designed domain‐general material which aimed to observe redundancy effects without interference from confounding variables. Results The results show that content redundancy increases learning outcomes and decreases cognitive load, while modal redundancy decreases learning outcomes and increases cognitive load. Conclusion On the theoretical level, these findings confirm the usefulness to distinguish content redundancy from modal redundancy. On the practical level, the empirical findings of the different effects of the two types of redundancy provide educators with important insights that can improve the design of multimedia learning materials.
    Type of Medium: Online Resource
    ISSN: 0007-0998 , 2044-8279
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 280391-4
    detail.hit.zdb_id: 1501130-6
    SSG: 5,2
    SSG: 5,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Medicinal Chemistry, American Chemical Society (ACS), Vol. 66, No. 13 ( 2023-07-13), p. 8666-8686
    Type of Medium: Online Resource
    ISSN: 0022-2623 , 1520-4804
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2023
    detail.hit.zdb_id: 1491411-6
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Die Urologie, Springer Science and Business Media LLC
    Type of Medium: Online Resource
    ISSN: 2731-7064 , 2731-7072
    Language: German
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 3123197-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 29, No. 12 ( 2023-06-13), p. 2266-2279
    Abstract: Interactions with tumor-associated microglia and macrophages (TAM) are critical for glioblastoma progression. Polysialic acid (polySia) is a tumor-associated glycan, but its frequency of occurrence and its prognostic value in glioblastoma are disputed. Through interactions with the opposing immune receptors Siglec-11 and Siglec-16, polySia is implicated in the regulation of microglia and macrophage activity. However, due to a nonfunctional SIGLEC16P allele, SIGLEC16 penetrance is less than 40%. Here, we explored possible consequences of SIGLEC16 status and tumor cell–associated polySia on glioblastoma outcome. Experimental Design: Formalin-fixed paraffin-embedded specimens of two independent cohorts with 70 and 100 patients with newly diagnosed glioblastoma were retrospectively analyzed for SIGLEC16 and polySia status in relation to overall survival. Inflammatory TAM activation was assessed in tumors, in heterotypic tumor spheroids consisting of polySia-positive glioblastoma cells and Siglec-16–positive or Siglec-16–negative macrophages, and by exposing Siglec-16–positive or Siglec-16–negative macrophages to glioblastoma cell–derived membrane fractions. Results: Overall survival of SIGLEC16 carriers with polySia-positive tumors was increased. Consistent with proinflammatory Siglec-16 signaling, levels of TAM positive for the M2 marker CD163 were reduced, whereas the M1 marker CD74 and TNF expression were increased, and CD8+ T cells enhanced in SIGLEC16/polySia double-positive tumors. Correspondingly, TNF production was elevated in heterotypic spheroid cultures with Siglec-16–expressing macrophages. Furthermore, a higher, mainly M1-like cytokine release and activating immune signaling was observed in SIGLEC16-positive as compared with SIGLEC16-negative macrophages confronted with glioblastoma cell–derived membranes. Conclusions: Collectively, these results strongly suggest that proinflammatory TAM activation causes the better outcome in patients with glioblastoma with a functional polySia-Siglec-16 axis.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: European Journal of Surgical Oncology, Elsevier BV, Vol. 49, No. 3 ( 2023-03), p. 611-618
    Type of Medium: Online Resource
    ISSN: 0748-7983
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2002481-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancers, MDPI AG, Vol. 12, No. 6 ( 2020-06-07), p. 1488-
    Abstract: Background: Aminopeptidase N (CD13) is present on tumor vasculature cells and some tumor cells. Truncated tissue factor (tTF) with a C-terminal NGR-peptide (tTF-NGR) binds to CD13 and causes tumor vascular thrombosis with infarction. Methods: We treated 17 patients with advanced cancer beyond standard therapies in a phase I study with tTF-NGR (1-h infusion, central venous access, 5 consecutive days, and rest periods of 2 weeks). The study allowed intraindividual dose escalations between cycles and established Maximum Tolerated Dose (MTD) and Dose-Limiting Toxicity (DLT) by verification cohorts. Results: MTD was 3 mg/m2 tTF-NGR/day × 5, q day 22. DLT was an isolated and reversible elevation of high sensitivity (hs) Troponin T hs without clinical sequelae. Three thromboembolic events (grade 2), tTF-NGR-related besides other relevant risk factors, were reversible upon anticoagulation. Imaging by contrast-enhanced ultrasound (CEUS) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) showed major tumor-specific reduction of blood flow in all measurable lesions as proof of principle for the mode of action of tTF-NGR. There were no responses as defined by Response Evaluation Criteria in Solid Tumors (RECIST), although some lesions showed intratumoral hemorrhage and necrosis after tTF-NGR application. Pharmacokinetic analysis showed a t1/2(terminal) of 8 to 9 h without accumulation in daily administrations. Conclusion: tTF-NGR is safely applicable with this regimen. Imaging showed selective reduction of tumor blood flow and intratumoral hemorrhage and necrosis.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Catheterization and Cardiovascular Interventions, Wiley, Vol. 97, No. 3 ( 2021-02-15), p. 431-436
    Abstract: Quantitative flow ratio (QFR) has been validated as an accurate surrogate of standard wire‐based fractional flow reserve. The clinical and angiographic outcomes of the Fantom sirolimus‐eluting bioresorbable coronary scaffold (BRS) have been previously studied and reported. We investigate the functional performance of the Fantom BRS. Methods The FANTOM II trial prospectively enrolled 240 patients with stable coronary artery disease or unstable angina, of which 235 patients received the Fantom BRS and were included in the present analysis. We performed an independent serial QFR analysis of the target vessel at baseline, post‐percutaneous coronary intervention (PCI), and at 6‐ or 9‐month and 24‐month follow‐up, using a QFR threshold ≤0.80 to define functional ischemia. Results QFR was analyzable in 178 patients at baseline, 185 post‐PCI, 178 at 6‐ or 9‐month follow‐up, and 30 at 24‐month follow‐up. At baseline, 119 patients (66.9%) had a QFR ≤0.80, whereas 12 (6.5%) post‐PCI, 13 (7.3%) at 6‐ or 9‐month follow‐up, and 3 (10.0%) at 24‐month follow‐up had a QFR ≤0.80. QFR improved from baseline to post‐PCI, and decreased from post‐PCI up to 24‐month follow‐up. During follow‐up period, 28 patients (11.9%) had target vessel revascularization, of which 21 had analyzable QFR and 16 patients (76.1%) had QFR ≤0.80 at the time of revascularization. Conclusions Off‐line serial QFR assessment demonstrated that around 30% patients did not have functionally significant lesions at baseline and the time with target vessel revascularization. PCI with the Fantom BRS improved functional ischemia with a slight decrease in QFR values over 24 months.
    Type of Medium: Online Resource
    ISSN: 1522-1946 , 1522-726X
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2021
    detail.hit.zdb_id: 2001555-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 1146-1146
    Abstract: Background: Tepotinib is approved to treat patients with NSCLC harboring MET exon 14 skipping. We previously provided preclinical proof of concept for tepotinib to treat NSCLC metastasized to the brain (Friese-Hamim M, et al. Lung Cancer 2022;163:77-86). Here, we report a detailed analysis of the effects of high-dose tepotinib in preclinical models of MET-amplified NSCLC brain metastases. Experimental methods: Using the previously reported intracranial MET-amplified patient-derived xenograft (PDX) models (LU5349 and LU5406), we measured the effects of tepotinib at the clinically relevant dose of 125 mg/kg QD and above (300 mg/kg QD) or a vehicle control on plasma concentrations, tumor growth (by T2-weighted MRI), intra-tumoral compound distribution (by MALDI MSI) and tumor vasculature (by DCE-MRI for Ktrans, a measure of tumor permeability), as well as biomarkers in tumor tissue (pMET, pAKT, Ki67, endoglin, caspase-3, and MoIgG). For model LU5349, animals were imaged on the day before euthanasia at Day (D) 0, 6 and 16 of treatment (n=10 per time point). Results: Plasma concentrations of tepotinib 2 hours after the final administration at each time point were not proportional by dose, indicating a limited absorption at the higher dose. By D16, tumor measurements (±SD) in tepotinib-treated mice (5.5 ± 3.4 mm3) were approximately 20-fold smaller than those in control mice (109.3 ± 50.1 mm3). At the lower dosing level, concentrations of tepotinib within the tumor regions decreased from 12 ± 6.4 µM (D0) to 3.2 ± 2.7 µM (D6) and then increased to 4.6 ± 1.2 µM (D16). In the higher dose group, concentrations of tepotinib in tumor regions decreased from 15.1 ± 8.7 µM (D0) to 6.8 ± 2.9 µM (D6) and then to 4.5 ± 3.6 µM (D16). Concentrations of tepotinib were below the limit of detection in non-tumor regions. Ktrans of the tumor was also affected by tepotinib treatment. While there was an apparent decline in mean Ktrans over time in control tumors (7.4 ± 3.0 × 10−2 s−1 [D0], 6.4 ± 2.8 × 10−2 s−1 [D6] , 5.6 ± 3.5 × 10−2 s−1 [D16]), tepotinib treatment caused a further reduction at both the lower (7.8 ± 3.2 × 10−2 s−1 [D0] , 2.9 ± 1.3 × 10−2 s−1 [D6], 2.7 ± 1.4 × 10−2 s−1 [D16] ) and higher dose (8.0 ± 3.0 × 10−2 s−1 [D0], 2.7 ± 1.1 × 10−2 s−1 [D6] , 2.9 ± 1.2 × 10−2 s−1 [D16]), indicating potential anti-vascular effects in the tumor. Conclusions: These preclinical analyses of tepotinib treatment of MET-amplified NSCLC brain metastases provide new insights, considering the observed tumor regression, the compound’s distribution to tumor versus non-tumor brain tissue and anti-vascular effects (via DCE-MRI), which may provide a predictive biomarker of response and patient survival. A possible causal association between reduced vascular permeability and the restricted distribution of tepotinib to tumor regions may exist. Analyses of the second PDX model and tumor biomarkers are ongoing and will be presented. Citation Format: Anderson Clark, Joachim Albers, Patrick McConville, Sylwia A. Stopka, Michael S. Regan, Marina DiPiazza, Nathalie Y.R. Agar. Tepotinib treatment inhibits tumor growth and affects tumor permeability in an intracranial PDX model of MET-amplified NSCLC brain metastasis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Inv ited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1146.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), ( 2023-09-29)
    Abstract: Purpose: Leiomyosarcoma (LMS) is an aggressive sarcoma for which standard chemotherapies achieve response rates under 30%. There are no effective targeted therapies against LMS. Most LMS are characterized by chromosomal instability (CIN), resulting in part from TP53 and RB1 co-inactivation and DNA damage repair defects. We sought to identify therapeutic targets that could exacerbate intrinsic CIN and DNA damage in LMS, inducing lethal genotoxicity. Experimental design: We performed clinical targeted sequencing in 287 LMS and genome-wide loss-of-function screens in 3 patient-derived LMS cell lines, to identify LMS-specific dependencies. We validated candidate targets by biochemical and cell-response assays in vitro and in 7 mouse models. Results: Clinical targeted sequencing revealed a high burden of somatic copy number alterations (median fraction of the genome altered=0.62) and demonstrated homologous recombination deficiency signatures in 35% of LMS. Genome-wide shRNA screens demonstrated PRKDC (DNA-PKcs) and RPA2 essentiality, consistent with compensatory non-homologous end joining hyper-dependence. DNA-PK inhibitor combinations with unconventionally low-dose doxorubicin had synergistic activity in LMS in vitro models. Combination therapy with peposertib and low-dose doxorubicin (standard or liposomal formulations) inhibited growth of 5 of 7 LMS mouse models without toxicity. Conclusion: Combinations of DNA-PK inhibitors with unconventionally low, sensitizing, doxorubicin dosing showed synergistic effects in LMS in vitro and in vivo models, without discernable toxicity. These findings underscore the relevance of DNA damage repair alterations in LMS pathogenesis and identify dependence on NHEJ as a clinically actionable vulnerability in LMS.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6215-6215
    Abstract: Background: Triple-negative breast cancer (TNBC) is the most lethal breast cancer subtype, exhibiting poor response rates toward current chemotherapy regimens and lacking additional effective treatment options. Approximately 30% of patients with TNBC respond well to anthracycline/taxane-based standard-of-care chemotherapy regimens. However, remaining patients demonstrate limited improvements in clinical outcomes, highlighting the critical need for effective strategies to improve anthracycline/taxane-based chemotherapy (1, 2). DNA-dependent protein kinase (DNA-PK), a member of the phosphoinositide 3-kinase-related kinase protein family, promotes nonhomologous end joining (NHEJ) as part of the DNA damage response. Peposertib is a potent, selective, and orally bioavailable DNA-PK inhibitor that strongly potentiates the antitumor effects of ionizing radiation and DNA double-strand break-inducing agents, including anthracyclines, in preclinical models. Here we report the synergistic antitumor effects of peposertib combined with topoisomerase II (TOPO II) inhibitors, particularly anthracyclines, in TNBC models in vitro and in vivo. Methodology: Combinations of peposertib with TOPO II inhibitors were evaluated in TNBC cell lines by viability assays, immunoblotting, and flow cytometry. Gene expression analysis was performed using the nCounter PanCancer pathways panel and results were confirmed by quantitative polymerase chain reaction (qPCR). In vivo efficacy was assessed in cell-line derived and patient-derived xenograft models. Results: When administered in combination with doxorubicin, epirubicin, and etoposide, peposertib exhibited synergistic antiproliferative activity in TNBC cell lines in vitro. The downstream analysis of pharmacodynamic biomarkers revealed induction of NHEJ mediated repair upon TOPO II inhibitor treatment and provided mechanistic insights into the synergistic antitumor effects of peposertib combined with TOPO II inhibitors. Furthermore, this combination treatment induced ataxia telangiectasia mutated (ATM)-dependent compensatory signaling and inflammatory responses, potentially creating a proinflammatory tumor microenvironment. To evaluate whether peposertib enhanced the antitumor effects of TOPO II inhibitors in vivo, we established a well-tolerated preclinical treatment schedule for the combined use of peposertib and pegylated liposomal doxorubicin, which was capable of achieving tumor regression in patient- and cell line-derived xenograft models of TNBC. Conclusion: Our findings suggest that cotreatment with the DNA-PK inhibitor peposertib can enhance the efficacy of anthracycline/TOPO II-based chemotherapies. This work was supported by the healthcare business of Merck KGaA, Darmstadt, Germany (CrossRef Funder ID: 10.13039/100009945). 1) Davison C et al. NPJ Breast Cancer. 2021;7(1):38. 2) Bianchini G et al. Nat Rev Clin Oncol. 2016;13(11):674-690. Citation Format: Steffie Revia, Christian Sirrenberg, Antonia Schach, Astrid Zimmermann, Frank T. Zenke, Joachim Albers. Peposertib, a DNA-PK inhibitor, enhances the antitumor efficacy of anthracyclines in triple-negative breast cancer models in vitro and in vivo. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6215.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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...