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: Anti-Cancer Drugs, Ovid Technologies (Wolters Kluwer Health), Vol. 34, No. 4 ( 2023-04), p. 532-543
    Abstract: Derazantinib (DZB) is an inhibitor of fibroblast growth factor receptors 1–3 (FGFR1–3), with additional activity against colony-stimulating-factor-1 receptor (CSF1R). We have profiled the activity of DZB in gastric cancer (GC) as monotherapy and combined with paclitaxel, and explored means of stratifying patients for treatment. The antiproliferative potency of DZB in vitro was quantified in 90 tumor cell lines and shown to correlate significantly with FGFR expression ( 〈 0.01) but not with FGFR DNA copy-number (CN) or FGFR mutations. In four GC cell lines in vitro , little or no synergy was observed with paclitaxel. In athymic nude mice, bearing cell-line derived xenografts (CDX) or patient-derived xenograft (PDX) GC models, DZB efficacy correlated highly significantly with FGFR gene expression ( r 2 = 0.58; P = 0.0003; n = 18), but not FGFR mutations or DNA-CN. In FGFR-driven GC models, DZB had comparable efficacy to three other FGFR inhibitors and was more efficacious than paclitaxel. DZB had dose-dependent plasma pharmacokinetics but showed low brain penetration at all doses. GC models (one CDX and six PDX) were tested for sensitivity to the combination of DZB and paclitaxel and characterized by immunohistochemistry. The combination showed synergy (5) or additivity (2), and no antagonism, with synergy significantly associated ( P 〈 0.05) with higher levels of M2-type macrophages. The association of strong efficacy of the combination in vivo with M2 macrophages, which are known to express CSF1R, and the absence of synergy in vitro is consistent with the tumor microenvironment also being a factor in DZB efficacy and suggests additional means by which DZB could be stratified for cancer treatment in the clinic.
    Type of Medium: Online Resource
    ISSN: 0959-4973
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2023
    detail.hit.zdb_id: 2025803-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Anti-Cancer Drugs, Ovid Technologies (Wolters Kluwer Health), Vol. 34, No. 9 ( 2023-10), p. 1035-1045
    Abstract: Derazantinib (DZB) is an inhibitor of the fibroblast growth factor receptors 1–3 (FGFRi) with similar potency against colony-stimulating factor receptor-1 (CSF1R), a protein important in the recruitment and function of tumor-associated macrophages. DZB inhibited pCSF1R in the macrophage cell line RAW264.7, and tumor cells GDM-1 and DEL, and had the same potency in HeLa cells transiently over-expressing FGFR2. DZB exhibited similar potency against pCSF1R expressed by isolated murine macrophages, but as in the cell lines, specific FGFRi were without significant CSF1R activity. DZB inhibited growth of three tumor xenograft models with reported expression or amplification of CSF1R, whereas the specific FGFRi, pemigatinib, had no efficacy. In the FGFR-driven syngeneic breast tumor-model, 4T1, DZB was highly efficacious causing tumor stasis. A murine PD-L1 antibody was without efficacy in this model, but combined with DZB, increased efficacy against the primary tumor and further reduced liver, spine and lung metastases. Immunohistochemistry of primary 4T1 tumors showed that the combination favored an antitumor immune infiltrate by strongly increasing cytotoxic T, natural killer and T-helper cells. Similar modulation of the tumor microenvironment was observed in an FGFR-insensitive syngeneic bladder model, MBT-2. These data confirm CSF1R as an important oncology target for DZB and provide mechanistic insight for the ongoing clinical trials, in which DZB is combined with the PD-L1 antibody, atezolizumab.
    Type of Medium: Online Resource
    ISSN: 0959-4973
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2023
    detail.hit.zdb_id: 2025803-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Rapid Communications in Mass Spectrometry, Wiley, Vol. 21, No. 7 ( 2007-04-15), p. 1093-1099
    Abstract: The in vitro metabolic pattern of BAL19403, a novel macrolide antibiotic, was investigated by capillary liquid chromatography/quadrupole time‐of‐flight mass spectrometry (LC/QTOF‐MS) in incubations with human microsomes. For the elucidation of the metabolic pathway, BAL19403 labeled with four deuterium atoms (D 4 ) was used, and detection of metabolites performed using mixtures of the unlabeled (H 4 ) BAL19403 and its D 4 analogue (1:1) as substrate. All metabolites appeared with similar chromatographic behavior. MS/MS spectra of BAL19403 and its metabolites are dominated by non‐informative fragment ions. Therefore, the structure of the metabolites was elucidated mainly by accurate mass measurements with subsequent proposals of elemental compositions. Main biotransformations were N‐demethylation, lactone ring hydrolysis, and oxidation. Additionally, N‐dealkylation of the aromatic moiety was identified. This dealkylation results not only in formation of an aldehyde, according to the classical pathway, but also in formation of the corresponding alcohol and carboxylic acid. Final elucidation of their structures was possible, since this dealkylation takes place vicinal to the deuterium‐labeled part of BAL19403 and interferes with D/H exchange. The degree of D/H exchange, determined by analysis of the metabolite isotopic pattern, was used to elucidate the adjacent functional group. Copyright © 2007 John Wiley & Sons, Ltd.
    Type of Medium: Online Resource
    ISSN: 0951-4198 , 1097-0231
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2007
    detail.hit.zdb_id: 2002158-6
    detail.hit.zdb_id: 58731-X
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Bioorganic & Medicinal Chemistry, Elsevier BV, Vol. 12, No. 13 ( 2004-7), p. 3503-3519
    Type of Medium: Online Resource
    ISSN: 0968-0896
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2004
    detail.hit.zdb_id: 1501507-5
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Bioorganic & Medicinal Chemistry Letters, Elsevier BV, Vol. 20, No. 15 ( 2010-08), p. 4635-4638
    Type of Medium: Online Resource
    ISSN: 0960-894X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 1501505-1
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society for Microbiology ; 2007
    In:  Antimicrobial Agents and Chemotherapy Vol. 51, No. 6 ( 2007-06), p. 1956-1961
    In: Antimicrobial Agents and Chemotherapy, American Society for Microbiology, Vol. 51, No. 6 ( 2007-06), p. 1956-1961
    Abstract: BAL19403 exemplifies a new family of macrolide antibiotics with excellent in vitro activity against propionibacteria. MICs indicated that BAL19403 was very active against erythromycin-resistant and clindamycin-resistant propionibacteria with mutations in the region from positions 2057 to 2059 ( Escherichia coli numbering) of the 23S rRNA, although it is less active against those rare clinical isolates in which a methyltransferase, ErmX, confers macrolide and lincosamide resistance by dimethylation of the adenine moiety at position 2058. BAL19403 was predominantly bacteriostatic toward the propionibacteria, and population analyses indicated resistance selection frequencies for BAL19403 and the comparator drugs (erythromycin, clindamycin) in the range 10 −8 to 10 −9 for cutaneous propionibacteria with diverse antibiotic resistance profiles. On the basis of its antipropionibacterial activity and its high anti-inflammatory activity, BAL19403 represents a promising topical treatment for mild to moderate inflammatory acne vulgaris.
    Type of Medium: Online Resource
    ISSN: 0066-4804 , 1098-6596
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2007
    detail.hit.zdb_id: 1496156-8
    SSG: 12
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society for Microbiology ; 2007
    In:  Antimicrobial Agents and Chemotherapy Vol. 51, No. 12 ( 2007-12), p. 4361-4365
    In: Antimicrobial Agents and Chemotherapy, American Society for Microbiology, Vol. 51, No. 12 ( 2007-12), p. 4361-4365
    Abstract: BAL19403 is a macrolide antibiotic from a novel structural class with potent activity against propionibacteria in vitro. The antibacterial spectrum of BAL19403 covers clinical isolates with mutations in the 2057 to 2059 region of 23S rRNA that confer resistance to erythromycin and clindamycin. The basis of this improved activity was investigated by ribosome binding assays and by a coupled transcription and translation assay. The latter was specifically developed for the use of ribosomes from Propionibacterium acnes . BAL19403 inhibited protein expression by ribosomes from erythromycin-sensitive and erythromycin-resistant P. acnes with similar potencies if the resistance was due to G2057A or A2058G mutations. BAL19403 showed a 〉 10-fold higher activity than erythromycin against ribosomes from a strain with the erm (X) gene. Erm(X) confers high levels of macrolide and lincosamide resistance by dimethylation of A2058. Assays with such ribosomes showed that BAL19403 was potent enough to inhibit half of the total activity with a 50% inhibitory concentration very close to the value measured with erythromycin-sensitive ribosomes. We concluded from our data that the P. acnes strain with the erm (X) gene had a mixed population of ribosomes, with macrolide-sensitive and macrolide-resistant species.
    Type of Medium: Online Resource
    ISSN: 0066-4804 , 1098-6596
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2007
    detail.hit.zdb_id: 1496156-8
    SSG: 12
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2020
    In:  Journal of Clinical Oncology Vol. 38, No. 4_suppl ( 2020-02-01), p. 421-421
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 4_suppl ( 2020-02-01), p. 421-421
    Abstract: 421 Background: DZB is an oral small-molecule Fibroblast Growth Factor Receptor 1/2/3 inhibitor (FGFRi) with clinical activity in FGFR2-fusion-positive cholangiocarcinoma. DZB was screened for activity in gastrointestinal cancer (GIC), by using a panel of GIC cell-lines, human tumor xenografts and 30 GIC patient-derived xenograft (PDX) models. Methods: DZB anti-proliferative potency was determined in 26 GIC cell lines to determine the GI50. The GIC cell-line, SNU-16 was grown s.c. in nude mice and treated daily for 3-weeks with DZB at the MTD of 75 mg/kg, p.o. Plasma and tumor were removed and analyzed for drug-levels and PD biomarkers to assess pathway inhibition. DZB (@MTD) was tested in the PDX-screen (15 biliary, 13 gastric and 2 colorectal cancer; n≥3/group) using models with FGFR-fusions, FGFR-mutations and/or differing FGFR copy-number (CN)/RNA-seq expression levels. Efficacy and tolerability were quantified as a dT/C (treated/control). Results: Cellular GI50s ranged from 0.02-20 μM; the most sensitive (GI50≤0.5 μM) had FGFR-fusions or high-expression. In mice, DZB induced stasis of SNU-16 tumors (dT/C∼0.0) and was well tolerated (dT/C 〉 1.0); the plasma PK was dose-dependent with a Cmax of 2 μM (4 hr), a Cmin of 0.5 μM. DZB induced dose- and time-dependent changes in the MAPK-pathway and expression of downstream genes, consistent with its mode of action. In PDX-models, efficacy varied from no-response to 100% regression. Known driver-mutations were associated with partial-responses (best dT/C = 0.42), but models with FGFR-fusions, especially FGFR2-fusions, were very sensitive leading to stasis or strong-regression, particularly in gastric cancer. High-expression of FGFR2 was also associated with strong responses. There was no direct correlation between CN and high RNA-seq values suggesting amplification was not always a predictor of high expression. Endpoint PD-analyses of the PDX-models is ongoing to identify other potential stratifiers and PD-markers of response. Conclusions: DZB showed convincing activity in GIC-models with FGFR-fusions and/or high expression. A clinical trial is planned in patients with gastric cancer to investigate DZB as mono- and combination-therapy.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5645-5645
    Abstract: Background: BAL0891 is a dual inhibitor of threonine tyrosine kinase (TTK) and polo-like kinase 1 (PLK1). These kinases collaborate in activating the mitotic spindle assembly checkpoint (SAC) at the kinetochore (KT) to regulate chromosome alignment and segregation prior to mitotic exit. In vitro, BAL0891 has a combined prolonged effect on TTK and a transient effect on PLK1, leading to rapid disruption of the SAC that potentiates aberrant mitotic progression of tumor cells. In this work, efficacy of BAL0891 was investigated in mouse models of human triple negative breast cancer (TNBC) including evaluation of dose-dependency, drug exposure, target occupancy and a screen of activity across a panel of PDX models. Methods: The MDA-MB-231 cell line was grown sc in nude mice and treated with BAL0891, administered IV weekly (QW) or twice-weekly (2QW). Thirteen sc TNBC PDX models were screened for BAL0891 response using 2QW administration. Efficacy was quantified as deltaT/C (treated/control tumors). Plasma and tumor were analyzed for drug levels or TTK target occupancy by LC-MS/MS. The latter used a biotinylated TTK-specific probe and streptavidin-mediated isolation of unoccupied TTK, trypsin digestion and quantification of TTK-representative peptides. Results: BAL0891 efficacy was tested in the TNBC xenograft model MDA-MB-231 with QW or 2QW IV dosing schedules. All treatments were well tolerated, with no drug-related animal deaths. With MTD dosing, tumor regressions were observed, while different MTD fractions for both QW and 2QW schedules showed dose-dependent anti-tumor activity. The weekly MTD group was followed for an additional 20 days after treatment cessation on day 100. Strikingly, 3 of 8 tumors continued to shrink resulting in 2 (25%) pathologically confirmed cures. Consistent with the potent efficacy of intermittent MTD dosing, and prolonged tumor drug exposure, tumor TTK was fully drug-occupied for ≥ 6 days after the last administration; target occupancy was also dose-and drug exposure-dependent. To further evaluate BAL0891 anti-cancer activity in TNBC, a screen in 13 TNBC PDX models was conducted. Seven models exhibited deltaT/C & lt; 50%, with regressions observed in 3. Of these, 2 models showed persistent regressions ≥ 70% vs. baseline. Interestingly, evaluation of TTK target occupancy in selected models showed high target occupancy independent of tumor response, indicating target dependency rather than drug availability is important for anti-cancer activity. Conclusion: BAL0891 is a novel dual TTK/PLK1 mitotic checkpoint inhibitor with potent anti-cancer activity in TNBC models. Intermittent IV administration is well tolerated and associated with prolonged tumor drug exposure, prolonged TTK inhibition and notable anti-tumor efficacy. These data support further investigation of BAL0891 for the treatment of cancer patients (incl. TNBC). Citation Format: Heidi A. Lane, Felix Bachmann, Elisa Zanini, Paul McSheehy, Karine Litherland, Nicole Forster-Gross, Luc Bury, Diep Vu-Pham, Jos de Man, Wilhelmina E. van Riel, Guido JR Zaman, Rogier C. Buijsman, Laurenz Kellenberger. BAL0891: A novel dual TTK/PLK1 mitotic checkpoint inhibitor (MCI) that drives aberrant tumor cell division resulting in potent anti-cancer activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5645.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Molecular Cancer Therapeutics Vol. 8, No. 12_Supplement ( 2009-12-10), p. C229-C229
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 8, No. 12_Supplement ( 2009-12-10), p. C229-C229
    Abstract: Background: BAL27862 is a synthetic small molecule potently inducing apoptosis in cancer cells due to tubulin depolymerization. BAL27862 has a broad in vitro anti-proliferative activity against a range of human tumor lines (low nM IC50s) and elicits significant antitumor responses in animal models of human cancer, including multidrug-resistant tumors, after oral or intravenous administration. Materials and Methods: Anti-proliferative activity was analyzed with monolayer (crystal violet) or soft agar (clonogenic) assays. Effects on microtubules (MTs) were assessed by immunofluorescence (IF) or immunoblotting (IB) for -tubulin; centrosome maturation by IF for -tubulin. Results: BAL27862 showed potent anti-proliferative activity in vitro on 8 NSCLC cell lines (IC50 range 8.5–17nM). Significant activity was also observed on 3 patient-derived NSCLC lines, highly resistant to paclitaxel (clonogenic IC50s: paclitaxel & gt;3.5 M; BAL27862 15nM, 29nM, 300nM). In all tumor histotypes analyzed, BAL27862 elicited a unique MT phenotype, distinct from that observed with conventional MT-targeting agents (paclitaxel, vinblastine, colchicine) or other mitotic inhibitors. In proliferating A549 NSCLC cells (anti-proliferative IC50: 16±2nM; MT depolymerization IC50: 10–20nM) BAL27862 caused a concentration-dependent effect on MTs. In interphase cells, at concentrations ≥15nM the MT network collapsed towards the MT-organizing center, with an absence of peripheral MTs. In dividing cells, an apparent fragmentation of the mitotic spindle occurred at 15nM, associated with the appearance of tiny MT asters. The asters decreased in size as concentrations increased (15–100nM), and were co-localized with condensed chromatin. A comparative titration (5–100nM) with the clinically used MT destabilizer vinblastine (VBL) indicated no phenotypic overlap. VBL-treated interphase cells had a mesh-like MT network with peripheral MTs intact, while in dividing cells aberrant, spindle-like structures were observed at concentrations ≥15nM. Using non-immortalized HS68 human fibroblasts, no obvious effects of BAL27862 (10–20nM) on centrosome number or size were observed, as defined by -tubulin staining; in contrast to an increase in size and decrease in apparent resolution of the centrosomes after treatment with VBL at the same concentrations. BAL27862 treatment also converted the abnormal spindle structures induced by the MT destabilizers VBL or colchicine into the distinct and dominant BAL27862 MT phenotype, further highlighting it's novel mechanism of MT disruption. Conclusions: BAL27862 is a new tubulin-interacting agent with a novel mechanism of action leading to a unique MT phenotype. These observations, together with a broad antitumor activity targeting refractory tumors, strongly support further development of BAL27862 as an anticancer agent. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):C229.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...