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 Medical Virology, Wiley, Vol. 95, No. 1 ( 2023-01)
    Abstract: The major challenge in COVID‐19 vaccine effectiveness is immune escape by SARS‐CoV‐2 variants. To overcome this, an Omicron‐specific messenger RNA (mRNA) vaccine was designed. The extracellular domain of the spike of the Omicron variant was fused with a modified GCN4 trimerization domain with low immunogenicity (TSomi). After immunization with TSomi mRNA in hamsters, animals were challenged with SARS‐CoV‐2 virus. The raised nonneutralizing antibodies or cytokine secretion responses can recognize both Wuhan S and Omicron S. However, the raised antibodies neutralized SARS‐CoV‐2 Omicron virus infection but failed to generate Wuhan virus neutralizing antibodies. Surprisingly, TSomi mRNA immunization protected animals from Wuhan virus challenge. These data indicated that non‐neutralizing antibodies or cellular immunity may play a more important role in vaccine‐induced protection than previously believed. Next‐generation COVID‐19 vaccines using the Omicron S antigen may provide sufficient protection against ancestral or current SARS‐CoV‐2 variants.
    Type of Medium: Online Resource
    ISSN: 0146-6615 , 1096-9071
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 752392-0
    detail.hit.zdb_id: 1475090-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 227-227
    Abstract: Zinc(II)-dipicolylamine (Zn-DPA) have been described to specifically complex with phosphatidylserine (PS), which has a higher level at the external surface of cancer cells in tumorigenic condition. BPRDP056 is a Zn-DPA-SN38 conjugate designed to provide a PS-targeting drug delivery of cytotoxic payload SN-38 at the tumor microenvironment, thereby, decrease the dosage of SN38, while induce apoptosis in cancer cells. The in vivo therapeutic efficacy of BPRDP056 against the growths of human tumors has been shown significant in mice subcutaneously bearing a tumor type of pancreas, prostate, colon, liver, breast and glioblastoma, as well as in mice with an orthotopic pancreatic tumor. BPRDP056 shrunk tumors at a lower dosing intensity (~20%) of SN38 compared to CPT-11 in all models tested. Micro-Western assays showed that BPRDP056 exhibited apoptotic cell death signal levels similar to those of CPT-11 in the treated tumors in mice. Furthermore, pharmacokinetic and preliminary toxicology studies showed that BPRDP056 has a good stability in circulation with an acceptable therapeutic safety window in mice. BPRDP056 has been demonstrated with a tumor targeting ability and thus increases the cytotoxic payload SN38 concentration in situ for improved efficacy. Its therapeutic spectrum against malignant neoplasm will be expected to cover the PS-rich tumor microenvironment of all cancer types. BPRDP056 is a first-in-class Small Molecule Drug Conjugate for anti-cancer therapy. Citation Format: Chia-Yu Hsu, Yun-Yu Chen, Chen-Fu Lo, Tai-Yu Chiu, Ching-Ping Chen, Chen-Lung Huang, Chung-Yu Huang, Min-Hsien Wang, Yu-Sheng Chao, Joe C. Shih, Teng-Kuang Yeh, Lun K. Tsou, Chiung-Tong Chen. BPRDP056, a novel small molecule drug conjugate specifically targeting phosphatidylserine for cancer therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 227.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3527-3527
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3527-3527
    Abstract: 14-3-3 family consists of seven highly conserved isoforms and most of them are identified as oncogenes in various types of cancer except for 14-3-3σ, a well-known tumor suppressor. To clarify the suppressor characteristic of 14-3-3σ divergent from others, we found that the major difference between the other oncogenic members and 14-3-3σ by protein alignment was an amino acid substitution (Y180H) by which the SH2-binding motif (YYEI) is disrupted and cannot be phosphorylation. Thus, we generated a H180Y 14-3-3σ mutant and investigated the impact of the acquired YYEI motif on the tumor suppression. First, we found that 14-3-3σ decreased cancer invasion. Surprisingly, the H180Y mutant not only enhanced cell invasion but increased cell viability. Meanwhile, the interaction between H180Y mutant and Src was higher than wild type. It indicates one amino acid substitution switches 14-3-3σ from tumor suppressor to oncogene. Hence, the YYEI motif might be important for the oncogenicity of other 14-3-3 proteins. Indeed, we demonstrated that 14-3-3ζ interacted with Src through Y178 phosphorylation, which is crucial for the binding of 14-3-3ζ with Src-SH2 domain. Owing to the importance of Y178 phosphorylation in the 14-3-3ζ/Src interaction, we introduced the Y178F 14-3-3ζ mutant to lung cancer cells and confirmed that Y178 phosphorylation is important for the increase of invasion and viability. Furthermore, we also revealed an amino acid switch of 14-3-3σ from YYEI to HYEI during evolutionary progression. Taken together, our findings suggest that YYEI motif is essential for 14-3-3 proteins to interact with Src and to regulate Src-mediated cell functions. Citation Format: Wen-Hsin Chang, Ching-Hsien Chen, Qi-Sheng Hong, Jian-Wei Chen, Sung-Liang Yu, Chiung-Tong Chen. YYEI motif is critical to oncogenicity of 14-3-3 proteins. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3527. doi:10.1158/1538-7445.AM2014-3527
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4413-4413
    Abstract: Drug delivered by conjugate with antibody is expected to increase drug concentrations at the tumor sites for improved antitumor effects. Showing some successes, the antibody-drug conjugate system has certain limitations such as difficulties in protein characterization, high cost and individual variations in expression of the epitope, which might abrogate the antibody recognition and cause premature drug release arising from antibody-drug linker instability and thus ineffectiveness. Phosphatidylserine (PS) exists in bulk amount in the tumor microenvironment. Studies showed that a small sized Zinc (II)-dipicolylamine (Zn-DPA) molecule was superior in locating PS surrounding the apoptotic sites in vivo. Although Zn-DPA has been constructed incorporating fluorescent dyes as optical tools for imaging the PS-exposed cell membranes to identify tumor sites from healthy cells in animals, drug conjugates with Zn-DPA has never been described. We aimed to design, synthesize and evaluate a Zinc-dipicolylAmine directed Pharmaceutical delivery System (ZAPS), a platform for spatial- and temporal-release of drug specifically at the targeted tumor site. Zn-DPA was conjugated through standard coupling conditions with various linkers to SN-38 and the Zn-DPA-linker-SN-38 conjugates were obtained and investigated for structure-activity relationships on the chemical stability in plasma and cytotoxicity against cancer cells. Promising ZAPS-SN-38 conjugates were evaluated for activities against tumor growths in nude mice. CPT-11 and SN-38 were included for comparisons. ZAPS-SN-38 conjugates were examined for tolerable doses in mice. Among the ZAPS-SN-38 conjugates synthesized, ZAPS001 was found chemically stable in mouse plasma and in vitro active against several cancer cells. ZAPS001 dose-dependently inhibited the growth of Colo205 tumors in nude mice. Zn-DPA-linker001 (ZAPS001 without SN-38) showed no inhibition effect on the tumor growth. Intriguingly, employing only 40% of the SN-38 delivered by CPT-11 (40 mg/kg), ZAPS001 resulted in 8-fold increase in antitumor activity compared to that of CPT-11 given at the same dose regimen. Furthermore, ZAPS001 is also active against pancreatic Mia-Paca2 and BxPC-3 tumors in mice. An increased SN-38 level delivered by ZAPS001 to the tumors in mice was also observed. ZAPS is thus capable of selectively associating with PS-exposing tumor cells/tissues to achieve site-specific delivery of anticancer agents. Our findings strongly support that ZAPS conferred an “in situ dose amplification” effect, i.e., the cytotoxics-induced PS exposure could specifically recruit more ZAPS-drug conjugates to the tumor site and further enhance its therapeutic effect. ZAPS provides spatial and temporal controls to increase drug concentration at targeted disease sites, reduce drug dosage, lessen the toxic side effects, and thus increase therapeutic index of the anticancer drugs. Citation Format: Lun K. Tsou, Yu-Wei Liu, Yun-Yu Chen, Chen-Fu Lo, Teng-Kuang Yeh, Chien-Huang Wu, Kak-Shan Shia, Joe C. Shih, Brian D. Gary, Koon Y. Pak, Chiung-Tong Chen. Zinc-dipicolylamine directed pharmaceutical delivery system (ZAPS) as an innovative cancer drug delivery platform. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4413. doi:10.1158/1538-7445.AM2015-4413
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 64, No. 13 ( 2004-07-01), p. 4621-4628
    Abstract: BPR0L075 is a novel synthetic compound discovered through research to identify new microtubule inhibitors. BPR0L075 inhibits tubulin polymerization through binding to the colchicine-binding site of tubulin. Cytotoxic activity of BPR0L075 in a variety of human tumor cell lines has been ascertained, with IC50 values in single-digit nanomolar ranges. As determined by flow cytometry, human cervical carcinoma KB cells are arrested in G2-M phases in a time-dependent manner before cell death occurs. Terminal deoxynucleotidyl transferase-mediated nick end labeling assay indicates that cell death proceeds through an apoptotic pathway. Additional studies indicate that the effect of BPR0L075 on cell cycle arrest is associated with an increase in cyclin B1 levels and a mobility shift of Cdc2 and Cdc25C. The changes in Cdc2 and Cdc25C coincide with the appearance of phosphoepitopes recognized by a marker of mitosis, MPM-2. Furthermore, phosphorylated forms of Bcl-2, perturbed mitochondrial membrane potential, and activation of the caspase-3 cascade may be involved in BPR0L075-induced apoptosis. Notably, several KB-derived multidrug-resistant cell lines overexpressing P-gp170/MDR and MRP are resistant to vincristine, paclitaxel, and colchicine but not to BPR0L075. Moreover, BPR0L075 shows potent activity against the growth of xenograft tumors of the gastric carcinoma MKN-45, human cervical carcinoma KB, and KB-derived P-gp170/MDR-overexpressing KB-VIN10 cells at i.v. doses of 50 mg/kg in nude mice. These findings indicate BPR0L075 is a promising anticancer compound with antimitotic activity that has potential for management of various malignancies, particularly for patients with drug resistance.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1937-1937
    Abstract: Small cell lung cancer (SCLC) accounts for approximately 15% of all lung cancers, leading to ~30,000 deaths each year in the United States. SCLC patients often present with metastasis at time of diagnosis, excluding surgery as a treatment option. While patients show high response rate to standard chemotherapy such as cisplatin/etoposide, they soon develop drug resistance and disease progression. Therefore, new therapeutic strategies are urgently needed for SCLC. BPR6K609S0 is a novel Aurora kinase inhibitor which has been designed to inhibit the kinase activity of Aurora A, and induces proteasome-mediated degradation of MYC. The BPR6K609S0 active molecule provokes cell apoptosis and inhibits proliferation of several SCLC cell lines with IC50 & lt; 100 nM. Oral administration of BPR6K609 induces & gt;60 % tumor regression in a NCI-H446 xenograft mouse model. In addition, BPR6K609 further reduces tumor progression in NCI-H446 xenograft mice pre-treated with LY3295668, an Aurora A-selective inhibitor which is currently under clinical investigation. These results support the clinical potential of BRP6K609S0 for the treatment of SCLC. Citation Format: Ya-Hui Chi, Chun-Ping Chang, Yi-Yu Ke, Wen-Hsing Lin, Wan-Ping Wang, Chia-Hua Tsai, Yen-Ting Chen, Yu-Jie Su, Ming-Chun Hung, Zhong-Wei Wu, Mine-Hsine Wu, Teng-Kuang Yeh, Ching-Ping Chen, Jen-Shin Song, Chiung-Tong Chen, Chuan Shih. BPR6K609: An Aurora kinase inhibitor targeting small cell lung cancer with MYC amplification [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1937.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2013
    In:  Blood Vol. 122, No. 21 ( 2013-11-15), p. 1448-1448
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1448-1448
    Abstract: Acute myeloid leukemia (AML) carrying t(8;21)(q22;q22) and inv(16)/t(16;16)(p13;q22) are classified as French-American-British (FAB) AML subtype M2 or monocytic with eosinophilic differentiation (M4Eo) by morphology and as core binding factor (CBF)-AML according to pathogenesis. CBF-AML accounts for approximately 15% of AML and frequently harbors c-KIT mutation (17∼46%). C-KIT mutated CBF-AML patients usually have higher baseline white blood cell count, higher relapse rate and shorter event free survival/overall survival after conventional chemotherapy than those without c-KIT mutation. It is conceived that c-KIT mutation is a crucial hit and cooperates with AML1-ETO resulting from t(8;21)(q22;q22) to cause overt AML. Imatinib, a tyrosine kinase inhibitor suppressing c-KIT activation, has been used in c-KIT mutated AML, systemic mastocytosis (SM) and gastrointestinal stromal tumor (GIST). However, c-KIT exon17 D816V mutation, a frequent mutation of CBF-AML and SM, is associated with primary imatinib resistance. 1J373, a multi-targeted tyrosine kinase inhibitor, which was initially designed as a FLT3 inhibitor but later found to target c-KIT as well. It has been shown to effectively inhibit the proliferation of FLT3-ITD mutated leukemia cell lines, MV4;11 and MOLM-13, both in vitro and in vivo. (unpublished data) Among a series of myeloid leukemia cell lines without FLT3-ITD mutation, including THP-1, HL-60, K562, KG-1, and kasumi-1, the sensitivity to 1J373 is closely associated with the presence of constitutive c-KIT activation (Figure 1A). The IC50 of 1J373 for cells with (K562, KG-1 and kasumi-1) and without (THP-1 and HL-60) constitutive c-KIT activation was below 50 nM and beyond 1000 nM, respectively. 1J373 suppressed the phosphorylation of c-KIT for cell lines with constitutively activated c-KIT, which suggested that 1J373 may suppress the proliferation of KG-1, K562, and kasumi-1 by inhibiting c-KIT (Figure 1B).We further compared the efficacy of 1J373 and imatinib in kasumi-1, a cell line with t(8;21)/AML1-ETO and c-KIT exon 17 N822K mutation. At 1000nM of concentration, the phosphorylation of c-KIT was effectively inhibited by imatinib at 2-hour but partially recovered after 8 hours; while 1J373 treatment resulted in a sustained inhibition for 24 hours. The inhibition of c-KIT activation by both agents was accompanied with corresponding changes in the phosphorylation status of its downstream signaling pathway molecules, including PI3K, AKT, mTOR, and MAPK (Figure 2). 1J373 induced cell cycle arrest of kasumi-1 at G1 phase with increase of subG1 population time-dependently and induced apoptosis of kasumi-1 through activation of caspase 8 and 9, and upregulation of proapoptotic proteins Bax and Bak. The in vivo experiments are in progress. In conclusion, 1J373, a multi-targeted tyrosine kinase inhibitor, can effectively inhibit the proliferation and induce the apoptosis of c-KIT activated leukemia cells. It has the potential to be used in clinical practice to treat c-KIT driven, particularly c-KIT mutated, AML.Figure 1c-KIT and phosphorylated c-KIT expression in myeloid leukemia cell linesFigure 1. c-KIT and phosphorylated c-KIT expression in myeloid leukemia cell linesFigure 2c-KIT and its downstream signalings expression in kasumi-1 cells treated with imatinib and 1J373Figure 2. c-KIT and its downstream signalings expression in kasumi-1 cells treated with imatinib and 1J373 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: 2013
    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: Anesthesiology, Ovid Technologies (Wolters Kluwer Health), Vol. 126, No. 5 ( 2017-05-01), p. 952-966
    Abstract: The authors investigated the pharmacology and signaling pathways of the opioid receptors modulated by compound 1, 1-(2,4-dibromophenyl)-3,6,6-trimethyl-1,5,6,7-tetrahydro-4H-indazol-4-one. Methods In vitro studies of compound 1 were assessed by using a radioligand-binding assay (n = 3), a cyclic adenosine monophosphate assay (n = 3), a β-arrestin assay (n = 3), an internalization assay (n = 3), and an immunohistochemistry (n = 8). In vivo studies of compound 1 were characterized using a tail-flick test (n = 5 to 6), tail-clip test (n = 7), von Frey hair test (n = 5), and charcoal meal test (n = 5). Results Compound 1 elicited robust effects in μ-opioid (mean ± SD; binding affinity: 15 ± 2 nM; cyclic adenosine monophosphate assay: 24 ± 6 nM), δ-opioid (82 ± 7 nM; 1.9 ± 0.1 μM), and κ-opioid (76 ± 9 nM; 1.4 ± 0.5 μM) receptor–expressing cells. Compound 1 acts as a full agonist of β-arrestin-2 recruitment in μ-opioid (1.1 ± 0.3 μM) and δ-opioid (9.7 ± 1.9 μM) receptor–expressing cells. Compound 1 caused less gastrointestinal dysfunction (charcoal meal test: morphine: 82 ± 5%; compound 1: 42 ± 5%) as well as better antinociception in mechanical pain hypersensitivity (tail-clip test: morphine: 10 ± 3 s; compound 1: 19 ± 1 s) and in cancer-induced pain (von Frey hair test: morphine: 0.1 ± 0.1 g; compound 1: 0.3 ± 0.1 g) than morphine at equi-antinociceptive doses. Conclusions Compound 1 produced antinociception with less gastrointestinal dysfunction than morphine.
    Type of Medium: Online Resource
    ISSN: 0003-3022
    RVK:
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2017
    detail.hit.zdb_id: 269-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Spandidos Publications ; 2012
    In:  International Journal of Oncology Vol. 41, No. 3 ( 2012-9), p. 1068-1084
    In: International Journal of Oncology, Spandidos Publications, Vol. 41, No. 3 ( 2012-9), p. 1068-1084
    Type of Medium: Online Resource
    ISSN: 1019-6439 , 1791-2423
    RVK:
    Language: English
    Publisher: Spandidos Publications
    Publication Date: 2012
    detail.hit.zdb_id: 2079608-0
    detail.hit.zdb_id: 1154403-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal of General Virology, Microbiology Society, Vol. 100, No. 3 ( 2019-03-01), p. 457-470
    Type of Medium: Online Resource
    ISSN: 0022-1317 , 1465-2099
    RVK:
    RVK:
    Language: English
    Publisher: Microbiology Society
    Publication Date: 2019
    detail.hit.zdb_id: 219316-4
    detail.hit.zdb_id: 2007065-2
    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...