GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (42)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (42)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5205-5205
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 5205-5205
    Kurzfassung: Background: Systematically reviewing published studies on a broad range of potential radiosensitizers, we previously reported that poly(ADP-ribose) polymerase inhibitors (PARPi) may have a unique radiosensitizing effect to enhance β-components of the linear-quadratic model. In contrast, many other radiosensitizing agents predominantly modified α-components. The aim of this study is to elucidate underlying mechanisms of “β-radiosensitization”. Methods and Results: We first confirmed that treatments with PARPi PJ34, olaparib, or veliparib two hours prior to and 48 hours after 10 Gy of ionizing radiation (IR) potentiated radiation-induced cell death in human cancer cells HCT116, HT29, H460 and A549 using the colony-forming assay. Tumor regrowth assay with 3-dimensional multicellular spheroids of HCT116 and H460 cells showed that PJ34 significantly delayed tumor regrowth when combined with single dose of 10 Gy but not in tumors treated with 10 Gy in 5 fractions. Similar results were found using HCT116 tumor xenograft model in mice. We next examined what cellular events may underlie the radiosensitization using PARPi PJ34 in HCT116 cells. Kinetics of DNA damage responses were first measured by phosphorylation of histone H2AX (γH2AX). PJ34 did not change an initial increase of γH2AX following 10 Gy IR, but γH2AX levels decreased faster in PJ34-treated cells than control cells. Immunocytochemical analysis of Rad51 showed that PJ34 reduced Rad51 foci formations up to 6 hours following 10 Gy IR. IR-induced chromosomal aberrations were quantitated by the C-banding of metaphase spreads. More dicentric chromosomes were found in PJ34-treated cells than the control. These data suggested that PARPi did not inhibit total DNA double-strand break (dsb) repair but altered the selection in DNA dsb repair pathways with increased end-joining type(s) of (mis)repair compensating for reduced homologous recombination repair. Cell cycle profiles showed sustained 4C DNA cell peaks in cells treated with PJ34 up to 72 hours following 10 Gy IR. Cell cycle markers (cyclin B1 and phosphorylated histone H3) indicated that the 4C DNA peaks of the PJ34- treated cells included substantial amount of G1 tetraploid cells in addition to G2- or M-phase diploid cells. Additionally, senescence-associated β-galactosidase staining showed that PJ34 promoted radiation-induced premature senescence, while smaller IR-induced sub-G1 populations were found in PJ34-treated cells than the control. Conclusion: PARPi increased IR-induced chromosomal aberrations through the altered selection in DNA dsb repair pathways. The increased quadratic misrepair enhanced β-components of IR-induced cell death mediated by the increased formation of G1 tetraploid cells and premature senescence. DNA dsb repair pathway selection may be a novel and effective target to elicit β-radiosensitization. Citation Format: Yuji Seo, Keita Yoshizaki, Keisuke Tamari, Yutaka Takahashi, Keisuke Otani, Masahiko Koizumi, Kazuhiko Ogawa. Poly(ADP-ribose) polymerase inhibitors induce β-radiosensitization through an altered selection of DNA double-strand break repair pathways [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5205. doi:10.1158/1538-7445.AM2017-5205
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2017
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 24_Supplement ( 2016-12-15), p. B66-B66
    Kurzfassung: Background and Aims: Pancreatic ductal adenocarcinoma (PDAC) is well characterized by dense fibrotic stroma with abundant cancer-associated fibroblasts (CAFs). As CAFs are activated during tumorigenesis and acquire tumor-promoting properties, activated CAFs have been implicated in PDAC progression; however, the precise mechanisms of their activation remain largely unknown. The bromodomain and extraterminal (BET) domain proteins are epigenetic reader proteins that recognize acetylated amino acid residues on histone tails and facilitate gene transcription. Recent studies have demonstrated therapeutic efficacy of BET inhibitors on various cancers including PDAC, mainly through suppression of c-myc transcription; however, how BET inhibitors suppress PDAC growth and their effects on CAFs remains largely unknown. Using patient-derived tumor xenografts (PDX) and primary CAFs, we investigated the therapeutic efficacy and dissected the underlying mechanisms of a BET inhibitor, JQ1, on human PDAC and CAFs. Methods: We established PDX lines and primary CAFs from surgically resected human PDAC specimen. For in vivo analyses, mice bearing subcutaneous tumor were treated with vehicle or JQ1. For in vitro analyses, patient-derived PDAC cells and CAFs were treated with vehicle or JQ1 and analyzed separately. To explore the pro-tumorigenic role of secretion from CAFs, PDAC cells were cultured with conditioned medium (CM) that was collected from DMSO- or JQ1- treated CAFs. Chromatin immunoprecipitation (ChIP) assay was performed to assess the binding of transcription factors and histone modifications which are associated with altered gene expression in CAFs by JQ1 treatment. Results: In vivo experiments revealed that volumes and weights of subcutaneous PDX tumors were significantly smaller in JQ1-treated mice than vehicle-treated mice. Unexpectedly, however, JQ1 exerted only minimal effects to the proliferation of PDAC cells that were isolated from PDX tumors and cultured in vitro, suggesting the involvement of cell-extrinsic mechanisms in the JQ1-mediated suppression of tumor growth in vivo. Of note, histopathological analysis of PDX tumors revealed that JQ1 treatment dramatically ameliorated desmoplastic change, with reduction in extracellular matrix (ECM) deposition and α-SMA expressing CAFs. As α-SMA expression and ECM production is a hallmark of activated CAFs, we hypothesized that JQ1 might inactivate CAFs, thereby reducing their tumor-promoting properties. To test this hypothesis, qPCR was performed to analyze gene expression in primary CAFs cultured in vitro and also in stromal cells in PDX tumors in vivo. As expectedly, JQ1 suppressed the expression of genes implicated in the properties of activated CAF, including ECM, cytokines and growth factors both in vitro and in vivo. Furthermore, when PDAC cells were cultured with CM from DMSO–treated CAFs, proliferation of PDAC cells were promoted along with activation of MAPK, AKT, and STAT3 pathways, which was abrogated when cultured with CM from JQ1-treated CAFs. Consistently, immunoblotting and immunohistochemistry of PDX tumors demonstrated that JQ1 reduced phosphorylation of ERK, AKT, and STAT3 in PDAC cells in vivo. Mechanistically, we found that JQ1 suppressed hedgehog and TGF-β/SMAD3 pathways, both of which play central roles in CAF activation, through disruption of BRD4 recruitment to the promoter regions of their target genes. Conclusions: BET proteins are critical regulators of CAF-activation in PDAC. Inactivation of CAFs by BET inhibition offers a novel therapeutic approach for PDAC. Citation Format: Keisuke Yamamoto, Keisuke Tateishi, Yotaro Kudo, Mayumi Hoshikawa, Mariko Tanaka, Takuma Nakatsuka, Hiroaki Fujiwara, Koji Miyabayashi, Ryota Takahashi, Yasuo Tanaka, Hideaki Ijichi, Yousuke Nakai, Hiroyuki Isayama, Yasuyuki Morishita, Taku Aoki, Yoshihiro Sakamoto, Kiyoshi Hasegawa, Norihiro Kokudo, Masashi Fukayama, Kazuhiko Koike.{Authors}. BET inhibition remodels tumor stroma and suppresses progression of human pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2016 May 12-15; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(24 Suppl):Abstract nr B66.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2016
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 2_Supplement ( 2016-01-15), p. A40-A40
    Kurzfassung: The role of genetic mutations in the pathogenesis of pancreatic ductal adenocarcinoma (PDAC) is well established. However, it is still unclear if epigenetic aberrations contribute to PDAC progression. We previously reported a novel role for the H3K27 demethylase KDM6B/JMJD3 in regulating PDAC progression (Carcinogenesis 2014;35(11):2404-14). KDM6B was downregulated in high grade PDACs and knockdown (KD) of KDM6B in PDAC cells increased the tumorigenicity and enhanced the aggressive phenotypes of these cells in vivo. Furthermore, CCAAT enhancer binding protein alpha gene (CEBPA) was identified as a direct target of KDM6B, and reduced KDM6B- C/EBPα axis was resulted in increased aggressiveness in PDAC cells. To dissect further the pathological effects caused by loss of KDM6B- C/EBPα function in PDAC cells, we tried to identify a surrogate molecular marker of the cells lacking of KDM6B- function. For the purpose, we used a cDNA microarray to compare the expression profiles of KDM6B- KD and control BxPC3 PDAC cells. 906 genes were upregulated and 639 downregulated in KDM6B- KD BxPC3 cells compared to the control cells. We focused on 58 genes encoding cell-surface molecules that were upregulated in KDM6B- KD BxPC3 cells and validated the expression of 9 surface marker candidates, including 3 that have already been reported to be expressed on PDAC tumor-initiating cells, namely, CD24, CD44, and CD133. Only CD47 was significantly upregulated in the KDM6B- KD BxPC3 cells as confirmed by both quantitative RT-PCR and flow cytometric analysis. CD47 was also upregulated in other PDAC cell lines following KDM6B knockdown. It has recently been reported that CD47 is upregulated in various malignancies and that an increase in CD47 expression is correlated with a poor prognosis. In line with the previous reports, CD47high cells formed about 4-fold more spheres than non-CD47high cells. The close relationship between CD47 expression and the sphere-forming ability was supported by the finding that CD47low cells formed even fewer spheres. To confirm these results in vivo, the sorted CD47high and non-CD47high cells were subcutaneously xenotransplanted into nude mice. All CD47high cells formed tumors more efficiently than the unfractionated KDM6B- KD cells, while the tumor-forming rate of non-CD47high cells was comparable to that of the Ctrl cells. In addition, when the cells were injected into the spleens of nude mice, CD47high cells demonstrated higher liver metastatic potential than the non-CD47high population. These data suggested that the increased tumor-initiating potential of KDM6B- KD cells was attributable to this induced CD47high population. Consistently, the expression of KDM6B and C/EBPα inversely correlated with CD47 expression and tumor grade in human PDAC tumors. Collectively, our data provides a link between epigenetic change and PDAC progression, thus offering a novel strategy to target PDAC aggressiveness by intervening in the dynamics of epigenetic process. Citation Format: Keisuke Yamamoto, Keisuke Tateishi, Yotaro Kudo, Koji Miyabayashi, Ryota Takahashi, Takuma Nakatsuka, Hiroaki Fujiwara, Yousuke Nakai, Yasuo Tanaka, Hideaki Ijichi, Hiroyuki Isayama, Kazuhiko Koike. Emergence of CD47- high expression cells confers enhanced tumorigenicity upon KDM6B suppression in pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Chromatin and Epigenetics in Cancer; Sep 24-27, 2015; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2016;76(2 Suppl):Abstract nr A40.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2016
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4585-4585
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4585-4585
    Kurzfassung: Osteosarcoama is one of the common malignancies at bone in children and adolescent. Recent study demonstrated that combination of anti-PD-L1 and anti-CTLA-4 antibodies provided grate control of metastatic osteosarcoma. However, a strong local treatment strategy including surgery or high precision radiation therapy is necessary to elucidate osteosarcoma. Radiation therapy plays an important role in local control for malignant tumors but previous studies demonstrated that radiation enhanced immune response in which not only local tumor regression at irradiated sites but also regression of metastatic tumor outside the radiation field were observed. Although this phenomenon, so called the abscopal effect, is rarely seen, recent studies demonstrated that combination of X-ray irradiation with checkpoint blockades provided higher probability of the abscopal effect for some kind of tumors. However, the effect of x-ray irradiation combined with checkpoint blockade on the abscopal effect for osteosarcoma has been totally unknown. We investigated whether local X-ray irradiation combined with the anti-PD-L1 and anti-CTLA-4 antibodies enhances local and distant antitumor efficacy for osteosarcoma. LM8 mouse osteosarcoma cells were inoculated into both legs of C3H mice. Mice were treated by 10 Gy X-ray irradiation alone to the tumor in the one side leg (RAD group) at day 12, 150 ug of anti-PD-L1 and anti-CTLA-4 antibodies (P1C4 group) at days 9, 12, and 15, or those combination (COMB group). Administration of anti-PD-L1 and anti-CTLA-4 antibodies provided tumor growth delay or complete response at day 37 for about 20% of the mice. X-ray irradiation strongly inhibited tumor growth at irradiated tumor but not in unirradiated tumor. On the other hand, the combination therapy provided the strongest tumor growth inhibition not only at irradiated tumor but also at unirradiated tumor for about 89% of the mice. Accordingly, lung metastasis in mice in COMB group was strongly reduced by 97% with the significant survival benefit compared with the mice in P1C4 group. Flow cytometric analysis revealed that mice in COMB group significantly recruited CD8 tumor infiltrating lymphocytes with moderate reduction of regularly T cells (Tregs), thereby increasing the CD8/Treg ratio. Furthermore, quantitative real time PCR showed significant induction of PD-L1 on irradiated LM8 cells in vitro. The radiation-induced upregulations of PD-L1, B7-1, and B7-2, the ligands of CTLA-4, were also confirmed by flow cytometry, indicating that the enhanced efficacy anti-PDL1 and CTLA-4 antibodies may associate with these upregulations by radiation. These results suggest that X-ray irradiation contributes to the enhancement of the efficacy for the distant metastasis as well as local control in the treatment of anti-PD-L1 and anti-CTLA-4 antibodies for osteosarcoma. Our data provide a rational to establish a new therapeutic strategy and start up a clinical trial for osteosarcoma. Citation Format: Yutaka Takahashi, Tomohiro Yasui, Keisuke Tamari, Kazumasa Minami, Masahiko Koizumi, Yuji Seo, Fumiaki Isohashi, Keisuke Ohtani, Ryosuke Kambe, Kazuhiko Ogawa. Radiation combined with checkpoint blockades enhanced antitumor efficacy for osteosarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4585. doi:10.1158/1538-7445.AM2017-4585
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2017
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 13_Supplement ( 2015-07-01), p. A55-A55
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 13_Supplement ( 2015-07-01), p. A55-A55
    Kurzfassung: TGF-beta signaling has a crucial role in pancreatic tumorigenesis, and almost all of pancreatic cancers carry at least one genetic alteration of TGF-beta related genes, such as SMAD4, TGFBR2, SMAD3, and BMPR2. However, the role of BMP signaling in pancreatic cancer remains unclear. Previous studies reported the depletion of BMP signaling resulted in the aggressive phenotype of cancer, and some reported BMP signaling played an important role in tumor progression and metastasis. We have already established pancreas-specific Tgbr2 knockout mice in the context of Kras activation, which clinically and histopathologically recapitulate human PDAC. With regard to PDAC, Smad4 mutation or deletion is more commonly observed, however the Smad4 knockout mice with activating Kras mutation was reported to show cystic type tumor of pancreas. Therefore, our Kras+Tgfbr2KO might be the closest approximation of the human PDAC in terms of histology. We examined the effect of BMP signaling on the tumorigenesis and progression of PDAC using this mouse model. We performed immunohistochemistry of murine PDAC to evaluate whether BMP signaling was related to the PDAC progression. We examined the effect of Bmp4 and Bmp7 on the proliferation, invasion and adhesion using murine PDAC and PanIN cells in vitro. We have already established the murine PDAC cell lines from Pancreas-specific Kras+Tgfbr2KO mice and murine PanIN cells from Pancreas-specific activating Kras mutation mice. Bmpr2 was knocked down in PanIN cell lines using shRNA, and we examined whether the effect of BMP signaling was canceled by Bmpr2 knockdown. In vivo, we evaluated the effect of BMP signaling on tumor growth and tumor-stromal interaction using the xenograft mouse model of Bmpr2-negative PanIN cells. The immunohistochemistry of murine pancreas tissues demonstrated that Smad1/5/8 was more strongly phosphorylated in PDAC compared to PanIN lesion. We also observed that Smad1/5/8 was phosphorylated in stromal cells surrounding tumor areas, which was likely to suggest the importance of BMP signaling in PDAC progression and tumor-stromal interaction. In vitro, both Bmp4 and Bmp7 did not affect the proliferation and invasion of PDAC and PanIN cells, but they increased the adhesion of PDAC and PanIN cells, and knockdown of Bmpr2 canceled the effect of Bmps. In vivo, we evaluated the growth of subcutaneous tumor allograft and the tumors of Bmpr2-negative PanIN cells showed slower tumor growth than tumors of the control, differently from the results in vitro. These results suggested that BMP signaling was associated with the tumor-stromal interaction and played important role in tumor progression. In this study we evaluated the role of BMP signaling in pancreatic cancer using pancreas-specific Kras+Tgfbr2KO mice, and demonstrated that BMP signaling played important role in the adhesion and progression of pancreatic cancer, which was due to the tumor-stromal interaction. Citation Format: Koji Miyabayashi, Hideaki Ijichi, Ryota Takahashi, Keisuke Yamamoto, Yoshinari Asaoka, Keisuke Tateishi, Yousuke Nakai, Hiroyuki Isayama, Harold L. Moses, Kazuhiko Koike. A role of bone morphogenetic protein signaling in pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Innovations in Research and Treatment; May 18-21, 2014; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2015;75(13 Suppl):Abstract nr A55.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2015
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2921-2921
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2921-2921
    Kurzfassung: Background: Genomic instability is a hallmark of cancer, which may lead to heterogeneity and provide an advantage on tumor survival and proliferation. However, we previously found that a greater number of genomic mutations were associated with reduced tolerance to radiation. The aim of this study is to elucidate a role of genomic instability on cellular radiosensitivity. Materials and Methods: We used 59 cancer cell lines derived from various types of solid tumors. Radiation-induced cell death was measured by the standard colony-forming assay at the dose range of 0 to 10 Gy. Based on the resulting survival curves, the mean inactivation dose (MID) was calculated as a single value representing cellular radiosensitivity. Somatic gene aberration data for the 59 cell lines were obtained from the Catalogue Of Somatic Mutations In Cancer cell line project v.80. The genomic analysis focused on cancer-related pathways defined by Kyoto Encyclopedia Genes and Genomes. The genomic mutations included nucleotide variations (substitutions, insertions, or deletions) and copy number variations (CNV). Only pathogenic mutations were selected and neutral mutations were disregarded according to the functional analysis through hidden Markov model. Both gain and loss in CNVs were included. Results: There were either nucleotide variations or copy number variations in 598 genes in the 59 cancer cell lines. The mutations were found in 16 caretakers, 51 gatekeepers, 105 oncogenes, and 449 passengers. There were significant positive correlations between the MIDs and the proportions of mutations in the gatekeepers or the oncogenes to the whole mutations. Namely, radioresistant cell lines harbored gatekeeper mutations or oncogene mutations at higher proportions. Conversely, radiosensitive cell lines showed significantly higher proportions of passenger mutations. No correlation was found in between the MIDs and the proportions of caretaker mutations. Conclusions: Genomic instability caused by caretaker mutations may result in not only increased tolerance to radiation through mutations in gatekeeper genes and oncogenes but also reduced tolerance to radiation through accumulating passenger mutations. A balance between the two opposing phenomena is a significant determinant of cancer cell radiosensitivity. Citation Format: Yuji Seo, Keisuke Tamari, Yutaka Takahashi, Kazumasa Minami, Keisuke Otani, Fumiaki Isohashi, Kazuhiko Ogawa. A dual role of genomic instability on radiosensitivity in cancer cells [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 2921.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 12, No. 12_Supplement ( 2014-12-01), p. B10-B10
    Kurzfassung: Pancreatic ductal adenocarcinoma (PDAC) is the most deadly cancer worldwide. Although many regimens have been tried against PDAC, an epidermal growth factor receptor (EGFR) inhibitor erlotinib in combination with gemcitabine is the only molecular target drug superior to gemcitabine alone. However, the mechanism by which PDAC with extremely frequent KRAS-mutation benefits from EGFR inhibition remains largely unknown. In this study, we evaluated the efficacy of erlotinib in combination with gemcitabine using a murine PDAC model with transforming growth factor-beta receptor II knockout plus Kras activation and investigated the mode of action. The mice were treated using the following drug doses and treatment schedules; erlotinib was administered from 3 weeks of age and gemcitabine was administered from 4 weeks of age. We isolated PDAC cells from the murine PDAC tissues. Effects of erlotinib on the proliferation and intracellular signaling of the murine PDAC cells or human PDAC cell lines were examined in vitro. We sacrificed the mice at 7 weeks of age, and excised the pancreatic tissues and processed for western blot analysis and immunohistochemistry. We evaluated the expression of EGFR ligands by real-time PCR and the heterodimer formation of EGFR with ErbB2 by immunoprecipitaiton after incubation with gemcitabine in vitro. We assessed whether the effect of gemcitabine on EGFR/ErbB2 activation is secondary to mitogen activated protein kinase (MAPK) signal activation after incubation with or without MEK inhibitor and gemcitabine by western blot analysis and real-time PCR. Gemcitabine + erlotinib inhibited PDAC progression and significantly prolonged the survival of the PDAC mice compared to gemcitabine alone. Gemcitabine or erlotinib also inhibited in vitro PDAC cell proliferation. Interestingly, Gemcitabine induced MAPK signaling, which was dramatically inhibited by adding erlotinib, even in the Kras-mutant PDAC cells. The suggested mechanisms were that gemcitabine induced EGFR ligand expression and also ErbB2 activation by increasing heterodimer formation with EGFR and maintaining high ErbB2 protein level in PDAC cells. We observed that the gemcitabine-induced MAPK signaling activation was in part due to induction of Egfr ligands(Egf, Tgf-a, Amphiregulin) expression by real-time PCR and ELISA. Using a phospho-RTK antibody array, we also observed that Gem induced Erbb2 activation in PDAC cells, and validated by western blot analysis, real-time PCR, and immunohistochemistry. Erlotinib inhibited the ErbB2 activation, partly by inhibiting heterodimer formation with EGFR and also decreasing ErbB2 protein expression in PDAC cells. We observed that gemcitabine-induced EGFR ligands up-regulation and EGFR/ErbB2 activation require intact MAPK signaling and these are secondary effects of MAPK signal activation and that gemcitabine induced the activation irrespective of KRAS status and gemcitabine sensitivity. This model helps us to evaluate an efficacy of new drugs and to investigate mechanisms of the mode of action and chemoresistance. This study provides clinical insights into potent therapeutic strategies for this difficult cancer. Citation Format: Koji Miyabayashi, Hideaki Ijichi, Ryota Takahashi, Dai Mohri, Keisuke Yamamoto, Yoshinari Asaoka, Tsuneo Ikenoue, Keisuke Tateishi, Harold L. Moses, Kazuhiko Koike. Epidermal growth factor receptor inhibitor prolongs survival in pancreatic cancer by blocking gemcitabine-induced mitogen-activated protein kinase signal. [abstract]. In: Proceedings of the AACR Special Conference on RAS Oncogenes: From Biology to Therapy; Feb 24-27, 2014; Lake Buena Vista, FL. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(12 Suppl):Abstract nr B10. doi: 10.1158/1557-3125.RASONC14-B10
    Materialart: Online-Ressource
    ISSN: 1541-7786 , 1557-3125
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2014
    ZDB Id: 2097884-4
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 7 ( 2021-04-01), p. 1654-1666
    Kurzfassung: Overcoming drug resistance is one of the biggest challenges in cancer chemotherapy. In this study, we examine whether targeting the long noncoding RNA taurine upregulated gene 1 (TUG1) could be an effective therapeutic approach to overcome drug resistance in pancreatic ductal adenocarcinoma (PDAC). TUG1 was expressed at significantly higher levels across 197 PDAC tissues compared with normal pancreatic tissues. Overall survival of patients with PDAC who had undergone 5-FU–based chemotherapy was shorter in high TUG1 group than in low TUG1 group. Mechanistically, TUG1 antagonized miR-376b-3p and upregulated dihydropyrimidine dehydrogenase (DPD). TUG1 depletion induced susceptibility to 5-FU in BxPC-3 and PK-9 pancreatic cell lines. Consistently, the cellular concentration of 5-FU was significantly higher under TUG1-depleted conditions. In PDAC xenograft models, intravenous treatment with a cancer-specific drug delivery system (TUG1-DDS) and 5-FU significantly suppressed PDAC tumor growth compared with 5-FU treatment alone. This novel approach using TUG1-DDS in combination with 5-FU may serve as an effective therapeutic option to attenuate DPD activity and meet appropriate 5-FU dosage requirements in targeted PDAC cells, which can reduce the systemic adverse effects of chemotherapy. Significance: Targeting TUG1 coupled with a cancer-specific drug delivery system effectively modulates 5-FU catabolism in TUG1-overexpressing PDAC cells, thus contributing to a new combinatorial strategy for cancer treatment.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 20, No. 9 ( 2021-09-01), p. 1713-1722
    Kurzfassung: Cholangiocarcinoma is a highly malignant cancer. Many patients need systemic chemotherapy to prevent tumor development and recurrence; however, their prognosis is poor due to the lack of effective therapy. Therefore, a new treatment option is urgently required. We recently identified glypican-1 (GPC1) as a novel cancer antigen of esophageal squamous cell carcinoma. We also demonstrated the efficacy and safety of GPC1-targeted ADC (GPC1–ADC) conjugating anti-GPC1 mAb possessing high internalization activity with monomethyl auristatin F (MMAF), which is a potent tubulin polymerizing inhibitor. In this study, we confirmed that GPC1 was highly expressed in cholangiocarcinoma cells and tissues. IHC analysis of 49 extrahepatic cholangiocarcinoma patient tumor specimens revealed high expression of GPC1 in 47% of patients. These patients demonstrated significantly poorer prognosis compared with the low-expression group in terms of disease-free survival and overall survival (P & lt; 0.05). GPC1 was also expressed in tumor vessels of cholangiocarcinoma, but not on the vessels of nontumor tissues. MMAF-conjugated GPC1–ADC showed potent tumor growth inhibition against GPC1-positive cholangiocarcinoma cells in vitro and in vivo. In a GPC1 knockout xenograft model, GPC1–ADC partially inhibited tumor growth. Vascular endothelial cells in tumor tissues of GPC1-negative xenograft mice expressed GPC1 and were arrested in the G2–M phase of cell cycle by GPC1–ADC. GPC1–ADC exhibits direct as well as indirect antitumor effects via inhibition of tumor angiogenesis. Our preclinical data highlight GPC1–ADC as a promising therapy for GPC1-positive cholangiocarcinoma.
    Materialart: Online-Ressource
    ISSN: 1535-7163 , 1538-8514
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2062135-8
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5487-5487
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5487-5487
    Kurzfassung: Prostaglandin E2 (PGE2) plays an important role in cancer cell growth, progression, and metastasis and exerts its effects by binding to E-prostanoid receptor (EP). In the previous study, we showed that soluble fragment of EP2 receptor containing the second extracellular region (FuEP2/Ex2) has neutralizing activity to PGE2 and inhibits the growth of prostate and endometrial cancer cells. Here, we established a stable transfectant expressing FuEP2/Ex2 from human ovarian cancer SKOV/ip cells (SKOV/ip-FuEP2/Ex2) and SKOV/ip-FuEP2/Ex2 and vector-control cells were injected into nude mice intraperitoneally. Tumor weight and ascites volume in SKOV/ip-FuEP2/Ex2-injected mice decreased significantly compared to control cell-injected mice. Furthermore, microvessel density and numbers of M2-poralized macrophage in tumor lesion were significantly decreased in SKOV/ip-FuEP2/Ex2-injected mice. By using RNAs from tumor lesion in each group, the cDNA microarray analysis was also conducted. Six of upregulated genes and eight of downregulated genes in SKOV/ip-FuEP2/Ex2-derived tumor were identified. In upregulated genes, MMP-7, TMPRSS4, and CYP1B1, which is involved in cancer progression and metastasis, were included. Treatment with AEBSF, an inhibitor of TMPRSS4, or siRNA for TMPRSS4 further reduced cell growth of SKOV/ip-FuEP2/Ex2 cells in vitro and in vivo. Our results suggest that FuEP2/Ex2 has a suppressive effect in peritoneal metastasis model of ovarian cancer and that MMP-7, TMPRSS4, and CYP1B1 may act as a survival factor under the condition in which EP-mediated signaling are inhibited. Moreover, targeting of TMPRSS4 may enhance the inhibitory effect of FuEP2/Ex2 on ovarian cancer metastasis. Citation Format: Tetsuyuki Takahashi, Hisanori Uehara, Keisuke Izumi. Inhibitory effect of soluble EP2 receptor on ovarian tumor growth in nude mice and usefulness of TMPRSS4 as a molecular target for synergistic efficacy. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5487. doi:10.1158/1538-7445.AM2013-5487
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2013
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...