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
Filter
  • American Association for Cancer Research (AACR)  (5)
  • Bang, Ju-Hee  (5)
Material
Publisher
  • American Association for Cancer Research (AACR)  (5)
Language
Years
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2024
    In:  Cancer Research Vol. 84, No. 6_Supplement ( 2024-03-22), p. 380-380
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 84, No. 6_Supplement ( 2024-03-22), p. 380-380
    Abstract: Background: Targeting endoplasmic reticulum (ER) stress is a potential therapeutic strategy in preclinical models of pancreatic duct adenocarcinoma (PDAC). GRP78 is known as the master regulator of the unfolded protein response (UPR) pathway, regulating ER stress by activating the UPR pathway to avoid cell apoptosis and regulates the activation of UPR pathway signals by binding PERK. Overexpression of GRP78 in PDAC results in the retention of misfolded and unassembled proteins, as well as the accumulation of ROS, leading to the promotion of genomic instability. However, there has been limited research on targeting GRP78 as a therapeutic strategy in PDAC. In this study, we aimed to investigate the anticancer effects of BOLD-100, an inhibitor of GRP78, in PDAC. Methods: We used 9 PDAC cell lines (Capan-1, AsPC-1, HPAFII, MIA-PaCa2, Panc-1, Capan-2, SNU-213, SNU-324, SNU-2918). The mouse xenograft model of Capan-1 was established for in vivo study. BOLD-100 (GRP78 inhibitor), AZD6738 (ATR inhibitor), NAC (N-Acetylcysteine; ROS scavenger) and TUDCA (Tauroursodeoxycholic acid; ER stress inhibitor) were used. MTT assay, CFA assay, cell cycle analysis, RT-PCR, western blot, immunoprecipitation, DCF-DA staining and Annexin V assay were used to elucidate the action of BOLD-100. The combination of BOLD-100 with AZD6738 has been evaluated in both in vitro and in vivo model. Results: BOLD-100 suppressed the proliferation of PDAC cells and decreased the mRNA level of GRP78, which in turn disrupted the interaction between GRP78 and PERK. BOLD-100 increased ER stress and ROS, leading to activation of PERK, eIF2a, and CHOP. Activation of UPR pathway induced CHOP-dependent apoptosis and inhibited PDAC cell growth. Moreover, TUDCA reversed the ROS accumulation induced by BOLD-100, confirming that ER stress regulates ROS levels. The accumulation of ROS upregulated the active forms of ATR and CHK1, suggesting the activation of the DNA damage repair pathway. Notably, the treatment of NAC abrogated the activation of ATR-CHK1 axis by BOLD-100-induced ROS accumulation. AZD6738 synergized with BOLD-100 in in vitro and in vivo, by suppressing BOLD-100-induced ATR phosphorylation. Conclusion: GRP78 could serve as one of the potential therapeutic targets in PDAC. The combination of BOLD-100 and AZD6738 demonstrates a synergistic effect suggesting GRP78/ATR dual targeting as a promising therapeutic option for patients with PDAC. Citation Format: Su In Lee, Ah-Rong Nam, Kyoung-Seok Oh, Jae-Min Kim, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Hyo Jung Kim, Jeesun Yoon, Tae-Yong Kim, Do-Youn Oh. Co-downregulation of GRP78 and ATR enhances apoptosis in pancreatic ductal adenocarcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 380.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2024
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4496-4496
    Abstract: Background: PARP inhibitors have shown antitumor activities against solid tumors with HRD (homologous recombination deficiency). The definition of HRD and other potential biomarkers besides HRD should be further evaluated for PARP inhibitors. JPI-547 is a novel PARP inhibitor, simultaneously targeting tankyrase1/2, other members of the PARP family, that are involved in the Wnt/β-catenin pathway. Method: Antiproliferative effect of JPI-547 and a variety of PARP inhibitors (olaparib, veliparib, talazoparib, niraparib, and rucaparib) were determined by MTT assay or clonogenic assay in 9 human pancreatic ductal adenocarcinoma (PDAC) cell lines (Capan-1, HPAF-II, Capan-2, AsPC-1, SNU-410, SNU-213, SNU-324, MIA-PaCa2, and PANC-1). Transcriptome data and gene dependency score of the cell lines were obtained from the CCLE database and DepMap respectively. DNA damage was monitored by immunofluorescent imaging of γ-H2AX and DR-GFP assay determined the homologous recombination repair (HRR) efficiency. A xenograft tumor model was established to substantiate the in vivo antitumor effect of JPI-547. Results: JPI-547 more potently blocks Poly(ADP-ribosyl)ation than olaparib, and induces a strong antiproliferative effect on Capan-1, a cell line with BRCA2 del. JPI-547 leads to cell cycle arrest and induces enhanced apoptotic cell death than olaparib. JPI-547 inhibits tumor growth of Capan-1 in vivo, suggesting the potent antitumor activity of JPI-547 against PDAC with HRD. Cell lines harboring RNF43 LOF mutations (HPAF-II, AsPC-1, and Capan-2), intrinsically addicted to Wnt/β-catenin pathway, are more sensitive to JPI-547 than cells with RNF43 wild types. Interestingly, RNF43 mutations could not distinguish the sensitivity of other PARP inhibitors except JPI-547. CTNNB1 gene dependency score and β-catenin levels positively correlate with cellular sensitivity to JPI-547. JPI-547-induced DNA damage was alleviated in HR-proficient PDAC cells. DR-GFP assays confirm that JPI-547 does not directly alter the HRR efficiency of Wnt-addicted PDAC cells. Collectively, these data indicate that the vulnerabilities of Wnt-addicted PDAC cells to JPI-547 were irrelevant to HRD mimicking. Rather, JPI-547 stabilizes AXIN-2 in Wnt-addicted PDAC cells and downregulates the active form of β-catenin level in the nucleus, thereby disrupting the transcription of its target genes. Knockdown of β-catenin neutralized the antiproliferative effect of JPI-547, suggesting that inhibition of the β-catenin pathway is an important mode of action by JPI-547 in Wnt-addicted PDAC cells. Conclusion: JPI-547 shows promising, preclinical antitumor effects against PDAC cells with HRD or Wnt-addiction, providing a rationale for further biomarker-driven clinical development of JPI-547 for the treatment of patients with PDAC. Citation Format: Kyoung-Seok Oh, Ah-Rong Nam, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Hyo Jung Kim, Su In Lee, Jae-Min Kim, Jeesun Yoon, Tae-Yong Kim, Banyoon Cheon, Hyunju Cha, John Kim, Do-Youn Oh. JPI-547, a dual inhibitor of PARP/Tankyrase, shows promising antitumor activity against pancreatic cancers with homologous recombination repair deficiency or Wnt-addiction. [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 4496.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2024
    In:  Cancer Research Vol. 84, No. 6_Supplement ( 2024-03-22), p. 5711-5711
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 84, No. 6_Supplement ( 2024-03-22), p. 5711-5711
    Abstract: Background: Targeting CDKs has emerged as a significant strategy in the development of new drugs. The CDK4/6 inhibitors have proven their efficacy in several tumor types, while the CDK2 and CDK9 inhibitors are currently undergoing clinical trials. In biliary tract cancer (BTC), gene expressions of CDK2 and CDK9 are elevated compared to normal tissue. CDK9, a transcriptional CDK, regulates RNA polymerase II (RNAP II), leading to enhanced transcription of oncogenes, such as MCL1. Aberrant activation of these CDKs is associated with cancer progression and evading apoptosis in BTC. Notably, MCL1 is frequently amplified in intrahepatic cholangiocarcinoma (16-21%). This finding underscores the potential significance of CDK2 and CDK9 as therapeutic targets in BTC. However, in BTC, targeting CDK2 or CDK9 has not yet been studied. In this study, we aimed to test CDK2/9 targeting and develop possible biomarker strategies in BTC. Method: A total 9 BTC cell lines (SNU245, SNU308, SNU478, SNU869, SNU1196, SNU2670, SNU2773, TFK1, and HUCCT1) were used. Fadraciclib (CDK2/9 dual inhibitor), Olaparib (PARP inhibitor), JQ1 (BRD4 inhibitor) was used. MTT assay, Colony formation assay, Annexin-V assay, and Cell cycle analysis were performed for evaluation of anti-cancer effects. The effect of Fadraciclib on homologous recombination (HR)-mediated DNA damage repair was assessed using focus formation assay and DRGFP assay. Combination of Fadraciclib with Olaparib or JQ1 was evaluated in vitro and in vivo. Result: The anti-tumor effect of Fadraciclib monotherapy was relatively higher in MCL1-High BTC cells (SNU869, SNU2773) compared to MCL-Low cells (SNU245, SNU2670). In MCL1-High cells, Fadraciclib inhibited CDK9, leading to downregulation of RNAP II phosphorylation at Serine 2 and MCL1. Fadraciclib also interfered with HR-mediated DNA damage response by inhibiting transcription of HR-related genes. The combination of Fadraciclib and Olaparib exhibited synergistic anti-tumor effects in vitro in MCL1-High cells and in vivo xenograft model. Conversely, in MCL1-Low cells, Fadraciclib increased BRD4 mRNA expression, leading to the restoration of CDK9-RNAP II activity. This reactivation of RNAP II restored cellular transcription homeostasis, thereby reactivating the transcription of oncogenes such as MCL1. Combination of Fadraciclib and JQ1 showed synergistic effects in vitro in MCL1-Low cells and in vivo xenograft model by inhibiting restored CDK9-RNAP II activity. Conclusion: MCL1-High BTC cells are sensitive to CDK2/9 inhibitor and show synergism between CDK2/9 inhibitor and PARP inhibitor. In MCL1-Low BTC, a combination of a CDK2/9 inhibitor and a BRD4 inhibitor might represent an optimal strategy for new drug development. Citation Format: Jae-Min Kim, Ah-Rong Nam, Kyoung-Seok Oh, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Su In Lee, Hyo Jung Kim, Jeesun Yoon, Tae-Yong Kim, Do-Youn Oh. Anti-tumor effects of fadraciclib, CDK2/9 inhibitor, in biliary tract cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 5711.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2024
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 84, No. 6_Supplement ( 2024-03-22), p. 2599-2599
    Abstract: Background: The combination approach of antibody-drug conjugates and immune checkpoint inhibitors (ICIs) is being tested to enhance efficacy and overcome resistance in solid tumors. Although trastuzumab deruxtecan (T-DXd) is approved in HER2-positive gastric cancer (GC), its impact on the GC immune microenvironment remains elusive. This study explores how T-DXd modulates the immune response, providing essential insights on the development of T-DXd and immunotherapy combinations. Methods: Comet assays and immunofluorescence assessed DNA damage. RNA-seq delineated the impact of T-DXd on GC cells (NCI-N87 and SNU-484). In vivo complex of enzyme assays and immunostaining of topoisomerase 1-DNA covalent cleavage complex (TOP1cc) were used to determine the action mechanism of T-DXd. RT-qPCR, and ELISA measured cGAS-STING cytosolic dsDNA sensing pathway activation. Further validation included cGAS knockdown and 2’3’-cGAMP addition to confirm the role of cGAS in T-DXd-induced promotion of inflammatory response. In coculture experiments, we utilized THP-1-derived dendritic cells (TDDCs) and PBMCs from healthy donors to prove the effect of T-DXd on immune cells. FACS analyses probed the activation markers of TDDCs and CD8+ T cells. Results: T-DXd-induced DNA damage upregulated PD-L1 expression in GC cells in an IRF1-dependent manner. RNA-seq revealed gene signatures associated with increased antitumor immune response, including proinflammatory response and type-I IFN, following T-DXd treatment. Mechanistically, T-DXd co-localized TOP1cc with γ-H2AX-positive micronuclei, activating the cGAS-STING pathway and triggering a robust IFN-I response. Knockdown of cGAS in cancer cells abolished the T-DXd-induced IFN-I response. T-DXd enhanced HLA-DR and CD86 expression in TDDCs, particularly when cocultured with GC cells. The physical interaction between cancer cells and TDDCs, along with the presence of cGAS in cancer cells, was found to be essential for the upregulation of dendritic cell (DC) activation marker by T-DXd. Furthermore, T-DXd elevated CD69, Granzyme B, and IFNG expression in CD8+ T cells exclusively under coculture setting with NCI-N87. Conclusion: T-DXd exerts immunostimulatory effects in GC cells through the activation of the cGAS-STING pathway, leading to a reprogramming of DC and CD8+ T cell phenotypes. This provides a molecular rationale for combining T-DXd with ICIs for the treatment of patients with HER2-positive tumors. Citation Format: Kyoung-Seok Oh, Ah-Rong Nam, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Hyo Jung Kim, Su In Lee, Jae-Min Kim, Jeesun Yoon, Tae-Yong Kim, Do-Youn Oh. Trastuzumab deruxtecan reprograms immune response by stimulating cGAS-STING pathway in gastric cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 2599.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2024
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3872-3872
    Abstract: Background: HER2 (Human epithelial growth factor receptor 2)-targeting therapies have been approved for patients with HER2-positive breast and gastric cancer and use have been attempted in various solid tumor types, including biliary tract cancer. However, resistance mechanism remains as a major challenge of HER2-targeting therapies. YAP (Yes-associated protein) is a major downstream effector of Hippo pathway, and it plays an essential role in cancer cell proliferation, survival and differentiation. Moreover, YAP is emerging as a key player of resistance mechanism of cytotoxic and targeted drugs. Yap is also an important immunosuppressive molecule as it works as a negative regulator of T cell tumor infiltration. In this study, we intend to elucidate the role of YAP in mechanism of trastuzumab resistance and T cell immune response in HER2-positive cancer cells. Methods: We established four trastuzumab-resistant (HR) cell lines (N87HR, SNU216HR, SNU2670HR and SNU2773HR) from HER2-postive gastric and biliary tract cancer cell lines. To inhibit the function of YAP, siRNA and Verteporfin (YAP-TEAD inhibitor) were used. MTT assay, cell cycle analysis, western blot and migration assay were performed to analyze the antitumor effects through YAP downregulation. In order to evaluate immune-modulation by YAP, human PBMC co-culture was used and immune markers were analyzed by RT-PCR and flow cytometry. Mouse xenograft models were established using SNU2773 and SNU2773HR cells. Results: We confirmed that the expressions of pYAP and YAP were elevated in HR cells (SNU216HR, SNU2670HR and SNU2773HR) compared to parental cells. Reducing the expressed YAP in HR cells inhibited tumor cell growth and migration and induced apoptosis. Immune suppression markers such as PD-L1, CD155, and galectin-9 were effectively decreased, while CD80, a stimulation marker, was increased by verteporfin treatment. Also, when YAP was decreased, CCL5 and CXCL10, well known CD8+ T cell recruitment cytokines, were increased. In HR cells treated with siYAP and verteporfin, there was a trend of increasing CD4+ and CD8+ T cells when co-culture with PBMC. In vivo experiment data showed greater tumor growth inhibition effects with SNU2773HR than SNU2773 xenograft models when treated with verteporfin. Conclusion: The expression of YAP is elevated in trastuzumab-resistant (HR) cells and inhibition of YAP shows anti-tumor effects and activation of T cell responses. Collectively, our data suggests that the inhibition of YAP is one of many promising strategies to overcome trastuzumab resistance and T cell regulatory mechanisms in HER2-positive cancers. Citation Format: Ah Rong Nam, Jeesun Yoon, Kyoung-Seok Oh, Jae-Min Kim, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Hyo Jung Kim, Su In Lee, Tae-Yong Kim, Do-Youn Oh. Targeting of YAP overcomes trastuzumab-resistance and promotes immune responses in HER2-positive cancers. [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 3872.
    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
    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...