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)  (17)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. B043-B043
    Abstract: Immune checkpoint blockade therapy targeting PD-L1/PD-1 have been widely used and shown remarkable clinical response in various cancers. However, therapeutic resistance after initial response is increasingly observed and the detailed mechanisms of resistance have yet to be fully understood. Recently, several mechanisms including loss-of-function alteration in genes related to antigen presentation and interferon-receptor signaling have been suggested to induce the resistance to PD-1 blockade treatment, yet few studies have focused on anti-PD-L1 (aPD-L1) blockade therapy. In current study, we performed whole exome sequencing and RNA-sequencing analysis in two non-small cell lung cancer (NSCLC) patients who were refractory during aPD-L1 therapy. We have identified the mutations in RNA splicing related gene, TARDBP/TDP-43, which were uncovered as hot spot mutations in Amyotrophic lateral sclerosis (ALS), and found two unique secreted PD-L1 variants, which lacked the transmembrane domain by aberrant RNA splicing. These secreted PD-L1 variants were shown to be stable by pulse chase assay, and worked as “decoys” of aPD-L1 antibody in the HLA-matched coculture system of iPSC-derived CD8 T cells and cancer cells. Moreover, expression of secreted mPD-L1 variant in mouse MC38 cancer cells conferred the resistance to PD-L1 blockade therapy, and soluble PD-L1 were time dependently accumulated in plasma using the MC38 syngeneic mice model. To further investigate the presence of secreted PD-L1 splicing variants in patients, we additionally analyzed 15 specimen who were resistant to anti-PD-L1 treatment by RNA-seq. We revealed that approximately 20% of therapeutic resistant patients harbored secreted PD-L1 variants and several new mutations in JAK1/2, the key mediator of interferon-receptor signaling, were found in a part of patients as well. Furthermore, we also identified the presence of secreted PD-L1 variants in the surgical specimens of squamous NSCLC patients without prior chemotherapy, targeted therapy, and /or immune-checkpoint inhibitor therapy. Consistent to the data, the plasma level of soluble PD-L1 in patients with secreted PD-L1 variants were high than those without the variants. Collectively, our results elucidated a novel resistant mechanism of PD-L1 blockade antibody mediated by secreted PD-L1 variants. The presence of sPD-L1 splicing variants or the level of soluble PD-L1 in plasma or pleural effusion may work as a biomarker to predict a patient’s response to PD-L1 blockade therapy. Citation Format: Bo Gong, Kazuma Kiyotani, Seiji Sakata, Ken Takahashi, Seiji Nagano, Shun Kumehara, Satoko Baba, Benjamin Besse, Noriko Yanagitani, Luc Friboulet, Makoto Nishio, Kengo Takeuchi, Hiroshi Kawamoto, Naoya Fujita, Ryohei Katayama. Identification of secreted PD-L1 variants as a decoy of PD-L1 blockade antibody mediating the therapeutic resistance [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B043. doi:10.1158/1535-7163.TARG-19-B043
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5347-5347
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5347-5347
    Abstract: REV7 is involved in DNA repair, cell cycle regulation, gene expression and carcinogenesis. In vitro studies show that REV7 interacts with several proteins and regulates their function, and that downregulation of REV7 expression in human cancer cells results in enhancement of sensitivity to DNA damaging agents. It has been reported that human REV7 is highly expressed in the adult testis by northern blot analysis. However, the significance of REV7 in mammalian development has not been elucidated. Here, we present analyses of Rev7-deficient (Rev7-/-) mice to clarify the significance of Rev7 in mouse development. In wild-type mice (Rev7+/+), Rev7 expression was ubiquitously observed in the embryo and confined to germ cells in the testes after birth. Rev7-/- mice exhibited growth retardation and a partial embryonic lethal phenotype. Mice that survived to adulthood were infertile in both sexes and showed germ cell aplasia in the testes and ovaries. Analyses of Rev7-/- embryos revealed that primordial germ cells (PGCs) were present at embryonic day 8.5 (E8.5). However, progressive loss of PGCs was observed during migration, and PGCs were absent in the genital ridges at E13.5. An increase of apoptotic cells was detected not only among PGCs but also in the forebrain of the Rev7-/- embryo, while cell proliferation was unaffected. Gene expression analysis of E8.5 embryos showed upregulation of Oct4 and Nanog and downregulation of c-Kit and Rev3 in Rev7-/- embryos compared with those in Rev7+/- embryos. These findings indicate that Rev7 is essential for PGC maintenance by prevention of apoptotic cell death in the mouse. Note: This abstract was not presented at the meeting. Citation Format: Yoshiki Murakumo, Naoki Watanabe, Shinji Mii, Masato Asai, Naoya Asai, Kaoru Niimi, Takuya Kato, Atsushi Enomoto, Masahide Takahashi. DNA repair protein Rev7 is required for primordial germ cell maintenance in the mouse. [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 5347. doi:10.1158/1538-7445.AM2014-5347
    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
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4250-4250
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4250-4250
    Abstract: CD109, a glycosylphosphatidylinositol (GPI)-anchored glycoprotein, is a member of the β2-macroglobulin/C3, C4, C5 family of thioester-containing proteins. It has been reported that CD109 is highly expressed in various tumor cell lines and in tumor tissues including squamous cell carcinomas (SCCs) of the lung, oral cavity, esophagus and uterine cervix, malignant melanoma of the skin, and urothelial carcinoma of the urinary bladder. CD109 is, therefore, thought to be a cancer-related gene. On the other hand, CD109 is a component of TGF-β receptor system and negatively regulates TGF-β signaling. However, its physiological function in vivo remains largely unknown. To investigate the physiological function of CD109 in vivo, we generated CD109-deficient (CD109-/-) mice in which exon 1 and 2 of CD109 genomic locus were replaced with the lacZ gene. CD109 protein is expressed in the skin, tongue, and testis of adult CD109+/+ mice, but not in CD109-/- mice. Macroscopically, CD109-/- mice showed hair growth impairment from postnatal day 7 (P7) and persisted until P28. Microscopically, hyperplasia of the epidermis and sebaceous glands became apparent at P7 in CD109-/- mice and sustained until P70, and the hair follicles of CD109-/- mice exhibited ectasia with kinked hair shafts at P14. The epidermis of CD109-/- mice showed apparent thickening of the basal / suprabasal layers, which are positive for CK14, compared with CD109+/+ mice, whereas no significant differences in Ki-67 or cleaved caspase-3-positive ratios were detected between the two groups. These results suggest that CD109 regulates the differentiation of keratinocytes in vivo, and impairment of hair growth and ectatic hair follicles may be secondary changes caused by narrowing of the infundibular portion of the hair follicles due to epithelial hyperplasia. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4250. doi:1538-7445.AM2012-4250
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 20 ( 2019-10-15), p. 5367-5381
    Abstract: Cancer-associated fibroblasts (CAF) constitute a major component of the tumor microenvironment. Recent observations in genetically engineered mouse models and clinical studies have suggested that there may exist at least two functionally different populations of CAFs, that is, cancer-promoting CAFs (pCAF) and cancer-restraining CAFs (rCAF). Although various pCAF markers have been identified, the identity of rCAFs remains unknown because of the lack of rCAF-specific marker(s). In this study, we found that Meflin, a glycosylphosphatidylinositol-anchored protein that is a marker of mesenchymal stromal/stem cells and maintains their undifferentiated state, is expressed by pancreatic stellate cells that are a source of CAFs in pancreatic ductal adenocarcinoma (PDAC). In situ hybridization analysis of 71 human PDAC tissues revealed that the infiltration of Meflin-positive CAFs correlated with favorable patient outcome. Consistent herewith, Meflin deficiency led to significant tumor progression with poorly differentiated histology in a PDAC mouse model. Similarly, genetic ablation of Meflin-positive CAFs resulted in poor differentiation of tumors in a syngeneic transplantation model. Conversely, delivery of a Meflin-expressing lentivirus into the tumor stroma or overexpression of Meflin in CAFs suppressed the growth of xenograft tumors. Lineage tracing revealed that Meflin-positive cells gave rise to α-smooth muscle actin-positive CAFs that are positive or negative for Meflin, suggesting a mechanism for generating CAF heterogeneity. Meflin deficiency or low expression resulted in straightened stromal collagen fibers, which represent a signature for aggressive tumors, in mouse or human PDAC tissues, respectively. Together, the data suggest that Meflin is a marker of rCAFs that suppress PDAC progression. Significance: Meflin marks and functionally contributes to a subset of cancer-associated fibroblasts that exert antitumoral effects.
    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
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 135-135
    Abstract: (a) Purpose: Gangliosides are molecules composed of glycosphingolipids linked with one or more sialic acids, and are known to be key components of lipid rafts, which act as platforms for signal transduction. Changes in ganglioside levels affect the expression of raft-associated proteins on the cell surface and lead to reduced membrane fluidity, thus, resulting in cellular dysfunction, such as impaired signal transduction. The ganglioside GM2 is one of the major series of gangliosides that has several biological functions, such as in cell adhesion and signal transduction. GM2 is highly expressed in several types of human malignant tumors, such as melanomas, gliomas, and neuroblastomas, but is absent or weakly expressed in normal tissues. However, the association between gangliosides and pancreatic ductal adenocarcinoma (PDAC) has not been elucidated yet. Thus, in this study, we aimed to clarify the expression and role of ganglioside GM2 in PDAC. (b) Experimental procedures: GM2 expression in eight human PDAC cell lines was examined by fluorescence-activated cell sorter (FACS) analysis. The morphology, growth, and stemness of GM2- and GM2+ sorted-cells were compared by transmission electron microscopy, growth assays, real-time PCR analysis of the markers of stemness, and sphere forming assays. Cell motility was evaluated by invasion assay. For comparing tumorigenicity between the GM2- and GM2+ sorted-cells, heterotopic implantations were performed. The expression levels and clinico-pathological roles of GM2 in patients with PDAC were examined by immunohistochemical analysis of 117 pancreatic tissue samples. (c) Results: The fraction of GM2+ cells was the highest (21.4%) in MIA PaCa-2 from among the eight cell lines, as revealed by FACS analysis. GM2-expressing MIA PaCa-2 cells had higher growth rates under adherent growth conditions. In 3D-culture, which enriches cancer stem cells (CSCs), most MIA PaCa-2 cells expressed GM2 and the cells responded to TGF-β1 treatment to promote invasion. Transplantation of the GM2-expressing cells into nude mice resulted in development of larger tumors as compared to that for control cells that did not express GM2. In the PDAC cases, GM2 expression was significantly associated with younger age, larger tumor size, advanced stage, and higher histological grade. (d) Conclusions: We show that a PDAC cell line overexpressing GM2 exhibits high growth rate and high tumor initiation in 2D-culture. Further, pancreatic CSC-like cells expressing GM2 exhibit responsiveness against TGF-β1, resulting in enhanced invasion in 3D-culture. Furthermore, GM2 expression is associated with the growth and advanced stage of human PDAC. Further studies will be required for the development of an early detection method for GM2-overexpressing pancreatic cancers and to develop novel therapeutic strategies targeting GM2. Citation Format: Norihiko Sasaki, Kenichi Hirabayashi, Masaki Michishita, Kimimasa Takahashi, Fumio Hasegawa, Fujiya Gomi, Yoko Itakura, Naoya Nakamura, Masashi Toyoda, Toshiyuki Ishiwata. Increased expression of ganglioside GM2 correlates with aggressiveness of human pancreatic ductal adenocarcinoma [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 135.
    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
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Cancer Research Vol. 69, No. 8 ( 2009-04-15), p. 3597-3604
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 69, No. 8 ( 2009-04-15), p. 3597-3604
    Abstract: Histone deacetylases (HDAC) are involved in carcinogenesis through their regulation of cell proliferation, differentiation, and survival. The inhibitors of HDAC exhibit profound synergistic effects in cancer treatment when combined with other anticancer drugs. However, the molecular mechanisms underlying this synergy are not fully understood. Here, we show that HDAC1 increases the resistance of cancer cells to oxidative stress by negatively regulating the expression of thioredoxin binding protein 2 (TBP-2). We found that the recruitment of HDAC1 to the TBP-2 promoter is mediated by a protein complex consisting of RET finger protein (RFP; also called TRIM27) and the trimeric transcription factor NF-Y. Accordingly, RNA interference–mediated depletion of RFP led to the disruption of the protein complex and a marked increase in the sensitivity of cancer cells to cisplatin, a potent inducer of oxidative stress. Furthermore, high levels of RFP expression correlated with down-regulation of TBP-2 in human colon cancers and were associated with poor clinical outcome. These findings reveal the diverse cancer-promoting activities of HDAC1 and identify RFP as a key regulator that provides cancer cells with resistance to anticancer drugs. [Cancer Res 2009;69(8):3597–604]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 322-322
    Abstract: Background;Recently, three-dimensional culture model has been reported as better tumor model than 2D culture. The importance of interaction between cancer and stromal cells has been widely recognized in tumor progression and resistance against treatment. So that ex vivo model which recaptures the tumor microenvironment is required to perform drug evaluation mimicking the patient tumor tissue. Methods; Layered 3D stromal tissues with microvascular network were produced by culturing normal fibroblasts and endothelial cells coated with the extra-cellular matrix (ECM) and natural polysaccharide, namely collagen and heparin. The layered 3D stromal tissues and co-cultured tumor were morphologically evaluated by HE staining, immunohistochemistry and immunofluorescence (IF). Drug sensitivity assay were conducted using popular colorectal cancer cell lines, and patient-derived cell lines (PDCs) established in JFCR under approved IRB. Remaining cancer cells post drug treatment were quantified by IF and imaging analysis. Furthermore, in vivo drug sensitivity was also evaluated. Results: The CD31 positive luminal structures which shows vascular endothelial tube formation were observed intra multi-layered tissues. The dense microvascular network tended to be formed nearby cancer cells. In comparison with 2D culture model, decreased drug sensitivities were represented in the layered 3D co-cultured model. The evaluated drug sensitivities in 3D model may reflect the response of cancer cells in in vivo. Conclusion; We developed the layered 3D stromal tissue culture system including blood micro-vessels. Our unique 3D ex vivo model appear to be a valuable tool for drug evaluation, and thus further studies are underway to confirm the usefulness of our model. Testing approved and/or developing compounds with PDCs in the model would potentially enable better prediction their efficacy. Citation Format: Yuki Takahashi, Kei Tsukamoto, Yuki Shimizu, Ryohei Katayama, Eiji Shinozaki, Satoshi Nagayama, Michiya Matsusaki, Shiro Kitano, Kensei Yamaguchi, Naoya Fujita. A unique ex vivo tumor model: 3D co-cultured system with cancer cells and stromal tissue for drug evaluation [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 322.
    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
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6027-6027
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6027-6027
    Abstract: Background: Multiple 3D culture models have been reported as a superior tumor model than 2D culture. The importance of interaction between cancer and stromal cells has been widely recognized in tumor progression and resistance to treatment. Therefore, ex vivo model recapturing the tumor microenvironment is needed to evaluate the drug efficacy under the condition imitating the patient tumor tissue. Purpose: We developed a unique 3D co-cultured tumor model with stromal tissues containing a microvascular network. Here, we investigated drug sensitivity in conventional 2D culture, our 3D co-cultured model and in vivo tumor. Methods: Drug sensitivity and gene expression on our model were evaluated using patient-derived cancer cells (PDC) established from colorectal cancer (CRC) patients in JFCR. The characteristics were compared with those of conventional 2D cultured cells or patient-derived xenograft (PDX). Results: In our 3D model, the drug sensitivities tended to be decreased in comparison with those of 2D. However, some drugs presented potent efficacy in our 3D model rather than 2D, and such drugs showed significant tumor growth suppression or tumor regression in vivo. Transcriptome profiles of our 3D model showed relatively higher similarity to those of in vivo tumors than those of 2D. Conclusion: Our study proposed the unique 3D co-cultured tumor model which may enable more accurate evaluation of drug sensitivities reflecting the in vivo circumstances. Citation Format: Yuki Takahashi, Shiro Kitano, Eiji Shinozaki, Satoshi Nagayama, Ryohei Katayama, Naoya Fujita. Layered 3D co-cultured tumor model including vascularized stromal tissue may reflect drug sensitivities in vivo tumor [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 6027.
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2837-2837
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2837-2837
    Abstract: Obesity-associated cancers, including colon cancer, are increasing in Western countries. It has been demonstrated that increased amounts of visceral fat and decreased levels of plasma adiponectin (APN) are associated with development of human colorectal cancer. Thus, we here investigated the function of APN in intestinal carcinogenesis. Male APN+/+, APN+/- or APN-/- C57BL/6J mice were injected with azoxymethane (AOM) once a week for 6 weeks, which led to the development of intestinal tumors. C57BL/6J mice with each genotype were sacrificed at the age of 55 weeks, and the number and size of colorectal tumors were examined by pathological study. Furthermore, these strains of mice were also crossed with Min mice (C57BL/6J background) to assess the effects of APN on intestinal polyp formation. At the age of 9 and 12 weeks, Min mice each with the APN genotype were sacrificed, and the number and size of intestinal polyps were examined by pathological study. Levels of adipocytokine and phosphorylation of AMPK were also evaluated to clarify the effect of APN on intestinal tumor development. The incidence of AOM-induced tumors was 40% of APN+/+, 50% of APN+/- and 71% (p & lt;0.05) of male APN-/- C57BL/6J mice, respectively. Multiplicities of AOM-induced tumors in each genotype were 0.5±0.7, 0.6±0.7 and 1.1±1.0 (p & lt;0.05), respectively. The total number of intestinal polyps developed in 9-week-old male APN+/- Min and APN-/-Min mice increased 2.4- and 3.2-fold compared with those in APN+/+Min mice, respectively, and was further enhanced at 12 weeks of age, being 3.2- and 3.4-fold, respectively. Studies with female mice gave similar results. Phosphorylation levels of AMPK in intestinal epithelial cells were decreased in APN-/- mice compared with APN+/+ mice. Among serum adipocytokines, levels of serum Pai-1 increased in APN-/- C57BL/6J mice and APN-/-Min mice with AOM injection. Activation of AMPK by metformin treatment suppressed Pai-1 expression in Min mice. AOM-induced colorectal aberrant crypt foci in APN-/- C57BL/6J mice were decreased by a Pai-1 blocker. In conclusion, APN-deficient mice developed more intestinal tumors. Decreased phosphorylation of AMPK and increased levels of Pai-1 were observed in APN-deficient mice compared with wild-type mice. APN and its receptors might be good targets for developing colorectal cancer chemopreventive agents. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2837. doi:10.1158/1538-7445.AM2011-2837
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5016-5016
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5016-5016
    Abstract: Background; Two dimensional single-layer culture still remains the preferred platform for most laboratory preclinical studies, although interaction between cancer and stromal cells has been well reported to be important in tumor progression and resistance against therapies. In order to mimic the patient tumor tissues, ex vivo model which recaptures the tumor microenvironment is required. Methods; Layered 3D stromal tissues were produced by culturing normal human dermal fibroblasts (NHDFs) and human umbilical vein endothelial cells (HUVECs) coated with extra-cellular matrix (ECM) and natural polysaccharide, namely collagen and heparin. The layered 3D stromal tissues and overlaid tumors were morphologically characterized by HE stain, immunohistochemistry (IHC) and immunofluorescence (IF). Furthermore, drug sensitivity assays were conducted using popular colorectal cancer cell lines, and patient-derived cell lines (PDCs) established in the laboratory of Japanese Foundation for Cancer Research. Cancer cell viability was evaluated by fluorescent labeling, enzymatic dissociation and cell counting analysis. IF with cancer specific markers and imaging analyses were also performed. Results; The 3D stromal tissues including CD31 positive luminal structure were multi-layered (approximately 20 layers), and formation of microvascular network was observed within several days. In comparison with 2D mono-culture or 3D mono/co-cultured spheroid model, decreased drug sensitivities were represented in our 3D co-cultured model. In the simultaneous treatment with cytotoxic anticancer agents and molecular targeted drugs, dose-responses were significantly different between the2D and 3D models. Conclusion; We developed the layered 3D stromal tissue culture system including blood micro-vessels. Drug evaluation with the co-cultured tumors may reflect the drug sensitivity of cancer cells in vivo. Our unique 3D ex vivo model represents a valuable tool for drug development in a fully human cell and matrix microenvironment, and thus testing patient-derived cells and approved compounds also enable better prediction their efficacy. Citation Format: Yuki Takahashi, Kei Tsukamoto, Shiro Kitano, Shinji Irie, Michiya Matsusaki, Satoshi Nagayama, Ryohei Katayama, Eiji Shinozaki, Naoya Fujita. A unique ex vivo drug evaluation model: 3D co-cultured system with tumor, stroma and blood microvessels [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5016.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...