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)  (9)
Material
Publisher
  • American Association for Cancer Research (AACR)  (9)
Language
Years
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 9 ( 2023-05-02), p. 1517-1530
    Abstract: Liposarcoma (LPS) is the most common soft-tissue sarcoma in adults with two major subtypes, well differentiated and dedifferentiated. Both subtypes are characterized with the pathognomonic giant ring or marker chromosomes that harbor high copy numbers of known oncogenes. Here, we reported a comprehensive molecular characterization of both tumor and normal tissues from the same patients with LPS, including whole-genome sequencing (WGS), transcriptome, enhancer landscape, and genome-wide three-dimensional (3D) genome structure by Hi-C. Tumor-specific transcripts and regulatory elements were identified, and enhancer coamplification and hijacking events were discovered as novel mechanisms upregulating oncogenes such as MDM2, CDK4, and HMGA2. Combining Hi-C, optical mapping, nanopore long reads, and WGS data partially resolved complex structural variations and reconstructed the local genome and the giant chromosome. Overall, this study provides a comprehensive resource for LPS research and offers insights into how altered enhancers and the 3D genome contribute to gene dysregulation in cancer. Significance: Comprehensive profiling of the enhancer landscape and 3D genome structure in liposarcoma identifies extensive enhancer-oncogene coamplification and enhancer hijacking events, deepening the understanding of how oncogenes are regulated in cancer.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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 ...
  • 2
    In: Cancer Research Communications, American Association for Cancer Research (AACR), Vol. 2, No. 11 ( 2022-11-10), p. 1404-1417
    Abstract: Tumor-associated macrophages (TAM) are the most abundant immune cells in the tumor microenvironment. They consist of various subsets but primarily resemble the M2 macrophage phenotype. TAMs are known to promote tumor progression and are associated with poor clinical outcomes. CD47 on tumor cells and SIRPα on TAMs facilitate a “don't-eat-me” signal which prevents cancer cells from immune clearance. Therefore, blockade of the CD47–SIRPα interaction represents a promising strategy for tumor immunotherapy. Here, we present the results on ZL-1201, a differentiated and potent anti-CD47 antibody with improved hematologic safety profile compared with 5F9 benchmark. ZL-1201 enhanced phagocytosis in combination with standards of care (SoC) therapeutic antibodies in in vitro coculture systems using a panel of tumor models and differentiated macrophages, and these combinational effects are Fc dependent while potently enhancing M2 phagocytosis. In vivo xenograft studies showed that enhanced antitumor activities were seen in a variety of tumor models treated with ZL-1201 in combination with other therapeutic mAbs, and maximal antitumor activities were achieved in the presence of chemotherapy in addition to the combination of ZL-1201 with other mAbs. Moreover, tumor-infiltrating immune cells and cytokine analysis showed that ZL-1201 and chemotherapies remodel the tumor microenvironment, which increases antitumor immunity, leading to augmented antitumor efficacy when combined with mAbs. Significance: ZL-1201 is a novel anti-CD47 antibody that has improved hematologic safety profiles and combines with SoC, including mAbs and chemotherapies, to potently facilitate phagocytosis and antitumor efficacy.
    Type of Medium: Online Resource
    ISSN: 2767-9764
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 3098144-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), ( 2023-09-26)
    Abstract: Bone marrow stromal cell (BMSC)-derived small extracellular vesicles (sEVs) promote drug resistance to bortezomib in multiple myeloma (MM) cells. Elucidating the components of BMSC sEV that induce drug resistance in MM cells could help identify strategies to overcome resistance. Considering the hypoxic nature of the myeloma microenvironment, we explored the role of hypoxia in regulating BMSC sEV cargo and investigated whether hypoxia-driven sEV miRNAs contribute to the drug resistance in MM cells. Hypoxia increased the release of sEVs from BMSCs, and these sEVs more strongly attenuated bortezomib sensitivity in MM cells than sEVs from BMSCs under normoxic conditions. RNA sequencing revealed that significantly elevated levels of miR-140-5p and miR-28-3p were enclosed in hypoxic BMSCs-derived sEVs. Both miR-140-5p and miR-28-3p conferred bortezomib resistance in MM cells by synergistically targeting SPRED1, a member of the Sprouty protein family that regulates MAPK activation. SPRED1 inhibition reduced sensitivity to bortezomib in MM cells through activating MAPK-related pathways and significantly promoted MM bortezomib resistance and tumor growth in a mouse model. These findings shed light on the role of hypoxia-induced miRNAs shuttled in BMSC-derived sEVs to MM cells in inducing drug resistance and identify the miR-140-5p/miR-28-3p/SPRED1/MAPK pathway as a potential targetable axis for treating MM.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Prevention Research Vol. 4, No. 11 ( 2011-11-01), p. 1863-1872
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 4, No. 11 ( 2011-11-01), p. 1863-1872
    Abstract: Alcohol drinking is a known risk factor for oral cancer in humans. However, previous animal studies on the promoting effect of ethanol on oral carcinogenesis were inconclusive. It is necessary to develop an animal model with which the molecular mechanism of ethanol-related oral carcinogenesis may be elucidated to develop effective prevention strategies. In this study, mice were first treated with 4-nitroquinoline-1-oxide (4NQO, 100 μg/mL in drinking water) for 8 weeks and then given water or ethanol (8%) as the sole drink for another 16 weeks. During the experiment, 8% ethanol was well tolerated by mice. The incidence of squamous cell carcinoma (SCC) increased from 20% (8/41) to 43% (17/40; P & lt; 0.05). Expression of 5-lipoxygenase (5-Lox) and cyclooxygenase 2 (Cox-2) was increased in dysplasia and SCC of 4NQO-treated tongues and further enhanced by ethanol. Using this mouse model, we further showed that fewer cancers were induced in Alox5−/− mice, as were cell proliferation, inflammation, and angiogenesis in the tongue, as compared with Alox5+/+ mice. Interestingly, Cox-2 expression was induced by ethanol in knockout mice, whereas 5-Lox and leukotriene A4 hydrolase (LTA4H) expression and leukotriene B4 (LTB4) biosynthesis were dramatically reduced. Moreover, ethanol enhanced expression and nuclear localization of 5-Lox and stimulated LTB4 biosynthesis in human tongue SCC cells (SCC-15 and SCC-4) in vitro. In conclusion, this study clearly showed that ethanol promoted 4NQO-induced oral carcinogenesis, at least in part, through further activation of the 5-Lox pathway of arachidonic acid metabolism. Cancer Prev Res; 4(11); 1863–72. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6174-6174
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6174-6174
    Abstract: Background: Cervical cancer ranks fourth in incidence and mortality among female tumors, with over 0.6 million patients diagnosed worldwide in 2020. A large percentage of cervical cancer patients will develop advanced diseases (stage IV or recurrent disease) and the 5-year survival rate of these patients was only 15-20% due to the lack of efficacy and adverse effects of first line chemodrugs. Recently, antibody drug conjugates (ADCs) emerge as a novel class of targeted therapeutics that selectively ablate tumor cells without damaging normal organs and tissues, which has shown promising efficacy in treating solid tumors. In this study, we sought to develop a cervical cancer-targeted ADC using a novel molecular target. Methods: Firstly, human cervical cancer and paracancerous tissue were stained by immunohistochemistry (IHC) in order to determine the difference of the expression level of a molecular target. Three human cervical cancer cell lines (C-33A, Si-Ha and Ca-Ski) and one normal cervical cell line (HCerEpiC) were used to determine the subcellular location of an ADC target on the cells by immunofluorescence (IF) staining, flow cytometry and single photon confocal microscopy. Secondly, endocytosis capability of target antibodies was visualized by confocal imaging and was quantified by flow cytometric analysis. Furthermore, a panel of ADC formulations with different chemical linkers and payloads were designed, constructed and characterized as ADC candidates for cervical cancer therapy. We next evaluated in vitro inhibitory activity of these ADC candidates on three cervical cancer cell lines by CCK8 assay. Finally, orthotopic and lung metastasis animal models of cervical cancer were established to determine the in vivo inhibitory activity and biosafety of constructed ADC candidates. Results: IHC staining of clinical specimens showed that the expression level of CD54 in cervical tumor tissues was significantly higher than those of paracancerous tissues. Fluorescence imaging and flow cytometry further determined the overexpression level, plasma membrane location and antigen-mediated endocytosis activity of CD54 in multiple human cervical cancer cell lines. Among several ADC formulations, an optimized CD54-targeted ADC linker and warhead combination was identified by comparing its IC50 with other ADC candidates and standard-of-care chemodrugs for cervical cancer. The optimized CD54 ADC exhibited promising anti-tumor activity on primary and lung metastatic cervical tumor models. Conclusion: In our study, we identified CD54 as a novel ADC target for cervical cancer. An optimized CD54-targeted ADC formulation was determined and its anti-tumor activity and biosafety profile were determined in multiple human cervical cancer cell lines in vitro and orthotopic and lung metastatic cervical tumor models in vivo, providing a promising targeted therapeutic candidate for the targeted treatment of cervical cancer. Citation Format: Shili Yao, Bing Zhu, Xinyan Wang, Rui Xu, Tong Yang, Peng Guo, Huarong Tang, Tao Zhu. Developing a novel antibody drug conjugate against cervical cancer [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 6174.
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3913-3913
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3913-3913
    Abstract: Background: As a key receptor tyrosine kinase (RTK) regulating tumorigenesis, AXL is overexpressed in multiple different types of cancers. AXL can be activated by homodimerization after binding with its ligand, growth arrest specific 6 protein (Gas6) or by heterodimerization with other RTKs. AXL overexpression and/or activation have been reported to promote cancer cell proliferation, migration/invasion and to induce resistance to targeted therapy. However, the AXL downstream signaling network has not yet been fully elucidated. Methods: We employed a CRISPR-CAS9-based knockout technology to create AXL inducible knockout cells in a human triple negative breast cancer (TNBC) cell line, HCC1395. In order to explore the downstream signaling events driven by AXL, we induced AXL knockout for 72 hours and used a 6-plex TMT-labeling-based quantitative proteomic approach to characterize the protein phosphorylation alterations induced by AXL knockout. IMAC and anti-phosphotyrosine antibody (pY1000)-based phosphopeptide enrichment methods were used to comprehensively quantify the phosphoproteome changes induced by AXL knockout. Results: In this study, we identified 38,525 phospho serine/threonine sites and 1,427 phosphotyrosine sites. In response to AXL knockout, 1,581 phospho serine/threonine sites and 40 phosphotyrosine sites showed significant differences between AXL-KO and AXL-WT groups with & gt;1.5-fold change and p & lt;0.05 as the cutoff. Among them, 982 phosphoserine/threonine sites and 30 phosphotyrosine sites were decreased, while 599 phospho serine/threonine sites and 10 phosphotyrosine sites were increased in AXL-KO cells. Signaling pathway enrichment analysis revealed that suppressing AXL expression could inhibit multiple important cancer related signaling pathways, including cell cycle, focal adhesion, MAPK signaling pathway, p53 signaling pathway and mTOR signaling pathway. Conclusion: Quantitative analysis of the phosphoproteome driven by AXL demonstrated that AXL plays a pivotal role in regulating canonical oncogenic signaling but also signaling pathways involved in RNA processing and DNA repair. Our data also suggest that targeting AXL could suppress these oncogenic signaling pathways and have therapeutic potential to improve clinical outcomes for patients with AXL overexpression. Citation Format: Li Wang, Nicole Pearson, Yuning Xiong, Santosh Renuse, Ran Cheng, Jodi M. Carter, Akhilesh Pandey, Xinyan Wu. Quantitative phosphoproteomic analysis of AXL signaling network in breast cancer [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 3913.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1601-1601
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1601-1601
    Abstract: Alcohol drinking is a known risk factor for oral cancer in humans. However, previous animal studies on the promoting effect of ethanol on oral carcinogenesis were inconclusive. It is necessary to develop an animal model with which the molecular mechanism of ethanol-related oral carcinogenesis may be elucidated in order to develop effective prevention strategies. In this study, mice were first treated with 4-nitroquinoline-1-oxide (4NQO, 100μg/ml in drinking water) for 8 weeks, and then given water or ethanol (8%) as the sole drink for another 16 weeks. During the experiment, 8% ethanol was well tolerated by mice. The incidence of squamous cell carcinoma (SCC) increased from 20% (8/41) to 43% (17/40; p & lt;0.05). Expression of 5-lipoxygenase (5-Lox) and cyclooxygenase 2 (Cox-2) was increased in dysplasia and SCC of 4NQO-treated tongues, and further enhanced by ethanol. Using this mouse model, we further demonstrated that fewer cancers were induced in Alox5-/- mice, as were cell proliferation, inflammation, and angiogenesis in the tongue, as compared with Alox5+/+ mice. Interestingly, Cox-2 expression was induced by ethanol in knockout mice, while 5-Lox and leukotriene A4 hydrolase (LTA4H) expression and leukotriene B4 (LTB4) biosynthesis were dramatically reduced. Moreover, ethanol enhanced expression and nuclear localization of 5-Lox and stimulated LTB4 biosynthesis in human tongue SCC cells (SCC-15 and SCC-4) in vitro. In conclusion, this study clearly demonstrated that ethanol promoted 4NQO-induced oral carcinogenesis, at least in part, through further activation of the 5-Lox pathway of arachidonic acid metabolism. 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 1601. doi:1538-7445.AM2012-1601
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5072-5072
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5072-5072
    Abstract: Gastric cancer (GC) ranks the fifth common cancer worldwide. Patients with GC suffer from a low 5-y survival rate due to the lack of effective treatments. Based on the genomic and molecular characteristics of GC, the Cancer Genome Atlas (TCGA) classified GC into four molecular subtypes: microsatellite-instable (MSI), Epstein-Barr virus-positive (EBV), chromosomal-instable (CIN), and genomically stable (GS). CIN tumors usually occur at gastroesophageal junction and normally belong to intestinal type of Lauren classification, which are characterized by DNA anueploidy, abnormal chromosomal copy numbers, and the frequent mutation of gene TP53, leading to chromosomal instability. CIN represents the most prevalent GC subtype ( & gt;50%) with a poor prognosis, highlighting a significant and unmet medical need for developing effective CIN-targeted therapeutics. Antibody drug conjugates (ADCs) are a new class of targeted therapeutics, composed of a humanized monoclonal antibody and small molecular cytotoxic drugs via chemical linkers. In this study, we designed and engineered a novel ADC targeting chromosome instable gastric cancer (CINGC). We first explored intercellular adhesion molecule-1 (ICAM1) as a novel molecular target for CINGC by characterizing its overexpression levels in a panel of human GINGC cell lines. ICAM1 is a cell surface transmembrane glycoprotein with functions of regulating cell-cell adhesion, signaling, and transendothelial migration of leukocytes to sites of inflammation. Many malignant tumors including GC were found to aberrantly overexpress ICAM1 on the surfaces of tumor cells. The high overexpression of ICAM1 on CINGC cells was validated by using flow cytometry (FCM) quantitatively and immunofluorescence (IF) staining qualitatively. To correlate it with clinically relevant GC tumor tissues, immunohistochemical (IHC) staining of ICAM1 was then conducted to determine the high expression of tumor tissue compared with para cancer tissue. The antigen-mediated endocytosis of ICAM1 monoclonal antibody was further detected by FCM quantitatively and IF visually. All the results showed that ICAM1 was an promising molecular target for CINGC, therefore, it was selected to engineer CINGC-targeted ADCs. We engineered a panel of ADC formulations with different linkers and cytotoxic payloads. We identified an optimal ADC formulation for CINGC by screening their cytotoxicity against CINGC cells in vitro in comparison with first line chemodrugs including 5-FU and oxaliplatin, which efficacy was validated by measuring its inhibitory activity on tumor growth, progression and metastasis via CINGC xenograft models. In conclusion, a novel ICAM1 ADC was designed and engineered to selectively recognize and ablate CINGC cells in vitro and in vivo. Our engineered new ADC offers the possibility of improving the clinical outcome and life quality of CINGC patients and expanding the scope of clinical use of drugs. Citation Format: Xinyan Wang, Li Yuan, Xiaoqing Guan, Shili Yao, Bing Zhu, Chun Liu, Tong Yang, Peng Guo, Jiangjiang Qin, Xiangdong Cheng. A novel antibody drug conjugateengineered for chromosome instable gastric cancer [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 5072.
    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) ; 2022
    In:  Cancer Research Vol. 82, No. 17 ( 2022-09-02), p. 3074-3087
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 17 ( 2022-09-02), p. 3074-3087
    Abstract: Hepatocellular carcinoma (HCC) is one of the most aggressive malignancies. Elucidating the underlying mechanisms of this disease could provide new therapeutic strategies for treating HCC. Here, we identified a novel role of DEAD-box helicase 24 (DDX24), a member of the DEAD-box protein family, in promoting HCC progression. DDX24 levels were significantly elevated in HCC tissues and were associated with poor prognosis of HCC. Overexpression of DDX24 promoted HCC migration and proliferation in vitro and in vivo, whereas suppression of DDX24 inhibited both functions. Mechanistically, DDX24 bound the mRNA618–624nt of laminin subunit beta 1 (LAMB1) and increased its stability in a manner dependent upon the interaction between nucleolin and the C-terminal region of DDX24. Moreover, regulatory factor X8 (RFX8) was identified as a DDX24 promoter-binding protein that transcriptionally upregulated DDX24 expression. Collectively, these findings demonstrate that the RFX8/DDX24/LAMB1 axis promotes HCC progression, providing potential therapeutic targets for HCC. Significance: The identification of a tumor-promoting role of DDX24 and the elucidation of the underlying regulatory mechanism provide potential prognostic indicators and therapeutic approaches to help improve the outcome of patients with hepatocellular carcinoma.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...