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)  (6)
Material
Publisher
  • American Association for Cancer Research (AACR)  (6)
Language
Years
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5623-5623
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5623-5623
    Abstract: CD137 (41-BB, tumor necrosis factor receptor superfamily 9), expressed on activated T cells and NK cells, has gained increased attention as an anti-tumor target because of remarkable clinical efficacy using either a single agent or in combination with other immune checkpoint inhibitors. We constructed CD137 humanized mice based on two different backgrounds, C57BL6 and BALB/c mice, as well as dual-target and triple-target mice crossbred with PD1/PDL1 humanized mice to create an ideal model to study CD137 target drugs. The normal immune cell population in peripheral blood were not affected in CD137 humanized mice, and hCD137 was expressed on T cells after the stimulation with a anti CD3 antibody. We verified the efficacy of anti-CD137 drugs in BALB/c-hCD137 mice inoculated with different cancer cell lines (i.e. CT26, EMT6 and 4T1). The drug Urelumab showed a dramatic response in the hCD137 mice model inoculated with CT26 and EMT6 but not with 4T1 cell line. In a rechallenge study, no tumors grew in tumor-free mice previously treated with Urelumab, with effector memory T cells increased in peripheral blood, indicating Urelumab has a relatively long-lasting anti-tumor effect. The combination of Urelumab and anti-PD1 has a stronger tumor growth inhibition effect than that of anti-CD137 or anti-PD1 monotherapy on dual-target or triple target mice. These data show that the humanized CD137 mouse is a suitable model for evaluating the monotherapies or combination therapies targeting CD137. Citation Format: Yunlong Jiang, Tingting Gu, Weiwei Yu, Hongyan Sun, Mingkun Zhang, Juan Liang, Shuai Li, Cunxiang Ju, Jing Zhao, Xiang Gao, Mark W. Moore. CD137 humanized mice for preclinical efficacy evaluation of therapeutic antibodies [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 5623.
    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 ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 930-930
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 930-930
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies due to its late diagnosis and limited response to treatment. B6-KPC mice ( LSL-KrasG12D/+, LSL-Trp53R172H/+, Pdx1-Cre) generate spontaneous PDAC which could recapitulate the pathogenesis and genomic features of human PDAC. Although B6-KPC model is a wildly used for the exploration of the mechanisms of PDAC, some drawbacks of the model, such as long time for tumorigenesis, individual variation and difficulties in tumor observation, limit its application in cancer therapy. It is necessary to develop the pancreatic tumor cell lines from these models. In this study, we successfully established and well-characterized the luciferase-labled cell line mPAKPC-Luciferase. This cells carried the KrasG12D and Trp53R172H mutation. The orthotopic mPAKPC-Luciferase tumor model was well established by implanting tumor cell line into pancreas of B6 wild type mice and the tumor growth was traced by in vivo imaging. Moreover, Gemcitabine or combined with CD40 agonist presented significant tumor growth inhibition in B6 bearing mPAKPC-Luciferase orthotopic mouse models. While anti-PD1 or anti-OX40 had no anti-tumor effects in this model accordance with the low T cell infiltration of mPAKPC modle as a “cold” tumor. In addition, the immune profiling of this cell line showed highly expressed of potential therapeutic targets, including Claudin 18.2, CD47, CD73. Thus, mPAKPC-Luciferase cell lines are valuable models for the PDAC preclinical therapies. Citation Format: Fang Zhu, Cunxiang Ju, Hongyan Sun, Weiwei Yu, Chao Ju, Shuai Li, Yunlong Jiang, Dongjing Jia, Steve Smith, Zhiying Li, Jing Zhao, Xiang Gao. Novel orthotopic pancreatic cancer cell lines derived from B6-KPC mice [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 930.
    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 ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 10, No. 1 ( 2004-01-01), p. 19-27
    Abstract: Purpose: Cyclic AMP response element binding protein binding protein (CBP), a nuclear transcriptional coactivator protein, is an important component of the cAMP signal transduction pathway. In this study, we systematically analyzed the pattern and frequency of CBP gene alterations in esophageal squamous cell carcinoma (ESCC) samples from Linzhou (Linxian), China. Experimental Design: Using microsatellite markers D16S475, D16S2622, and D16S523 within the chromosome 16p13.3 locus flanking the CBP gene, we observed loss of heterozygosity (LOH), microsatellite instability (MSI), or homozygous deletion in 16 of 26 ESCC samples. Additional ESCC samples were analyzed using different sets of microsatellite markers (CS1–CS5) within the introns or in close proximity to the 3′ end of the CBP gene. Results: The data showed that CBP gene LOH or MSI occurred in 9 of 19 ESCC samples. A detailed genetic alteration map of the CBP gene showed that an LOH or MSI hot spot occurred within intron 2 of the CBP gene. Furthermore, ESCC samples were investigated for CBP gene mutation by conformation sensitive gel electrophoresis and DNA sequencing. These results revealed that most of the shifted fragments contained internal tandem duplication (ITD), frequently in the regions encoding the histone acetyltransferase domain and COOH-terminal transactivating domain one of the CBP gene. The presence of ITD within the CBP gene was additionally confirmed by Southern blot analysis and sequencing. Conclusions: These studies show that LOH and ITD of the CBP gene are frequent genetic events in human ESCC. These alterations may have functional importance in the development of human ESCC.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2584-2584
    Abstract: The Fc gamma receptor (FcγR), a member of the immunoglobulin superfamily, binds the IgG Fc segment of antibodies and is mainly expressed on the surface of immune cell. The FcγR binds to the Fc region of an antibody that is attached to target cells, transmitting an activation or inhibition signal to the FcγR expressing cells, resulting in various biological responses and functions. Many therapeutic antibodies are developed based on the engineering of the Fc region to fine-tune the FcγR mediated effects. There are four subtypes of mouse FcγRs, including Fcgr1, Fcgr2b, Fcgr3, and Fcgr4. In contrast, human FcγRs consist of at least seven members, including FCGR1A, FCGR1B, FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B. Additionally, the expression pattern of even the orthologous FcγR genes varies between humans and mice. e.g., the distribution of mouse FCGR3 and FCGR2. Therefore, animals harboring the murine FcγRs are not ideal models for the accurate preclinical evaluation of human therapeutic antibodies. To address this problem, we established an FcγRs fully humanized mouse model by injecting an engineered human bacterial artificial chromosome construct carrying all seven human FcγR genes into mouse zygotes. The transgene carriers were mated with another strain in which all mouse FcγR genes were knocked out to establish the colony. Unlike the endogenous mouse FcγR genes expression profiles, the transgenic human FcγR genes regulated under the control of the human control elements retain the human expression pattern. For example, the transgenic human FCGR3A was observed on mouse NK cells and macrophages, whereas the FCGR3B was detected on mouse neutrophils only, which were consistent with their expression patterns in humans. Antibody-dependent cellular cytotoxicity (ADCC) experiment indicated that a human therapeutic antibody could direct NK cells derived from the FcγRs fully-humanized mice to kill target tumor cells. Moreover, a significant anti-tumor effect could also be achieved by an ADCC based human antibody in tumor-bearing FcγRs humanized mice. Taken together, the FcγR gene fully-humanized model established in this study demonstrates a human FcγR expression pattern and functionality, thus providing a valuable tool for accurate preclinical evaluation of human therapeutic antibodies. Citation Format: Mingkun Zhang, Cunxiang Ju, Ying Li, Yunlong Jiang, Weiwei Yu, Shuai Li, Hongyan Sun, Zhiying Li, Jing Zhao, Xiang Gao. Development of an Fc gamma receptor fully-humanized mouse model to support the accurate preclinical study of therapeutic antibodies [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 2584.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Clinical Cancer Research Vol. 22, No. 2_Supplement ( 2016-01-15), p. A70-A70
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 2_Supplement ( 2016-01-15), p. A70-A70
    Abstract: Purpose: Epigenetic changes, particularly in DNA methylation, have been implicated in acquired resistance to platinum in ovarian cancer (OC). The goal of the current study was to analyze and integrate global RNA expression and DNA methylation profiles of platinum resistant tumors compared to untreated, platinum-sensitive ovarian tumors, as well as to measure genomic and epigenomic changes induced by guadecitabine (SGI-110) in tumors. Methods: An ongoing phase I/II multi-institutional clinical trial uses the novel DNA methyltransferase (DNMT) inhibitor guadecitabine to re-sensitize recurrent platinum resistant OC to carboplatin. Patients enrolled in this trial had recurrent platinum resistant OC. Tumor biopsies were collected at baseline and after two cycles of guadecitabine administered daily for 5 days at a low (30mg/m2) dose (28 days per cycle). RNA and DNA were extracted from 48 and 57 baseline tumors and analyzed for next generation sequencing approaches to interrogate transcriptomes (RNA-seq) and methylomes (Infinium Human Methylation450 (HM450) arrays), respectively. Differential gene expression and DNA methylation profiles were generated and used for Ingenuity Pathway Analysis (IPA) to identify the top altered pathways in response to guadecitabine. Expression of DNMTs was examined by real-time RT-PCR and immunohistochemistry. LINE1 methylation and promoter methylation of selected genes (MAGE-A2, MAGE-A3, MAGE-A11, NY-ESO, RASSF1, MLH1, and HOXA11) were quantified by pyrosequencing before and after guadecitabine treatment (n=12 paired samples). Results: Analysis of a limited number of paired samples before and after treatment (n=8) revealed significant changes in global gene expression profiles induced by guadecitabine, with 960 altered genes representing immunopathway enrichment including cytokine production in macrophages and T helper cells by IL-17A and IL-17F, granulocyte /agranulocyte adhesion and inflammation, IL-8 signaling, p38 MAPK signaling, cAMP-mediated signaling, and innate immunity. Epigenetic profiling using HM450 revealed extensive methylation changes when comparing recurrent platinum resistant ovarian tumors (n=42) to primary, untreated ovarian cancer specimens analyzed as part of the TCGA project (n=10). Six hundred and four promoters were significantly differentially methylated (adjusted p & lt;0.05, absolute methylation changes β & gt;0.2), among which, 498 and 106 were hypermethylated or hypomethylated respectively in recurrent platinum resistant ovarian tumors. IPA analysis of baseline tumor transcriptome and methylome demonstrated significant enrichment in a wide range of pathways associated with cancer, stem cells, inflammation and the immune system. DNMT1, 3A, and 3B mRNA levels in the tumors were highly variable (n=19). Analysis of a limited number of paired samples (n=7) revealed no significant changes in global methylation or in DNMT expression levels induced by treatment with guadecitabine (adjusted p & gt;0.05). However, the DNMT inhibitor induced significant methylome alterations in selected patients. Significant hypomethylation of MAGE-A3 and MAGE–A11 promoters (p & lt;0.05) was detected. Correlations between methylation changes and clinical outcomes are being explored. Conclusions: These data suggest that treatment with the DNMT inhibitor guadecitabine induces a reactivation of immune responses in human OC. Correlations between methylation changes and expression profiles are being explored. Citation Format: Fang Fang, Horacio Cardenas, Dave Miller, Aaron Buechlein, Qing Yu, Yunlong Liu, Guanglong Jiang, Pietro Taverna, Harold Keer, Doug Rusch, Daniela Matei, Kenneth P. Nephew. Epigenome and genome alterations in platinum resistant ovarian tumors. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: Exploiting Vulnerabilities; Oct 17-20, 2015; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(2 Suppl):Abstract nr A70.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. B158-B158
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer-related mortality in the US. Most patients present with advanced disease, and ∼95% die within five years. Treatment with chemotherapy has not changed the natural course of this disease, and just recently, with combination of chemotherapeutic agents, the median survival reached a year. Several mechanisms are proposed to play a role in the aggressive, treatment-resistant phenotype of PDAC, including adaptation to hypoxia, which leads to increased potential for metastasis and impairs the efficacy of chemotherapy and radiotherapy. Hypoxia-Inducible Factor-1α (HIF1α), a major oxygen sensor in cells, is a transcription factor that is rapidly degraded under normoxic conditions but upregulates a number of genes under hypoxic conditions that contribute to survival, metastasis, and angiogenic signaling in the tumor microenvironment. One of the most notable HIF targets is Carbonic Anhydrase IX (CA9), which promotes tumor cell survival and metastasis by maintaining a steady intracellular pH while acidifying the microenvironment, thereby encouraging epithelial-mesenchymal transition and contributing to extracellular matrix degradation. AP Endonuclease1/ Redox Effector Factor 1 (APE1/Ref-1) is a dual function protein that possesses a DNA repair function as well as the ability to reduce transcription factors and enable them to bind to their DNA target sequences. APE1/Ref-1 regulates several transcription factors involved in preventing apoptosis, survival mechanisms, and hypoxia signaling, including HIF-1α. Therefore, we hypothesized that APE1/Ref-1 inhibition impairs HIF-1α-mediated signaling, and this leads to decreased survival and invasion of tumor cells exposed to hypoxic conditions. Methods: We performed co-immunoprecipitation (co-IP) studies to look at the interaction of APE1/Ref-1 with transcriptional targets, HIF-1α, STAT3, and NFκB along with RT-PCR and Western blotting to confirm expression of hypoxia signaling genes. Luciferase reporter assays were used to quantitate transcriptional activation under hypoxia. Boyden chamber was used to look at migration and invasion as well as proliferation based assays following manipulation of APE1/Ref-1 and hypoxia. Results: HIF-1α and STAT3, but not NFκB associate with APE1/Ref-1 under hypoxia. Moreover, we found that knockdown of APE1/Ref-1 protein diminishes HIF-mediated transcription in hypoxia, as shown by analysis of luciferase reporter assays. Next, we showed that, in hypoxia, APE1/Ref-1 inhibition diminishes HIF-1α-induced downstream targets including CA9 and ANGPTL4 further indicating that APE1/Ref-1 redox activity regulates HIF signaling. Importantly, we found that hypoxia, in the presence or absence of APE1/Ref-1, no longer induced CA9 mRNA levels in HIF-deficient MEFs, proving that hypoxia-dependent regulation of CA9 expression is fully mediated by HIF-1α. A blockade of both CA9 activity via small molecule and CA9 transcription via APE1/Ref-1 leads to decreased PDAC cell proliferation under hypoxia. These data indicate that APE1/Ref-1 inhibition interferes with ηψπoξια-mediated signaling and can further sensitize PDAC cells to CA9/12 inhibition even under the conditions of extreme oxygen deprivation. Ongoing experiments will determine the role APE1/Ref-1 plays in the survival and invasion of tumor cells exposed to hypoxia. Citation Format: Derek P. Logsdon, Huiwen Cheng, Meihua Luo, Safi Shahda, Yangyang Hao, Yan Tong, Zhangsheng Yu, Nicholas Zyromski, Ernestina Schipani, Yunlong Liu, Claudiu T. Supuran, Mircea Ivan, Mark R. Kelley, Melissa L. Fishel. Targeting APE1/Ref-1 results in inhibition of hypoxia signaling genes. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B158.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...