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)  (14)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Clinical Cancer Research Vol. 22, No. 12 ( 2016-06-15), p. 3105-3117
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 12 ( 2016-06-15), p. 3105-3117
    Abstract: Purpose: Hepatocellular carcinoma (HCC) lacks effective curative therapy. Hypoxia is commonly found in HCC. Hypoxia elicits a series of protumorigenic responses through hypoxia-inducible factor-1 (HIF1). Better understanding of the metabolic adaptations of HCC cells during hypoxia is essential to the design of new therapeutic regimen. Experimental Design: Expressions of genes involved in the electron transport chain (ETC) in HCC cell lines (20% and 1% O2) and human HCC samples were analyzed by transcriptome sequencing. Expression of NDUFA4L2, a less active subunit in complex I of the ETC, in 100 pairs of HCC and nontumorous liver tissues were analyzed by qRT-PCR. Student t test and Kaplan–Meier analyses were used for clinicopathologic correlation and survival studies. Orthotopic HCC implantation model was used to evaluate the efficiency of HIF inhibitor. Results: NDUFA4L2 was drastically overexpressed in human HCC and induced by hypoxia. NDUFA4L2 overexpression was closely associated with tumor microsatellite formation, absence of tumor encapsulation, and poor overall survival in HCC patients. We confirmed that NDUFA4L2 was HIF1-regulated in HCC cells. Inactivation of HIF1/NDUFA4L2 increased mitochondrial activity and oxygen consumption, resulting in ROS accumulation and apoptosis. Knockdown of NDUFA4L2 markedly suppressed HCC growth and metastasis in vivo. HIF inhibitor, digoxin, significantly suppressed growth of tumors that expressed high level of NDUFA4L2. Conclusions: Our study has provided the first clinical relevance of NDUFA4L2 in human cancer and suggested that HCC patients with NDUFA4L2 overexpression may be suitable candidates for HIF inhibitor treatment. Clin Cancer Res; 22(12); 3105–17. ©2016 AACR.
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. LB-310-LB-310
    Abstract: Background & Aims: Liver is a major metabolic organ, yet the detailed metabolic alterations driving hepatocellular carcinoma (HCC) remain elusive. The rapid growing nature of HCC results in oxygen deprivation or hypoxia in regions of tumors with insufficient blood supply. Hypoxia unbalances the electron flow through the electron transport chain (ETC) resulting in reactive oxygen species (ROS) accumulation. Here we aim at delineating the mechanisms by which HCC evades oxidative stress. Methods: We performed transcriptome sequencing to study the gene expression profile in both HCC patients and HCC cell line. The mRNA expression of 100 paired HCC and corresponding non-tumorous tissues were analyzed. Stable RNAi knockdown by shRNA and genetic knockout by TALEN were established in HCC cells for functional characterization. Results: We demonstrated that HCC cells specifically utilized the mitochondrial protein NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4-like 2 gene (NDUFA4L2), in the complex I of the ETC, to survive hypoxia. NDUFA4L2 was drastically over-expressed in human HCC and closely associated with poor clinical outcomes in HCC patients. We confirmed that NDUFA4L2 was regulated by HIF-1α in HCC cells. Inactivation of HIF-1α/NDUFA4L2 in different HCC cell lines increased mitochondrial activity and oxygen consumption, resulting in ROS accumulation and ROS-mediated apoptosis in HCC cells. Knockdown of NDUFA4L2 markedly suppressed HCC growth and metastasis in vitro and in vivo. In addition, HIF inhibitors, digoxin and sorafenib, significantly suppressed growth of tumors that expressed high level of NDUFA4L2 in orthotopic HCC model. Conclusions: Our results have unprecedentedly uncovered the clinical relevance and oncogenic roles of NDUFA4L2 in HCC. Citation Format: Robin Kit-Ho Lai, Irix Ming-Jing Xu, David Kung-Chun Chiu, Aki Pui-Wah Tse, Larry Lai Wei, Cheuk-Ting Law, Derek Lee, Chun-Ming Wong, Maria Pik Wong, Irene Oi-Lin Ng, Carmen Chak Lui Wong. NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4-like 2 (NDUFA4L2) reduces oxidative stress to promote hepatocellular carcinoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr LB-310.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 1121-1121
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 1121-1121
    Abstract: BACKGROUND: The anaplastic lymphoma kinase (ALK) gene is frequently involved in chromosomal translocations resulting in fusion genes with different partners found in various lympho-proliferative conditions. In non-small cell lung cancers (NSCLC), the fusion protein encoded by echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusion gene was recently reported to confer oncogenic properties. This study aimed to identify other possible ALK fusion genes and analyze their oncogenic potentials. METHODS: Immunohistochemical analysis was used to screen for aberrant ALK expression in primary NSCLC. 5’ rapid amplification of cDNA ends was used to screen for potential novel 5’ fusion partners of ALK other than EML4-ALK. Reverse transcription polymerase chain reaction and fluorescence in situ hybridization were used to confirm the identity of 5’fusion partner. The genomic breakpoint was verified using genomic sequencing. Overexpression of the novel ALK fusion gene and variants 3a and 3b of EML4-ALK was performed to assess downstream signaling and functional effects. RESULTS: We identified a novel gene resulting from fusion of kinesin family member 5B (KIF5B) exon 15 to ALK exon 20 in a primary lung adenocarcinoma. Western blot of the clinical tumor tissues showed expression of a protein whose size correlated with that of the predicted KIF5B-ALK. Overexpression of KIF5B-ALK in mammalian cells led to STAT3 and AKT activation, enhanced cell proliferation, migration and invasion. These oncogenic properties were comparable to those of EML4-ALK variants. CONCLUSIONS: The discovery of the novel KIF5B-ALK variant with transforming functions further consolidates the role of aberrant ALK signaling in lung carcinogenesis. 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 1121. doi:10.1158/1538-7445.AM2011-1121
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. LB-052-LB-052
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. LB-052-LB-052
    Abstract: Lung cancer is the leading cause of cancer mortality. Adaptive responses to microenvironmental stresses are important survival mechanisms of cancer cells that lead to long term treatment failure, with accentuated resilience in cell subsets identified by cancer stem cell markers such as CD44 and ALDH. Reactive oxidative species (ROS) are harmful by-products of both cancer cell proliferation and treatment-induced cell death, the elimination of which is critical for maintaining cellular homeostasis and survival. We previously observed the calcium/calmodulin-dependent protein kinase II alpha (CaMK2A) could enhance cancer stem cell (CSC) properties and attenuate ROS in lung cancer cells. To understand the molecular mechanisms, we focused on its relation to the master transcription factor for ROS regulation, NRF2, and the downstream pathways. To test if NRF2 could be a substrate activated by CaMK2A, total NRF2 proteins were measured in nuclear protein extracted from lung cancer cells. CaMK2A-overexpression (OE) increased, while shRNA-mediated CaMK2A-knockdown (shCaMK2A) reduced nuclear NRF2, indicating NRF2 is a mediator of CaMK2A. To identify downstream molecular pathways, screening for known NRF2 target genes was performed using qPCR. The results identified Glutathione S-transferase P1 (GSTP1), an anti-oxidant and detoxifying enzyme, was a target observed consistently in CaMK2A-OE and shCaMK2A cell lines. This finding was confirmed at the protein levels by western blot, supporting CaMK2A/NRF2/GSTP1 might be a candidate axis playing a supportive role for lung CSC. To obtain further support, we studied the expression levels of GSTP1 in 44 clinical lung cancers from our cohort and the TCGA database, respectively, which showed significant up-regulation in cancers compared to corresponding normal lung samples. Further investigations showed GSTP1 protein levels were higher in tumor-spheres than monolayers of five established lung cancer cell lines. Stable knockdown of GSTP1 led to significant reduction of tumor sphere formation, expression of CD44 and ALDH1, as well as mouse xenograft tumorigenicity. On the other hand, sensitivities to chemotherapy and mutant EGFR targeting drugs were increased. Thus, the in vitro and in vivo data supported that GSTP1 played a role in the maintenance of the stem cell-like phenotypic state in lung cancer. To assess if GSTP1 maintained the CSC phenotype through regulating oxidative stress, its effects on ROS levels was measured using cellROX deep red probes. In shGSTP1 cells, the ROS level was significantly raised while it was reciprocally reduced on GSTP1- OE. Furthermore, ROS challenge experiments using N-acetyl-L-cysteine (NAC) in shGSTP1 cell lines resulted in increased cell sphere formation. Together, the results support the CaMK2A/NRF2/GSTP1 axis increases cancer cell resilience through ROS elimination and enhancement of stem cell-like properties of lung carcinoma, indicating its potential role as a long-term treatment target. Citation Format: SIQI WANG, Zhijie Xiao, Pui Chi Tin, Xuyuan Gao, Judy Wai Ping Yam, Maria Pik Wong. Stress response through theCaMK2A/NRF2/GSTP1 axis strengthens cancer cell resilience and mediates stemness phenotype through ROS attenuation [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 LB-052.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1949-1949
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1949-1949
    Abstract: The p38 MAPK cascade regulates body responses to a wide range of stress stimuli in a context-dependent and tissue-specific manner. The isoform p38alpha (p38) mediates both tumor suppressive and tumor-supportive functions in different cancer models. However, its role in lung cancer is not clear. Since EGFR is frequently mutated in lung adenocarcinomas in non-smoking women and p38 is a downstream target of EGFR signaling, it is possible that p38 plays a tumor-supportive role. Using immunohistochemistry to analyze nuclear activated p38 expression in 90 lung adenocarcinomas, we observed p38 expression was significantly associated with well differentiated compared to poorly-differentiated carcinomas (p & lt;0.001) and better patient outcome (p=0.019, disease-free survival; p=0.03, overall survival). In vitro p38 suppression in lung cancer cell lines did not lead to enhanced cell death. On the other hand, p38 suppression alone or in combination with EGFR inhibition led to increased CD44high/ALDHhigh lung tumor initiating cells (TIC); isolated lung TIC also showed higher p38 expression. Together, the data implicate p38 plays a tumor suppressive role in lung cancer by driving tumor cell differentiation. Disrupting pathways that inhibit p38 expression or enhancement of p38 expression might be useful for lung cancer treatment. Citation Format: Jing Liu, Lai-han Leung, Zhi-jie Xiao, Vicky PC Tin, Maria Pik Wong. P38MAPK plays a tumor suppressive role through differentiation of ALDHhigh/CD44high tumor iniating cells in primary human lung adenocarcinoma. [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 1949. doi:10.1158/1538-7445.AM2014-1949
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. LB-251-LB-251
    Abstract: The cancer stem cell (CSC) theory suggests that cancer is maintained by a subpopulation of cancer cells which possesses stem cell characteristics, including self renewal, tumor initiation and pluripotent differentiation abilities. A large series of CSC markers has been reported in various cancer types. CD133 is the most frequently used marker and has been applied to isolate cancer stem cells from cancers of the liver, brain, colon and lung, etc. CD44+/CD24-/low were used as CSC markers in breast cancers. CD34 and Sca-1 were used for identification of murine lung stem cells but Sca-1 is not expressed in human tissues. Expression of embryonic genes such as BMI1 and POU5F1 were found in CSC from different tissues and both proteins are key components in maintaining ‘stemness’ of embryonic stem (ES) cells and induced-pluripotent stem (iPS) cells. It still remains unclear whether additional markers are needed to detect lung CSC. To our knowledge, limited studies using CSC markers on a panel of non-small cell lung cancer (NSCLC) cell lines including those raised from Chinese lung cancer patients have been documented. This study aimed to examine the expression profiles of 6 CSC markers (CD24, CD34, CD44, CD133, BMI1 and POU5F1) in a panel of 10 NSCLC cell lines, 163 NSCLC tumor specimens, fetal lung, reactive and regenerating lung tissues. NSCLC cell lines showed expression of CSC markers at various frequencies that ranged from 0-99.7%, suggesting absence of a single universal marker. Six cell lines expressed CD44 while only HCC1833 expressed CD133. Expression of CD44 and CD133 were mutually exclusive. The CD133+ and CD44+ populations showed distinct properties from CD133- and CD44- cells, respectively. Both CD133+ and CD44+ expanded into tumor-spheroids in vitro in non-adherent medium but not CD133- and CD44- cells. CD44+ H1299 cells expressed ‘stemness’ markers including Oct4, Nanog and Sox2 by RT-PCR and immunofluorescence but were lost after induced-differentiation. The clonogenicity of CD44+ cells is significantly higher than CD44- cells and self-renewal ability was maintained after serial passages. CD44+ cells were cisplatin-resistant and initiated tumor formation in nude mice. The xenograft tumors showed poor differentiation that resembled the parental cell line. Freshly sorted tumor cells showed both CD44+ and CD44- populations, suggesting in vivo differentiation. Immunohistochemistry (IHC) results showed CD44 expression in epithelia of developing airways and regenerating lung. In lung cancers, CD44 expression was associated with squamous cell carcinoma (SCC) (P=0.046). When only 96 adenocarcinomas (AD) were included, CD44 expression was associated with non-smokers (P=0.024). Overall, CD44+cells in NSCLC are capable of tumor-initiation and possess stem cell characteristics. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr LB-251.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3193-3193
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3193-3193
    Abstract: Lung cancer is the leading cause of cancer death worldwide. Non-small cell lung cancer (NSCLC) accounts for over 80% of all the lung cancer cases. The application of advanced gene sequencing technology has identified several specific driver genetic mutations in NSCLC, such as EGFR, KRAS and EML4-ALK etc. The most common types of EGFR activating mutation are L858R substitution mutation on exon 21 and in-flame deletion mutation on exon 19. It leads to the high demand of using small molecule tyrosine kinase inhibitors specific to these mutations as a new anti-cancer strategy. For example, Gefitinib is used for treating NSCLC with EGFR activating mutation. Despite the great achievement in the initial response of Gefitinib treatment, drug resistance remains an unresolved problem. The discovery of new inhibitors or drugs targeting the molecules responsible to Gefitinib resistant is urgent needed. The application of natural products to achieve therapeutic properties has been tried for a long time along human civilization. Plants, animals and mineral products are the main sources of the natural drugs. In this study, we have found a single compound, designated as MUST-1, which is purified from a Chinese medicine herb, is a potential anti-cancer compound in Gefitinib resistant NSCLC. MUST-1, which is a natural alkaloid, is isolated from a commonly used Chinese medicinal herb, which has been utilized clinically to treat inflammatory diseases, anti-microbe, anti-inflammation and anti-oxidation functions traditionally. Quantitative analysis using flow cytometer of the control and the MUST-1-treated cells after Annexin V/PI staining showed that MUST-1 significantly induced apoptosis in NSCLC cells after 24 hours treatment, the effect is especially effective in those NSCLC cell lines with EGFR mutation(s) and resisted to Gefitinib treatment. Western blot analysis demonstrated that MUST-1 initially activated c-Jun-N-terminal kinase (JNK) and p38 phosphorylation, triggered EGFR degradation and suppressed the EGFR downstream anti-apoptotic AKT signaling. Moreover, NF-κB pathway suppression was also observed. JC-1 staining was used to prove the change in mitochondrial membrane potentials, our result showed that mitochondrial disruption plays an important role in the MUST-1-mediated apoptosis. Further Western blot analysis showed that Caspases-3/7 was activated during apoptosis. Treatment with pan-caspases inhibitor can attenuate the apoptotic effect of MUST-1, suggesting that apoptosis induced by MUST-1 is Caspases-dependent. Overall, our data suggest that MUST-1 is particularly effective in Gefitinib-resistant NSCLC. It may potentially be developed as an alternative treatment option for the NSCLC patients who have developed Gefitinib resistance. Citation Format: Xing-Xing Fan, Maria Pik Wong, Zhi-Wei Cao, Na Li, Jin-Lin Wu, Hua Zhou, Zhi-Hong Jiang, Liang Liu, Elaine Lai-Han Leung. Apoptotic effect of a single compound derived from natural product in Gefitinib-resistant non-small cell lung cancer cells. [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 3193. doi:10.1158/1538-7445.AM2014-3193
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2003-2003
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2003-2003
    Abstract: Lung cancer development is often mediated through oxidative and inflammatory stresses caused by tobacco and environmental carcinogens, suggesting dysregulation of stress mediators might play a role during carcinogenesis. The p38 MAPK pathway is activated by multiple stress stimuli and inflammatory cytokines, and mediates a wide range of cellular processes in a context-specific manner. P38α, a member of the p38 family, is known to regulate the self-renewal and differentiation of adult lung stem cells. While p38α has been reported to mediate tumor suppressive or supportive roles in different cancers, inactivation of p38α in the lung epithelium is involved in KRASG12V induced tumorigenesis. However, whether p38α could be involved in regulating tumor initiating cells (TIC) of lung cancer remains unclear. To investigate this possibility, immunohistochemistry was first used to analyse the nuclear expression of phosphorylation-activated p38α (p-p38α) in 90 human primary lung adenocarcinomas. Results showed higher expression of p-p38α was significantly correlated with longer progression free and overall survivals, as well as a more differentiated tumor histological phenotype, indicating p38α might play a tumor suppressive role and drive tumor differentiation in lung cancer. To further address its functional role on lung TIC, p38α was suppressed by siRNA or pharmacologically inhibited by SB203580. This led to increase of the TIC subset that coexpresses ALDH and CD44 (ALDH+/CD44+-TIC), as well as up-regulated expression of the pluripotency genes SOX2, OCT4, and NANOG in lung cancer cell lines. Functionally, p38α inhibition by SB203580 promoted tumor spheres formation for two consecutive generations. NFAT is a family of transcription factors reported to regulate TIC in various cancers which are activated via de-phosphorylation leading to nuclear translocation. We hypothesize the TIC supportive function of p38α inhibition might be mediated through loss of phosphorylation and subsequent activation of NFATc4. In line with this hypothesis, we showed, using Western blot, p38α inhibition increased the expression of nuclear NFATc4. Moreover, NFATc4 knockdown led to suppressed sphere formation and pluripotency genes expression in lung cancer cell lines. Together, our results indicated p38α mediates a tumor suppressor pathway through downstream NFATc4 inactivation and TIC inhibition. The p38α -NFATc4 stress pathway might serve as a potential target for long term lung cancer treatment through TIC regulation. Citation Format: Zhi-Jie Xiao, Jing Liu, Si-Qi Wang, Xu-Yuan Gao, Vicky Pui-Chi Tin, Maria Pik Wong. P38α suppresses tumor initiating cell phenotypes through inhibition of NFATc4 in non-small cell lung cancer [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 2003.
    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 ...
  • 9
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 8, No. 8 ( 2009-08-01), p. 2142-2151
    Abstract: Epidermal growth factor receptor (EGFR) mutations are common in lung adenocarcinomas, especially from nonsmoking women of Asian descent. We have previously shown EGFR mutations occur in & gt;70% of lung adenocarcinoma from nonsmokers in our population with a complex mutational profile, including 13% of EGFR double mutations. In this study, we investigated the in vitro gefitinib response of four EGFR double mutants identified in untreated patients, including Q787R+L858R, E709A+G719C, T790M+L858R, and H870R+L858R. The phosphorylation profiles of EGFR and downstream effectors AKT, STAT3/5, and ERK1/2 were compared by immunoblot analyses among the single and double mutants transfected into H358 cells. Results showed that mutants responded to in vitro gefitinib treatment with different sensitivities. The G719C and L858R single mutants showed the highest gefitinib sensitivity compared with the corresponding coexisting single mutants E709A, Q787R, H870R, and T790M. The double mutants E709A+G719C, Q787R+L858R, and H870R+L858R showed attenuated responses to gefitinib in the EGFR and downstream effector phosphorylation profiles compared with G719C or L858R alone. T790M+L858R showed strong resistance to gefitinib. Clinically, the patient whose tumor contained H870R+L858R showed tumor stabilization by 250 mg oral gefitinib daily but cerebral metastasis developed 6 months later. Correlation with the in vitro phosphorylation profile of H870R+L858R suggested that treatment failure was probably due to inadequate suppression of EGFR signaling by the drug level attainable in the cerebrospinal fluid at the given oral dosage. Overall, the findings suggested that rare types of EGFR substitution mutations could confer relative gefitinib resistance when combined with the common activating mutants. [Mol Cancer Ther 2009;8(8):2142–51]
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 3034-3034
    Abstract: Personalized therapy is becoming a dominant cancer therapeutic strategy, Epidermal Growth Factor Receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been developed to treat non-small-cell lung cancer (NSCLC) patients with EGFR mutation but they only inhibit EGFR phosphorylation, acquired-resistance still remains unavoidable without EGFRT790M removal. Here, we report a new treatment strategy to overcome such drug resistance by selectively inducing EGFRT790M degradation via specific stimulation of methionine 790 (M790) oxidation. The basal reactive oxygen species (ROS) levels in EGFRT790M containing gefitinib-resistant NSCLC cell lines were substantially high and 63 clinical lung tumors showed higher NADPH oxidase 2 (NOX2) expression than 61 normal lung tissues which may contribute to high basal ROS in cancer. Interestingly, only NOX3 was upregulated by sanguinarine, a pharmacological agent to elevate ROS, resulted in EGFR over-oxidation and degradation as well as apoptosis. By contrast, such responses were lacking in EGFRWild-type (WT) cells, suggesting a new promising drug development strategy with wide therapeutic window between EGFRT790M and EGFRWT. Selective EGFRT790M degradation was manipulated by REDOX imbalance between NOX3 and methionine reductase (MsrA). When NOX3 was activated, NADPH, the key substrate of MsrA, was depleted to generate excessive ROS, resulting in inhibition of MsrA physiological reduction function on oxidized-methionine, thus localized methionine 790 oxidation of EGFRT790M occurred. Furthermore, the in vivo tumor suppression effect of sanguinarine, NOX3 upregulation and EGFR degradation were confirmed. These findings indicate that specific inducing EGFRT790M degradation via manipulating REDOX imbalance between NOX3 and MsrA would be promising strategy for treating gefitinib-resistant NSCLC. Citation Format: Elaine Lai-Han Leung, Xing-Xing Fan, Maria Pik Wong, Zhi-Hong Jiang, Zhong-Qiu Liu, Xiao-Jun Yao, Lin-Lin Lu, Yan-Ling Zhou, Li-Fong Yau, Vicky Pui-Chi Tin, Liang Liu. Overcoming gefitinib resistance in non-small-cell lung cancer by inducing epidermal growth factor receptor (EGFR) degradation via methionine 790 oxidation. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3034.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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...