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
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2007
    In:  Clinical Cancer Research Vol. 13, No. 3 ( 2007-02-01), p. 851-857
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 13, No. 3 ( 2007-02-01), p. 851-857
    Abstract: Purpose: Hepatitis B virus (HBV) reactivation is one unique pathogenesis in Asian carriers with liver toxicity after radiotherapy for hepatobiliary malignancies. This study attempts to delineate the biological mechanism of radiation-induced HBV reactivation. Experimental Design: Primary cultures of hepatocytes (PCC) were prepared from the noncancerous liver tissue removed perioperatively from 12 HBV carriers with hepatocellular carcinoma (HCC). The conditioned medium of irradiated PCCs, HCC, and endothelial cells from patients was transferred to PCCs or HepG2.2.15 cells (a human hepatoblastoma cell line transfected with HBV DNA) before subsequent irradiation. Forty-eight hours after irradiation, HBV DNA was measured by real-time quantitative PCR. Specific cytokines were determined by cytokine array and ELISA analysis. Preradiotherapy and postradiotherapy sera from 10 HBV carriers and 16 non-HBV carriers were analyzed for viral loads and cytokine activities. Results: Radiation induced HBV DNA replication in (a) irradiated PCCs cultured with the conditioned medium from irradiated PCCs (2.74-fold; P = 0.004) and endothelial cells (9.50-fold; P = 3.1 × 10−10), but not from HCCs (1.07-fold), and in (b) irradiated HepG2.2.15 cells (17.7-fold) cocultured with human umbilical vascular endothelial cells. Cytokine assay revealed increased expression of interleukin-6 (IL-6) in conditioned medium from irradiated human umbilical vascular endothelial cells. All 16 patients with liver irradiated had the increased serum IL-6 compared with 3 of 10 patients with irradiation excluding liver (P & lt; 0.001). All nine HBV carriers with liver irradiated had postradiotherapy increases in both HBV DNA and IL-6. Conclusions: Radiation-induced liver toxicity with HBV reactivation is from a bystander effect on irradiated endothelial cells releasing cytokines, including IL-6.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1464-1464
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1464-1464
    Abstract: Over-expression of epidermal growth factor receptor (EGFR) had been correlated with poor prognosis in bladder cancer, but clinical trials using EGFR inhibitors alone in the treatment of bladder cancer did not show definite improvement. On the other hand, radiotherapy is the key element of bladder-preserving protocol for patients with muscle-invasive bladder cancer, and several preclinical data reported the advantage of combing radiation and tyrosine kinase inhibitor against EGFR. It is known that aberrantly activated signal transduction pathways can influence radiosensitivity of cancer cells. Herein, we attempt to demonstrate whether radiation can activate signal transduction pathways in bladder cancer cells, and whether the combined inhibition of such pathways can further enhance radiosensitivity. We used a RTK signaling antibody array to investigate the relative levels of phosphorylated signaling proteins. We detected that not only EGFR but also HER-2 and Akt (Ser473) were activated in T24 human bladder cancer cells after irradiation, and the phenomenon was more prominent in 10Gy dose than in 2.5Gy dose. The increased phosphorylation of EGFR and Her-2 was confirmed by Western blot. Then we performed colony formation assay of T24 and NTUB1 human bladder cancer cell lines for 7 days after treatment combining irradiation and erlotinib (an EGFR inhibitor), lapatinib (a reversible EGFR/Her-2 dual inhibitor) or afatinib (an irreversible EGFR/Her-2 dual inhibitor) in different doses. The combination index (CI) analysis revealed synergy between radiation (2.5Gy and 10 Gy) and afatinib in various doses. For T24 cells with afatinib 100nM, the CI was 0.97 in 2.5Gy and 0.66 in 10Gy. For NTUB1 cells with afatinib 100nM, the CI was 0.92 in 2.5Gy and 0.80 in 10Gy. In contrast, erlotinib or lapatinib showed only minor influence to enhance radiation effect in human bladder cancer cells. Using flow cytometry, the relative distribution of cells among various cell cycle phases were determined in cell lines treated with either DMSO, radiation 2.5Gy, afatinib 200nM, or the combination of radiation and afatinib. When compared with radiation alone, treatment combining radiation and afatinib resulted in accumulation of cells in sub-G1 phase but decrease in G2-M phase. Western blot showed the expression of cleaved poly (ADP-ribose) polymerase (PARP) was increased in the combination group when compared with radiation or afatinib alone. The results implicated that the adding of afatinib might enhance radiation effect by increasing apoptosis in bladder cancer cells. Our data clearly show that both EGFR and Her-2 signaling can be activated after radiation in human bladder cancer cells. Afatinib, an EGFR/Her-2 dual inhibitor, effectively radiosensitizes the bladder cancer cells by enhancing apoptosis. The result provides a basis for future in vivo and human studies. 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 1464. doi:1538-7445.AM2012-1464
    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 ...
  • 3
    In: British Journal of Cancer, Springer Science and Business Media LLC, Vol. 128, No. 9 ( 2023-05-18), p. 1753-1764
    Abstract: Although trimodality therapy resecting tumours followed by chemoradiotherapy is emerged for muscle-invasive bladder cancer (MIBC), chemotherapy produces toxicities. Histone deacetylase inhibitors have been identified as an effective strategy to enhance cancer radiotherapy (RT). Methods We examined the role of HDAC6 and specific inhibition of HDAC6 on BC radiosensitivity by performing transcriptomic analysis and mechanism study. Results HDAC6 knockdown or HDAC6 inhibitor (HDAC6i) tubacin exerted a radiosensitizing effect, including decreased clonogenic survival, increased H3K9ac and α -tubulin acetylation, and accumulated γH2AX, which are similar to the effect of panobinostat, a pan-HDACi, on irradiated BC cells. Transcriptomics of shHDAC6-transduced T24 under irradiation showed that shHDAC6 counteracted RT-induced mRNA expression of CXCL1, SERPINE1, SDC1 and SDC2, which are linked to cell migration, angiogenesis and metastasis. Moreover, tubacin significantly suppressed RT-induced CXCL1 and radiation-enhanced invasion/migration, whereas panobinostat elevated RT-induced CXCL1 expression and invasion/migration abilities. This phenotype was significantly abrogated by anti-CXCL1 antibody, indicating the key regulator of CXCL1 contributing to BC malignancy. Immunohistochemical evaluation of tumours from urothelial carcinoma patients supported the correlation between high CXCL1 expression and reduced survival. Conclusion Unlike pan-HDACi, the selective HDAC6i can enhance BC radiosensitization and effectively inhibit RT-induced oncogenic CXCL1-Snail-signalling, thus further advancing its therapeutic potential with RT.
    Type of Medium: Online Resource
    ISSN: 0007-0920 , 1532-1827
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2002452-6
    detail.hit.zdb_id: 80075-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 35, No. 15_suppl ( 2017-05-20), p. 4042-4042
    Abstract: 4042 Background: The optimal use of the metabolic tumor response measured by 18 fluorodeoxyglucose positron emission tomography (FDG-PET) in the treatment of esophageal cancer is currently unknown. We launched a phase II clinical trial to evaluate the early metabolic response to one-cycle chemotherapy in locally advanced esophageal squamous cell carcinoma (ESCC) patients, who subsequently received neoadjuvant chemoradiation (neo-CRT) followed by surgery. Methods: ESCC patients with stage T3 or N1M0 or M1a (AJCC, 6th edition) were enrolled to receive one-cycle chemotherapy, day 1 and 8 doses of paclitaxel, cisplatin, and 24-hour infusional 5-fluorouracil and leucovorin, followed by paclitaxel/cisplatin- based 40Gy neo-CRT and surgery. FDG-PET was performed at baseline and day 14 of the one-cycle chemotherapy. The primary endpoint is pathological complete response (pCR) to neo-CRT. We hypothesized that early PET responders, defined as 〉 35% reduction of maximum standardized uptake value (SUV max ) from the baseline, would significantly improve pCR. Results: Between Feb 2008 and Mar 2012, 66 patients (M: F = 61: 5) were enrolled. Their clinical stages were: II or III, 56; IVA, 10. Forty seven received surgery. The pCR rate per surgical population was 34.0%. The median progression-free survival (PFS) and overall survival (OS) for the whole study group was 16 months (95% CI 9-27) and 22 months (95% CI 16-40), respectively. A total of53 patients were evaluable for PET response. The early PET response was not associated with high pCR rate or better survivals. However, in an exploratory analysis, the post-chemotherapy SUV max was an independent prognostic factor for pCR, PFS and OS. A predictive model for pCR composed of weight loss and the post-chemotherapy SUV max was established with an AUC of 0.84. Conclusions: Our study failed to validate the predictive value of predefined early PET response to one-cycle chemotherapy for pCR to neo-CRT in locally advanced ESCC patients. However, the FDG-PET SUV max after one-cycle chemotherapy may have prognostic and predictive significance, and may be explored in further studies. Clinical trial information: NCT01034332.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2017
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4429-4429
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4429-4429
    Abstract: Erlotinib is a first-generation EGFR (epidermal growth factor receptor) tyrosine kinase inhibitor and have well-established efficacy in non-small cell lung cancer patients with activating EGFR mutations. Afatinib, on the other hand, is a second-generation EGFR tyrosine kinase inhibitor with anti-HER2 activity and was found to be of benefit to patients with advanced lung adenocarcinoma who failed previous gefetinib or erlotinib. In previous study we have demonstrated the in vitro and in vivo radiosensitising activity of afatinib in a murine bladder cancer model. However, the radiosensitizing effect of afatinib and erlotinib, has never been compared in cancer cells including bladder cancer. We performed RTK (Receptor Tyrosine Kinase) antibody arrays to investigate the relative levels of phosphorylation in T24 human bladder cancer cell line. We found that EGFR and HER2 signals were activated after radiation10 Gy. Afatinib 100 nM suppressed both irradiation-induced EGFR and HER2 signals but erlotinib 100 nM only suppressed irradiation-induced EGFR signal. Clonogenic assay of T24 and NTUB1 human bladder cancer cell lines was then examined. Afatinib (100-500 nM) showed better radiosensitizing effect (radiation dose: 2-10 Gy) than erlotinib (300-1500 nM) in both T24 and NTUB1 cells by significantly decreasing the numbers of colonies 7 days after treatment. The effect is more prominent in high doses of afatinib. Flow cytometry was used to determine the distribution of cells among various cell cycle phases. T24 and NTUB1 cells were treated with vehicle, radiation 2.5 Gy, afatinib 200 nM, erlotinib 200 nM or the combination of radiation and afatinib or erlotinib. When compared with radiation or drug alone, treatment combining radiation and afatinib in T24 cells resulted in a significant increase of cells in sub-G1 phase but the phenomenon was not observed in the combination of erlotinib and radiation. A similar trend was found in NTUB1 cells. The results implicated that afatinib might enhance radiation effect by increasing apoptosis in bladder cancer cells. To study the DNA damage status after treatment, we checked intra-nuclear γH2AX foci by mmunofluorescence microscopy. T24 and NTUB1 cells were treated with vehicle, radiation 2.5 Gy, afatinib 100 nM, erlotinib 100 nM or the combination of radiation and afatinib or erlotinib. Radiation alone significantly increased γH2AX foci and combining radiation with afatinib further increased the foci in both T24 and NTUB1 cells. The enhancement was absent in the combination of radiation and erlotinib. Our data clearly show that afatinib, a new generation EGFR tyrosine kinase inhibitor with anti-HER2 activity, is superior to erlotinib as a radiosensitizer in the treatment of human bladder cancer cells. Citation Format: Yu-Chieh Tsai, Tsung-Fan Tuan, Pei-Yin Ho, Wei-Lin Liu, Liang-Yu Chang, Yeong-Shiau Pu, Ann-Lii Cheng, Jason Chia-Hsien Chen. Comparison of afatinib and erlotinib as radiosensitizing agents in bladder cancer cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4429. doi:10.1158/1538-7445.AM2013-4429
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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...