38 research outputs found

    Expression and prognostic relevance of Cyclophilin A and matrix metalloproteinase 9 in esophageal squamous cell carcinoma

    Get PDF
    AIMS: To guide clinicians in selecting treatment options for esophageal squamous cell carcinoma (ESCC) patients, reliable markers predictive of clinical outcome are desirable. This study analyzed the correlation of cyclophilin A (CypA) and matrix metalloproteinase 9 (MMP9) in ESCC and their relationships to clinicopathological features and survival. METHODS: We immunohistochemically investigated 70 specimens of ESCC tissues using CypA and MMP9 antibodies. Then, the correlations between CypA and MMP9 expression and clinicopathological features and its prognostic relevance were determined. RESULTS: Significant correlations were only found in high level of CypA and MMP9 expression with tumor differentiation and lymph node status. Significant positive correlations were found between the expression status of CypA and that of MMP9. Overexpression of CypA and metastasis were significantly associated with shorter progression free survival times in univariate analysis. Multivariate analysis confirmed that CypA expression was an independent prognostic factor. CONCLUSIONS: CypA might be correlated with the differentiation, and its elevated expression may be an adverse prognostic indicator for the patients of ESCC. CypA/MMP9 signal pathway may be attributed to the malignant transformation of ESCC, and attention should be paid to a possible target for therapy. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1166551968105508

    Elucidation of Resistance Mechanisms to Second-Generation ALK Inhibitors Alectinib and Ceritinib in Non–Small Cell Lung Cancer Cells

    Get PDF
    Crizotinib is the first anaplastic lymphoma kinase (ALK) inhibitor to have been approved for the treatment of non–small cell lung cancer (NSCLC) harboring an ALK fusion gene, but it has been found that, in the clinic, patients develop resistance to it. Alectinib and ceritinib are second-generation ALK inhibitors which show remarkable clinical responses in both crizotinib-naive and crizotinib-resistant NSCLC patients harboring an ALK fusion gene. Despite their impressive activity, clinical resistance to alectinib and ceritinib has also emerged. In the current study, we elucidated the resistance mechanisms to these second-generation ALK inhibitors in the H3122 NSCLC cell line harboring the EML4-ALK variant 1 fusion in vitro. Prolonged treatment of the parental H3122 cells with alectinib and ceritinib led to two cell lines which are 10 times less sensitive to alectinib and ceritinib than the parental H3122 cell line. Although mutations of ALK in its kinase domain are a common resistance mechanism for crizotinib, we did not detect any ALK mutation in these resistant cell lines. Rather, overexpression of phospho-ALK and alternative receptor tyrosine kinases such as phospho-EGFR, phospho-HER3, and phospho-IGFR-1R was observed in both resistant cell lines. Additionally, NRG1, a ligand for HER3, is upregulated and responsible for resistance by activating the EGFR family pathways through the NRG1-HER3-EGFR axis. Combination treatment with EGFR inhibitors, in particular afatinib, was shown to be effective at overcoming resistance. Our study provides new mechanistic insights into adaptive resistance to second-generation ALK inhibitors and suggests a potential clinical strategy to combat resistance to these second-generation ALK inhibitors in NSCLC

    Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression

    No full text
    Abstract Background Estrogen promotes breast cancer development and progression mainly through estrogen receptor (ER). However, blockage of estrogen production or action prevents development of and suppresses progression of ER-negative breast cancers. How estrogen promotes ER-negative breast cancer development and progression is poorly understood. We previously discovered that deletion of cell cycle inhibitors p16Ink4a (p16) or p18Ink4c (p18) is required for development of Brca1-deficient basal-like mammary tumors, and that mice lacking p18 develop luminal-type mammary tumors. Methods A genetic model system with three mouse strains, one that develops ER-positive mammary tumors (p18 single deletion) and the others that develop ER-negative tumors (p16;Brca1 and p18;Brca1 compound deletion), human BRCA1 mutant breast cancer patient-derived xenografts, and human BRCA1-deficient and BRCA1-proficient breast cancer cells were used to determine the role of estrogen in activating epithelial-mesenchymal transition (EMT), stimulating cell proliferation, and promoting ER-negative mammary tumor initiation and metastasis. Results Estrogen stimulated the proliferation and tumor-initiating potential of both ER-positive Brca1-proficient and ER-negative Brca1-deficient tumor cells. Estrogen activated EMT in a subset of Brca1-deficient mammary tumor cells that maintained epithelial features, and enhanced the number of cancer stem cells, promoting tumor progression and metastasis. Estrogen activated EMT independent of ER in Brca1-deficient, but not Brca1-proficient, tumor cells. Estrogen activated the AKT pathway in BRCA1-deficient tumor cells independent of ER, and pharmaceutical inhibition of AKT activity suppressed EMT and cell proliferation preventing BRCA1 deficient tumor progression. Conclusions This study reveals for the first time that estrogen promotes BRCA1-deficient tumor initiation and progression by stimulation of cell proliferation and activation of EMT, which are dependent on AKT activation and independent of ER

    Additional file 1: of Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression

    No full text
    Figure S1. Characterization of mammary tumors developed in mutant mice in Balb/cB6 mixed background. Representative mammary tumors spontaneously developed in p18-/-;Brca1MGKO, p16-/-;Brca1MGKO and p18-/- mice were immunostained with the antibodies indicated. The representative cells are enlarged in the insets

    Additional file 4: of Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression

    No full text
    Figure S4. Estrogen promotes lung metastasis of Brca1-deficient mammary tumors. (A, B) Metastatic p18-/-;Brca1MGKO tumor cells were inoculated into the MFPs of NSG mice with either E2 or placebo supplement. When newly generated tumors reached maximum size allowed by the IACUC in 3–6 weeks, or the mice became moribund, lungs were dissected for analysis. Representative gross pictures (A) and H.E. staining (B) of lungs are shown

    Additional file 7: of Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression

    No full text
    Figure S7. Estrogen stimulates ER-positive cell proliferation that is blocked by 4OHT. MCF-7 cells were treated with DMSO and 5 nM E2 with or without 5 µM 4OHT. The number of viable cells was determined on day 1, day 3, and day 5 (A). Cells treated for 72 h were collected and analyzed by western blot (B); *p

    Additional file 3: of Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression

    No full text
    Figure S3. Estrogen promotes Brca1-proficient and deficient mammary tumor initiation (A) Western blot analysis of mammary tumors regenerated by p18-/- or p18-/-;Brca1MGKO tumor cells with E2 supplement. (B) Representative gross pictures of p18-/- and p16-/-;Brca1MGKO tumors generated by transplantation. We transplanted 1 x 107 p18-/- or 6 x 104 p16-/-;Brca1MGKO tumor cells into MFPs of NSG mice with or without E2 supplement. Gross pictures were taken 6-7 weeks post-transplantation. (C) Representative H.E. staining of primary p18-/- tumors and tumors generated by p18-/- tumor cells with E2 supplement. Note the well-differentiated cells with glandular structure in both primary and regenerated tumors. (D) Representative H.E. staining of p16-/-;Brca1MGKO tumors generated in the presence or absence of E2 supplement. Note the poorly differentiated cells with increased fibroblast-like cells in the tumors with E2 treatment. Spindle cells (black arrows), cells with high nuclear-cytoplasm ratio (green arrows), mitotic cells (red arrows), and necrosis (yellow arrows) are indicated

    Additional file 2: of Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression

    No full text
    Figure S2. p18-/-;Brca1MGKO and p16-/-;Brca1MGKO tumor cells generate reproducible ER-negative Brca1-deficient mammary tumors. (A) A primary mammary tumor (donor tumor A) developed in a 15-month-old p16-/-;Brca1MGKO mouse was analyzed by FACS. As a control, tumor-free mammary glands from age-matched p16-/- mice were analyzed. Note a predominant CD24?+?CD29high population in the tumor. (B, C) We transplanted 6?×?104 FACS-sorted Lin- cells from a p16-/-;Brca1MGKO tumor (donor tumor B) into MFPs of four NSG mice. Representative tumors generated were analyzed by IHC (B) and western blot (C). (D, E) Representative p18-/- and p18-/-;Brca1MGKO tumor cells were cultured and analyzed. MCF7 cells were used as a positive control of ERa expression. (F, G) We transplanted 1?×?106 cultured p18-/-;Brca1MGKO tumor (donor tumor A) cells into MFPs of four NSG mice. Representative tumors generated were analyzed by IHC (F) and western blot (G). p18-/- tumors were used as control in (C) and (G)

    Tumor location as a novel high risk parameter for stage II colorectal cancers

    No full text
    <div><p>Current studies do not accurately evaluate the influence of tumor location on survival of colorectal cancer patients. This study aimed to explore whether tumor location could be identified as another high-risk factor in stage II colorectal cancer by using data identified from the Surveillance, Epidemiology, and End Results database. All colorectal cancer patients between 2004 and 2008 were grouped into three according to tumor location. Of 33,789 patients diagnosed with stage II colorectal cancer, 46.8% were right colon cancer, 37.5% were left colon cancer and 15.7% were rectal cancer. The 5-year cancer specific survivals were examined. Right colon cancer was associated with the female sex, older age (> 50), and having over 12 lymph nodes resected. Conversely, rectal cancer was associated with the male sex, patients younger than 50 years of age and insufficient lymph node resection. The characteristics of left colon cancer were between them and associated with Asian or Pacific Islander populations, T4 stage, and Grade II patients. The prognostic differences between three groups were significant and retained after stratification by T stage, histological grade, number of regional nodes dissected, age at diagnose, race and sex. Furthermore, the significant difference of location was retained as an independent high-risk parameter. Thus, stage II colorectal cancers of different locations have different clinic-pathological features and cancer-specific survivals, and tumor location should be recognized as another high-risk parameter in stage II colorectal cancer.</p></div
    corecore