10 research outputs found

    Fulminant Marginal Keratitis Induced by Atezolizumab, a Programmed Death Ligand 1 Inhibitor for Lung Cancer

    Get PDF
    Introduction: With the increasing use of immune checkpoint inhibitors, ocular adverse events have gained attention. We describe a case of atypical keratitis presumably induced by atezolizumab, a programmed cell death ligand 1 inhibitor. Case Presentation: A 73-year-old Japanese woman developed ring-shaped marginal infiltrations with epithelial breakdown of the corneas in both eyes. The patient had advanced small cell lung cancer and had received intravenous carboplatin, etoposide, and atezolizumab. She was treated with topical administration of 0.1% sodium phosphate betamethasone and 0.5% moxifloxacin six times daily. On day 14 following initial presentation, marked reduction of bilateral corneal infiltration was observed. During the succeeding cycles of chemotherapy, marginal keratitis did not recur, and then, the topical steroid was gradually tapered. Conclusions: Cancer immunotherapy, including atezolizumab, may lead to active T-cell recruitment into the cornea, which result in autoimmune corneal keratitis. We believe that this report is informative to both ophthalmologists and oncologists involved in the treatment of patients receiving cancer immunotherapy

    Dual antiplatelet therapy inhibits neutrophil extracellular traps to reduce liver micrometastases of intrahepatic cholangiocarcinoma

    No full text
    The involvement of neutrophil extracellular traps (NETs) in cancer metastasis is being clarified, but the relationship between intrahepatic cholangiocarcinoma (iCCA) and NETs remains unclear. The presence of NETs was verified by multiple fluorescence staining in clinically resected specimens of iCCA. Human neutrophils were co-cultured with iCCA cells to observe NET induction and changes in cellular characteristics. Binding of platelets to iCCA cells and its mechanism were also examined, and their effects on NETs were analyzed in vitro and in in vivo mouse models. NETs were present in the tumor periphery of resected iCCAs. NETs promoted the motility and migration ability of iCCA cells in vitro. Although iCCA cells alone had a weak NET-inducing ability, the binding of platelets to iCCA cells via P-selectin promoted NET induction. Based on these results, antiplatelet drugs were applied to these cocultures in vitro and inhibited the binding of platelets to iCCA cells and the induction of NETs. Fluorescently labeled iCCA cells were injected into the spleen of mice, resulting in the formation of liver micrometastases coexisting with platelets and NETs. These mice were treated with dual antiplatelet therapy (DAPT) consisting of aspirin and ticagrelor, which dramatically reduced micrometastases. These results suggest that potent antiplatelet therapy prevents micrometastases of iCCA cells by inhibiting platelet activation and NET production, and it may contribute to a novel therapeutic strategy

    Gasdermin C Is Upregulated by Inactivation of Transforming Growth Factor β Receptor Type II in the Presence of Mutated Apc, Promoting Colorectal Cancer Proliferation.

    No full text
    Mutations in TGFBR2, a component of the transforming growth factor (TGF)-β signaling pathway, occur in high-frequency microsatellite instability (MSI-H) colorectal cancer (CRC). In mouse models, Tgfbr2 inactivation in the intestinal epithelium accelerates the development of malignant intestinal tumors in combination with disruption of the Wnt-β-catenin pathway. However, no studies have further identified the genes influenced by TGFBR2 inactivation following disruption of the Wnt-β-catenin pathway. We previously described CDX2P-G19Cre;Apcflox/flox mice, which is stochastically null for Apc in the colon epithelium. In this study, we generated CDX2P-G19Cre;Apcflox/flox;Tgfbr2flox/flox mice, with simultaneous loss of Apc and Tgfbr2. These mice developed tumors, including adenocarcinoma in the proximal colon. We compared gene expression profiles between tumors of the two types of mice using microarray analysis. Our results showed that the expression of the murine homolog of GSDMC was significantly upregulated by 9.25-fold in tumors of CDX2P-G19Cre;Apcflox/flox;Tgfbr2flox/flox mice compared with those of CDX2P-G19Cre;Apcflox/flox mice. We then investigated the role of GSDMC in regulating CRC tumorigenesis. The silencing of GSDMC led to a significant reduction in the proliferation and tumorigenesis of CRC cell lines, whereas the overexpression of GSDMC enhanced cell proliferation. These results suggested that GSDMC functioned as an oncogene, promoting cell proliferation in colorectal carcinogenesis. In conclusion, combined inactivation of both Apc and Tgfbr2 in the colon epithelium of a CRC mouse model promoted development of adenocarcinoma in the proximal colon. Moreover, GSDMC was upregulated by TGFBR2 mutation in CRC and promoted tumor cell proliferation in CRC carcinogenesis, suggesting that GSDMC may be a promising therapeutic target

    Tumorigenesis in <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice and histological analysis of tumors.

    No full text
    <p>(A) Body weights of <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> and control (wild-type) mice. Solid line, <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice; dotted line, control mice. Points, means; bars, SDs. (B) Genotyping of <i>Apc</i> and <i>Tgfbr2</i> in tumor, normal jejunum, and colon tissues. (C) Images of dissected terminal ileums and colons of <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice, showing multiple polypoid lesions in the proximal colon. Scale bar, 1 cm. (D) Images of dissected terminal ileum and colon of control (wild-type) mice. Scale bar, 1 cm. il, ileum; ce, cecum; an, anus. The locations of polypoid lesions are indicated by arrowheads. (E) Histological analysis of hematoxylin and eosin (H&E)-stained sections of tumors from <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice showing well-differentiated adenocarcinoma, and higher-power H&E-stained sections of cancer tissue (bottom right). Bar, 100 μm. (F) Histological analysis of H&E-stained sections of normal colon from control (wild-type) mice. Bar, 10 μm. (G) Tgfbr2 immunofluorescence staining of tumors from <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice and <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup> mice. The nuclei are shown in green, and Tgfbr2 is shown in red. upper panel: intact Tgfbr2 was not detected in tumors from <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice; lower panel: Tgfbr2 was detected in tumors in <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup> mice. Bar, 10 μm. (H, I) Immunohistochemical staining for β-Catenin in tumors from <i>CDX2P-G19Cre</i>;<i>Apc</i><sup><i>flox/flox</i></sup>;<i>Tgfbr2</i><sup><i>flox/flox</i></sup> mice. (J) Immunohistochemical staining for β-Catenin in normal epithelium from control (wild-type) mice. (H) Bar, 100 μm. (I, J) Bar, 20 μm.</p

    GSDMC immunohistochemical staining of normal colonic tissues (A, B) and CRC specimens (C, D).

    No full text
    <p>(A), (C) High-magnification image. (B), (D) Low-magnification image. GSDMC was not expressed in normal colonic tissues, whereas GSDMC was diffusely distributed in the cytoplasm of cancer cells. Scale bar 100 μm (E) The expression of <i>GSDMC</i> mRNA was significantly increased in cancer tissues compared with that in normal tissues. Data are shown as box plots. The horizontal lines represent the median score, the bottom and top of the boxes represent the 25th and 75th percentiles, respectively, and the whiskers represent the range of expression level.</p

    GSDMC promoted xenograft tumor growth <i>in vivo</i>.

    No full text
    <p>(A) Quantitative reverse transcription PCR was used to analyze the expression of GSDMC in LoVo cells stably expressing <i>GSDMC</i> shRNA2 or nonsilencing shRNA. (B)–(D) Effects of GSDMC silencing in LoVo cells on the growth of xenograft tumors in nude mice (n = 8). Cells stably expressing <i>GSDMC</i> shRNA2 or nonsilencing shRNA were injected subcutaneously into nude mice. The growth of subcutaneous tumors was measured every 3 days using Vernier caliper, and volume determined by the formula V = 1/2 (length × width<sup>2</sup>). The tumors were then removed from the nude mice and imaged (C). Tumor weights are shown in (D). *<i>p</i> < 0.05, ** <i>p</i> < 0.01</p

    GSDMC promoted tumor growth <i>in vitro</i>.

    No full text
    <p>(A), (B) Quantitative reverse transcription (qRT)-PCR of the expression of <i>GSDMC</i> mRNA after transfection of DLD1 and LoVo cells with siRNAs. (C), (D) MTS assays were used to analyze the effects of <i>GSDMC</i> knockdown on cell proliferation in DLD-1 and LoVo cells for up to 4 days. (E), (F) Colony formation assays in DLD-1 and LoVo cells. Upper panel: number of colonies; lower panel: imaging results. Colonies were monitored for up to 11 days after cell seeding on the plates. (G), (H) Upper panel: qRT-PCR; lower panel: western blotting analysis in SW480 or WiDr cells transfected with pDON-5/GSDMC or pDON-5. (I), (J) MTS assays in SW480 or WiDr cells transfected with pDON-5/GSDMC or pDON-5. Cell growth was monitored up to 6 days. (K), (L) Colony formation assays in SW480 or WiDr cells transfected with pDON-5/GSDMC or pDON-5. Those in WiDr cells did not show significant differences (<i>p</i> = 0.127). Upper panel: number of colonies; lower panel: imaging results. Colonies were monitored for up to 11 days after cell seeding on the plates. Representative data from three independent experiments are shown. *<i>p</i> < 0.05.</p
    corecore