18 research outputs found

    Full Wafer Process Control Through Object Detection Using Region-Based Convolutional Neural Networks

    No full text
    International audienceFull wafer measurement techniques are used in the semiconductor industry to acquire information at a large scale to control process variation or detect potential defects. This process usually results in the generation of full wafer images, containing various objects that need to be identified to evaluate their impact on the final product performance. Artificial intelligence is very powerful to automate this identification routine. In this paper, we present the application of Region-based Convolutional Neural Networks (RCNN) for enhanced process control from full wafer images gathered by two industrial metrology equipments

    Sirtuin-1 Activation Controls Tumor Growth by Impeding Th17 Differentiation via STAT3 Deacetylation.

    No full text
    International audienceSirtuin-1 deacetylates proteins and has emerged as a critical regulator of different cellular processes, particularly inflammation. Basal SIRT1 activity was previously found to limit Th9 and enhance Th17 differentiation in mice, but the effect of pharmacological SIRT1 activation on T cell differentiation and antitumor responses remains unclear. Here, we find that SIRT1 pharmacological agonists selectively impede mouse and human Th17 cell differentiation. SIRT1 activation induces STAT3 deacetylation, thus reducing its ability to translocate into the nucleus, bind to Rorc promoter, and induce its transcription. SIRT1 agonists reduce tumor growth in mice by blocking Th17 cell differentiation. In cancer patients, the SIRT1 agonist metformin reduced the frequency of Th17 cells and STAT3 acetylation levels. Altogether, these data underscore that SIRT1 activation impedes Th17 cell differentiation and thereby limits tumor growth and suggest that SIRT1 activators may directly target IL-17A functions

    Accumulation of MDSC and Th17 Cells in Patients with Metastatic Colorectal Cancer Predicts the Efficacy of a FOLFOX-Bevacizumab Drug Treatment Regimen

    No full text
    International audienceHost immunity controls the development of colorectal cancer, and chemotherapy used to treat colorectal cancer is likely to recruit the host immune system at some level. Athough preclinical studies have argued that colorectal cancer drugs, such as 5-fluorouracil (5-FU) and oxaliplatin, exert such effects, their combination as employed in the oncology clinic has not been evaluated. Here, we report the results of prospective immuno-monitoring of 25 metastatic colorectal cancer (mCRC) patients treated with a first-line combination regimen of 5-FU, oxaliplatin, and bevacizumab (FOLFOX-bevacizumab), as compared with 20 healthy volunteers. Before this therapy was initiated, T regulatory cells (Treg), Th17, and granulocytic myeloid-derived suppressor cells (gMDSC) were increased significantly in mCRC, but only a high level of gMDSC was associated with a poor prognosis. Chemotherapy modulated the Treg/Th17 balance by decreasing Treg and increasing Th17 cell frequency by 15 days after the start of treatment. Increased Th17 frequency was associated with a poor prognosis. FOL-FOX-bevacizumab treatment elicited a decrease in gMDSC in 15 of 25 patients and was associated with a better survival outcome. Notably, the gMDSCs that expressed high levels of PD-L1, CD39, and CD73 exerted a robust immunosuppressive activity, relative to other myeloid cells present in blood, which could be reversed by blocking the CD39/CD73 and PD-1/PD-L1 axes. Our work underscores the critical prognostic impact of early modifications in Th17 and gMDSC frequency in mCRC. Furthermore, it provides a clinical rationale to combine FOLFOX-bevacizumab chemotherapy with inhibitors of ATP ectonucleotidases and/or anti-PD-1/PD-L1 antibodies to more effectively treat this disease. (C) 2016 AACR

    Bleomycin exerts ambivalent antitumor immune effect by triggering both immunogenic cell death and proliferation of regulatory T cells.

    Get PDF
    Bleomycin (BLM) is an anticancer drug currently used for the treatment of testis cancer and Hodgkin lymphoma. This drug triggers cancer cell death via its capacity to generate radical oxygen species (ROS). However, the putative contribution of anticancer immune responses to the efficacy of BLM has not been evaluated. We make here the observation that BLM induces immunogenic cell death. In particular, BLM is able to induce ROS-mediated reticulum stress and autophagy, which result in the surface exposure of chaperones, including calreticulin and ERp57, and liberation of HMBG1 and ATP. BLM induces anti-tumor immunity which relies on calreticulin, CD8(+) T cells and interferon-γ. We also find that, in addition to its capacity to trigger immunogenic cell death, BLM induces expansion of Foxp3+ regulatory T (Treg) cells via its capacity to induce transforming growth factor beta (TGFβ) secretion by tumor cells. Accordingly, Treg cells or TGFβ depletion dramatically potentiates the antitumor effect of BLM. We conclude that BLM induces both anti-tumor CD8(+) T cell response and a counteracting Treg proliferation. In the future, TGFβ or Treg inhibition during BLM treatment could greatly enhance BLM anti-tumor efficacy

    BLM induces in vivo Treg accumulation throught TGFb production.

    No full text
    <p>A: Spleens from CT26 tumor-bearing mice, treated with PBS or BLM, were harvested and the cells were analyzed through flow cytometry for Foxp3<sup>+</sup>CD4<sup>+</sup> Treg detection. B: Treg from PBS- or BLM- treated CT26-bearing mice were isolated from spleen and co-cultured with OT-I in presence of SIINFEKL (SII). After 3 days of culture, IFNγ was titrated in the supernatant using ELISA method. C: Schematic representation for D and E panel experiments. D: GFP<sup>+</sup> CD4<sup>+</sup> cells were sorted from CD45.2 FOXP3-EGFP mice, then injected i.v. in CD45.1 mice bearing CT26 tumor. The mice then received PBS, IL-2 or BLM treatment. All mice received EdU injection. One day after treatment, spleens and tumors were collected and the proliferation status of transferred cells was assessed by revealing EdU by flow cytometry. E: CD4<sup>+</sup> CD62L<sup>+</sup> GFP<sup>−</sup> naive T cells were sorted from CD45.2 FOXP3-EGFP mice, and injected i.v. in CD45.1 mice bearing CT26 tumor. The mice then received PBS or BLM treatment. Spleens were harvested and the cells were analyzed through flow cytometry for Treg detection. F: Mice were injected with 1.10<sup>6</sup> CT26 cells i.p. Ten days later mice received PBS or BLM injection. The following day ascites were collected, and TGFβ was assessed using ELISA method. G: CT26 cells were treated with PBS or BLM <i>in vitro</i> for 24 h, then the supernatant was collected and assessed for TGFβ using ELISA method. H: Mice bearing CT26 tumor cells were treated with PBS or BLM. The day after, CT26 tumors were collected and tumor cells were separated from the tumor infiltrating lymphocytes. Tumor cells were stained for LAP and analyzed by flow cytometry. I: GFP<sup>+</sup> CD4<sup>+</sup> cells were sorted from CD45.2 FOXP3-EGFP mice, then cultured <i>in vitro</i> under TCR-stimulating conditions. We added culture supernatant of CT26 treated for 24 h with PBS of BLM. In some wells, blocking anti-TGFβ antibody was added. After three days of culture, the cells were incubated with BrdU for 3 h, and then proceeded to BrdU detection by flow cytometry. J: Same as A, and mice received injection of blocking anti-TGFβ antibody. All data presented are representative of one out of two (panels B, D, E, G and I) or three experiments (panels A, F, H and J). Graphs show mean +/− SEM.</p

    BLM treatment triggers characteristic events of immunogenic cell death.

    No full text
    <p>A: Western blotting analysis of phosphorylated, total eIF2α and β-actin in CT26 treated 24 h with the indicated drugs. B: indicated tumor cells were treated <i>in vitro</i> with different chemotherapeutic drugs for 24 h, then CRT (left panel) or ERp57 (right panel) exposure was detected by flow cytometry. C: tumor cells were treated with BLM, with or without N-acetylcystein (NAC) for 24 h, then CRT (left panel) or ERp57 (right panel) exposure was detected by flow cytometry. C: HMGB1 was titrated with ELISA method in supernatant of CT26 cells treated 24 h with indicated drugs. E: CT26 cells were cultured on glass slides and treated with indicated drugs for 24 h. The cells were then submitted to anti-LC3 (green) and DAPI (blue) labeling and observed under an epifluorescence microscope. F: CT26 cells were treated with indicated drugs for 24 h, then stained using Cyto-ID autophagy detection kit and analysed by flow cytometry. G: ATP was titrated in supernatant of CT26 treated with indicated drugs for 24 h. All data presented are representative of one out of three experiments. B-D, E and F show mean +/− SEM.</p

    BLM in vivo antitumor effect through immune-based mechanisms.

    No full text
    <p>A: Footpad injection of different vaccines was performed: PBS-, DOX- or BLM-treated B16-OVA, or ovalbumin protein. 5 days later, the popliteal lymph nodes were harvested and cells were rechallenged with OVA peptide SIINFEKL. After 3 days of culture, IFNγ was titrated in the supernatant using ELISA method. B: shControl CT26 (left panel) or shCRT CT26 (right panel) were injected in the flank of mice. When the tumor reached about 25 mm<sup>2</sup>, mice were treated with PBS or BLM and tumor growth was monitored with a caliper over time. C and D: CT26 cells were injected in the flank of mice. When the tumor reached about 25 mm<sup>2</sup>, mice were treated with PBS or BLM. Some of the mice received injection of isotype control or depleting anti-CD8 (C) or anti-IFNγ (D). We monitored tumor growth with a caliper over time. All data presented are representative of one out of two experiments. Graphs show mean +/− SEM.</p

    BLM anti-tumor effect is enhanced by Treg or TGFβ depletion.

    No full text
    <p>Five hundred thousand CT26 cells were injected in the flank of mice. When the tumor reached about 25 mm<sup>2</sup>, mice were treated with PBS or BLM. Some of the mice received injections of isotype control antibody or depleting anti-CD4 (A), anti-CD25 (B) or blocking anti-TFGβ (C) antibodies. Tumor growth was monitored with a caliper over time. All data presented are representative of one out of two experiments. Graphs show mean +/− SEM.</p

    BLM has no obvious effect on innate immune cells.

    No full text
    <p>A: Spleens from CT26 tumor-bearing mice, treated with PBS or BLM, were harvested and the cells were analyzed through flow cytometry. The graph shows proportion of different splenocyte populations identified by immunostaining. B: Mice received a single injection of PBS, LPS or BLM. One day later, spleens were collected and the maturation status of dendritic cells defined as CD11c<sup>hi</sup> and MHC-II<sup>+</sup> was assessed by flow cytometry, with CD40 (left panel) and CD86 (right panel) staining. C: We injected CT26 cells in the thigh of mice. When the tumor reached about 25 mm<sup>2</sup>, we treated them with PBS, single or five BLM injections. One day after last injection, inguineal lymph nodes were collected and NK cell activation was analysed through CD69 immunostaining. D: Same as A with further identification of MDSC sub-populations with Ly6G and Ly6C immunostaining. The cells are gated as CD11b<sup>hi</sup> among the live cells. E: MDSC from PBS- or BLM- treated CT26-bearing mice were isolated from spleen and co-cultured with OT-I in presence of SIINFEKL. After 3 days of culture, IFNγ was titrated in the supernatant using ELISA method. All data presented are representative of one out of three experiments. Graphs show mean +/− SEM.</p
    corecore