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
  • Scott, Ailing  (6)
  • Wang, Ying  (6)
  • Medicine  (6)
  • 1
    In: Gut, BMJ, Vol. 69, No. 1 ( 2020-01), p. 18-31
    Abstract: Peritoneal carcinomatosis (PC) occurs frequently in patients with gastric adenocarcinoma (GAC) and confers a poor prognosis. Multiplex profiling of primary GACs has been insightful but the underpinnings of PC’s development/progression remain largely unknown. We characterised exome/transcriptome/immune landscapes of PC cells from patients with GAC aiming to identify novel therapeutic targets. Design We performed whole-exome sequencing (WES) and whole transcriptome sequencing (RNA-seq) on 44 PC specimens (43 patients with PC) including an integrative analysis of WES, RNA-seq, immune profile, clinical and pathological phenotypes to dissect the molecular pathogenesis, identifying actionable targets and/or biomarkers and comparison with TCGA primary GACs. Results We identified distinct alterations in PC versus primary GACs, such as more frequent CDH1 and TAF1 mutations, 6q loss and chr19 gain. Alterations associated with aggressive PC phenotypes emerged with increased mutations in TP53, CDH1, TAF1 and KMT2C , higher level of ‘clock-like’ mutational signature, increase in whole-genome doublings, chromosomal instability (particularly, copy number losses), reprogrammed microenvironment, enriched cell cycle pathways, MYC activation and impaired immune response. Integrated analysis identified two main molecular subtypes: ‘mesenchymal-like’ and ‘epithelial-like’ with discriminating response to chemotherapy (31% vs 71%). Patients with the less responsive ‘mesenchymal-like’ subtype had high expression of immune checkpoint T-Cell Immunoglobulin And Mucin Domain-Containing Protein 3 (TIM-3), its ligand galectin-9, V-domain Ig suppressor of T cell activation (VISTA) and transforming growth factor-β as potential therapeutic immune targets. Conclusions We have uncovered the unique mutational landscape, copy number alteration and gene expression profile of PC cells and defined PC molecular subtypes, which correlated with PC therapy resistance/response. Novel targets and immune checkpoint proteins have been identified with a potential to be translated into clinics.
    Type of Medium: Online Resource
    ISSN: 0017-5749 , 1468-3288
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2020
    detail.hit.zdb_id: 1492637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1071-1071
    Abstract: Background: The prognosis of gastric adenocarcinoma (GAC) patients with metastases is very poor. Understanding of molecular biology is limited. Hedgehog (Hh) signaling plays an important role in many tumor types and expression of Shh/Gli-1, two major molecules in Hh pathway has been documented in GAC. However, their clinical impact on GAC patients particularly in peritoneal metastasis remains elusive. Methods: Expression of Gli1 and Shh were examined using IHC in tissue microarrays containing more than 500 cases of GAC tissues with clinical annotation. The prognostic variables were determined using univariate and multivariate Cox regression analyses. GAC cell lines, patient-derived peritoneal metastatic cells and novel PDX metastatic model were used to determine the functional role of Shh/Gli-1 in vitro and in vivo. Genetic knockout Gli-1 using LentiCRISPR/Cas9 and Hh inhibitor GDC0449 as well as BET inhibitor were used to test their antitumor activities in GAC cell line and patient-derived cells. Cell proliferation, colony formation, invasion, tumor sphere assays and immunofluorescence were performed to evaluate their functionality and effects of targeted therapy. Results: Both Gli1 and Shh expression are significantly overexpressed in GAC tissue. Among 519 GAC cases, 80.76% and 87.02% were positive for nuclear Gli-1 and cytoplasmic Shh expression respectively, while the strong nuclear expression rate for Gli-1 is 69.56% and 50.10% for Shh. In the univariate Cox analysis, the overall survival was shorter for patients with high Gli-1 (p=0.018) or high Shh expression (p=0.038). In the multivariate cox analysis for both markers, only Gli-1 remained as an independently prognostic for short survival. We also observed high Gli-1 nuclear expression correlated with the presence of lymph node metastasis (p=0.032). Gli1 was highly expressed in most human malignant ascites cells. Interestingly, Gli-1 was significantly upregulated in mouse PDX-ascites cells compared to primary mice tumors. Genetic knockdown Gli-1 or pharmacologically inhibition of Gli-1 by GDC0449 Hh inhibitor or BET inhibitor JQ1 decreased Gli-1 and restored E-cadherin expression and significantly suppressed malignant cell properties and reduced population of cancer stem cells (ALDH1+ or CD133+) in patients' derived metastatic cells. Conclusions: These findings indicate that overexpression of Gli1 and Shh plays an important role in progression of peritoneal metastases of GAC. Targeting Gli1/Hh signaling may provide novel therapeutic strategies for GAC patients with peritoneal metastases. Keywords:Hh pathway, Gli1, Shh, Gastric cancer, metastasis Citation Format: Yan Xu, Yuan Li, Bovey Liu, Melissa Pool Pizzi, Yongxi Song, Kazuto Harada, Ailing Scott, Lang Ma, Jiankang Jin, Xiaochuan Dong, Ying Wang, Brian D. Badgwell, Jeannelyn S. Estrella, Roy-Chowdhuri Sinchita, Fatemeh G. Amlashi, Zhenning Wang, Shumei Song, Jaffer A. Ajani. Overexpression of SHH and GLI1 contributes to poor prognosis and peritoneal metastases in gastric adenocarcinoma [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 1071.
    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 ...
  • 3
    In: Gut, BMJ, Vol. 72, No. 4 ( 2023-04), p. 624-637
    Abstract: Many cancers engage embryonic genes for rapid growth and evading the immune system. SOX9 has been upregulated in many tumours, yet the role of SOX9 in mediating immunosuppressive tumour microenvironment is unclear. Here, we aim to dissect the role of SOX9-mediated cancer stemness attributes and immunosuppressive microenvironment in advanced gastric adenocarcinoma (GAC) for novel therapeutic discoveries. Methods Bulk RNAseq/scRNA-seq, patient-derived cells/models and extensive functional studies were used to identify the expression and functions of SOX9 and its target genes in vitro and in vivo. Immune responses were studied in PBMCs or CD45 + immune cells cocultured with tumour cells with SOX9 high or knockout and the KP-Luc2 syngeneic models were used for efficacy of combinations. Results SOX9 is one of the most upregulated SOX genes in GAC and highly expressed in primary and metastatic tissues and associated with poor prognosis. Depletion of SOX9 in patient-derived GAC cells significantly decreased cancer stemness attributes, tumour formation and metastases and consistently increased CD8 + T cell responses when cocultured with PBMCs/CD45 + cells from GAC patients. RNA sequencing identified the leukaemia inhibitory factor (LIF) as the top secreted molecule regulated by SOX9 in tumour cells and was enriched in malignant ascites and mediated SOX9-induced M2 macrophage repolarisation and inhibited T cell function. Conclusion Epithelial SOX9 is critical in suppressing CD8 + T cell responses and modified macrophage function in GAC through the paracrine LIF factor. Cotargeting LIF/LIFR and CSF1R has great potential in targeting SOX9-mediated cancer stemness, T cell immunosuppression and metastases suggesting the novel combination therapy against advanced GAC.
    Type of Medium: Online Resource
    ISSN: 0017-5749 , 1468-3288
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2023
    detail.hit.zdb_id: 1492637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Gut, BMJ, Vol. 71, No. 3 ( 2022-03), p. 467-478
    Abstract: Gastric adenocarcinoma (GAC) is a lethal disease with limited therapeutic options. Genetic alterations in chromatin remodelling gene AT-rich interactive domain 1A ( ARID1A ) and mTOR pathway activation occur frequently in GAC. Targeting the mechanistic target of rapamycin (mTOR) pathway in unselected patients has failed to show survival benefit. A deeper understanding of GAC might identify a subset that can benefit from mTOR inhibition. Methods Genomic alterations in ARID1A were analysed in GAC. Mouse gastric epithelial cells from CK19-Cre-Arid1A fl/fl and wild-type mice were used to determine the activation of oncogenic genes due to loss of Arid1A. Functional studies were performed to determine the significance of loss of ARID1A and the sensitivity of ARID1A-deficient cancer cells to mTOR inhibition in GAC. Results More than 30% of GAC cases had alterations (mutations or deletions) of ARID1A and ARID1A expression was negatively associated with phosphorylation of S6 and SOX9 in GAC tissues and patient-derived xenografts (PDXs). Activation of mTOR signalling (increased pS6) and SOX9 nuclear expression were strongly increased in Arid1A −/− mouse gastric tissues which could be curtailed by RAD001, an mTOR inhibitor. Knockdown of ARID1A in GAC cell lines increased pS6 and nuclear SOX9 and increased sensitivity to an mTOR inhibitor which was further amplified by its combination with fluorouracil both in vitro and in vivo in PDXs. Conclusions The loss of ARID1A activates pS6 and SOX9 in GAC, which can be effectively targeted by an mTOR inhibitor. Therefore, our studies suggest a new therapeutic strategy of clinically targeting the mTOR pathway in patients with GAC with ARID1A deficiency.
    Type of Medium: Online Resource
    ISSN: 0017-5749 , 1468-3288
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2022
    detail.hit.zdb_id: 1492637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Gut, BMJ, Vol. 70, No. 1 ( 2021-01), p. 55-66
    Abstract: Peritoneal carcinomatosis (PC; malignant ascites or implants) occurs in approximately 45% of advanced gastric adenocarcinoma (GAC) patients and associated with a poor survival. The molecular events leading to PC are unknown. The yes-associated protein 1 ( YAP1 ) oncogene has emerged in many tumour types, but its clinical significance in PC is unclear. Here, we investigated the role of YAP1 in PC and its potential as a therapeutic target. Methods Patient-derived PC cells, patient-derived xenograft (PDX) and patient-derived orthotopic (PDO) models were used to study the function of YAP1 in vitro and in vivo. Immunofluorescence and immunohistochemical staining, RNA sequencing (RNA-Seq) and single-cell RNA-Seq (sc-RNA-Seq) were used to elucidate the expression of YAP1 and PC cell heterogeneity. LentiCRISPR/Cas9 knockout of YAP1 and a YAP1 inhibitor were used to dissect its role in PC metastases. Results YAP1 was highly upregulated in PC tumour cells, conferred cancer stem cell (CSC) properties and appeared to be a metastatic driver. Dual staining of YAP1/EpCAM and sc-RNA-Seq revealed that PC tumour cells were highly heterogeneous, YAP1 high PC cells had CSC-like properties and easily formed PDX/PDO tumours but also formed PC in mice, while genetic knockout YAP1 significantly slowed tumour growth and eliminated PC in PDO model. Additionally, pharmacologic inhibition of YAP1 specifically reduced CSC-like properties and suppressed tumour growth in YAP1 high PC cells especially in combination with cytotoxics in vivo PDX model. Conclusions YAP1 is essential for PC that is attenuated by YAP1 inhibition. Our data provide a strong rationale to target YAP1 in clinic for GAC patients with PC.
    Type of Medium: Online Resource
    ISSN: 0017-5749 , 1468-3288
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 1492637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Gut, BMJ, Vol. 70, No. 12 ( 2021-12), p. 2238-2248
    Abstract: Gastro-oesophageal cancers (GEC) are resistant to therapy and lead to poor prognosis. The cancer stem cells (CSCs) and antiapoptotic pathways often confer therapy resistance. We sought to elucidate the antitumour action of a BCL-2 inhibitor, AT101 in GEC in vitro, in vivo and in a clinical trial. Methods Extensive preclinical studies in vitro and in vivo were carried out to establish the mechanism action of AT101 on targeting CSCs and antiapoptotic proteins. A pilot clinical trial in patients with GEC was completed with AT-101 added to standard chemoradiation. Results Overexpression of BCL-2 and MCL-1 was noted in gastric cancer tissues (GC). AT-101 induced apoptosis, reduced proliferation and tumour sphere formation in MCL-1/BCL-2 high GC cells. Interestingly, AT101 dramatically downregulated genes ( YAP-1/Sox9 ) that control CSCs in GEC cell lines regardless of BCL-2/MCL-1 expression. Addition of docetaxel to AT-101 amplified its antiproliferation and induced apoptosis effects. In vivo studies confirmed the combination of AT101 and docetaxel demonstrated stronger antitumour activity accompanied with significant decrease of CSCs biomarkers (YAP1/SOX9). In a pilot clinical trial, 13 patients with oesophageal cancer (EC) received AT101 orally concurrently with chemoradiation. We observed dramatic clinical complete responses and encouraging overall survival in these patients. Clinical specimen analyses revealed that AT-101 dramatically reduced the expression of CSCs genes in treated EC specimens indicating antitumour activity of AT101 relies more on its anti-CSCs activity. Conclusions Our preclinical and clinical data suggest that AT-101 overcomes resistance by targeting CSCs pathways suggesting a novel mechanism of action of AT101 in patients with GEC.
    Type of Medium: Online Resource
    ISSN: 0017-5749 , 1468-3288
    RVK:
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 1492637-4
    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...