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) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 188-188
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 188-188
    Abstract: Adipose stromal cells (ASC) constitute a significant component of breast stroma. Emerging evidence demonstrates a functional interaction between ASC and breast cancer (BC) cells. Here, we report that stromal discoidin domain receptor 1 (DDR1) is important for ASC-BC communication. Using a DDR1 knockout mouse model, we find that syngeneic mammary tumors grow more slowly in KO mice than wild-type littermate controls. In vitro, loss of DDR1 in stromal vascular fraction (SVF) impairs its ability to stimulate migration and invasion of tumor cells. Furthermore, we show that DDR1-dependent IL-6 production in SVF is responsible for the SVF effect on tumor cells. These results uncover a previously unrecognized role of stromal DDR1 in BC progression and a potential therapeutic target for BC treatment. Citation Format: Xiujie Sun. Regulation of breast cancer progression by stromal DDR1 [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 188.
    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 ...
  • 2
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2018
    In:  The Journal of Immunology Vol. 200, No. 1_Supplement ( 2018-05-01), p. 166.27-166.27
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 200, No. 1_Supplement ( 2018-05-01), p. 166.27-166.27
    Abstract: PD-1/PD-L1 provides a mechanism of immune escape, the blockage of which has reinvigorated interest in the treatment of urothelial cancer. We recently reported on cell-intrinsic PD-L1 effects in melanoma and ovarian cancer, and considered that effects in bladder cancer cells could differ based on distinct mutational landscapes. Using CRISPER/Cas9 methodology, we knockout PD-L1 in the murine bladder cancer cell line MB49 and the human bladder caner cell line RT. We show here that cell-intrinsic MB49 and RT4 cells express PD-L1 and PD-1 that each mediate cell-intrinsic signals. PD-L1 knockout has little effect on cells proliferation in vitro, but significant difference could be found in tumor size when we used MB49 Ctrl or MB49 PD-L1KO cells to challenge C57B16 mice. αPD-L1 and αPD-1 antibodies reduced bladder cancer cell proliferation in vitro demonstrating direct signaling effects. Bladder cancer cell-intrinsic PD-L1 promoted mTORC1 and tumor initiating cell generation similar to melanoma and ovarian cancer cells. By contrast to melanoma and ovarian cancer cells, bladder cancer cell PD-L1 promoted autophagy and had little effect on in vivo immune-independent growth. Base one the functional role, PD-L1 knockout increased proliferation inhibition induced by Rapamycin in both mouse and human bladder cancer cells. Furthermore, we found intrinsic PD-L1 suppress cytokine production, including CXCL10. Corresponding with increased CXCL10 secretion, more CXCR3+ NK cells could be attracted by PD-L1KO cancer cells. Overall, our findings further illustrated the intrinsic role of PD-L1 in bladder cancer development and found PD-1 expression on human bladder cancer cells for the first time.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2018
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2019
    In:  The Journal of Immunology Vol. 202, No. 1_Supplement ( 2019-05-01), p. 195.16-195.16
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 202, No. 1_Supplement ( 2019-05-01), p. 195.16-195.16
    Abstract: PD-L1 expression in both tumor and host cells correlates with antitumor therapeutic efficacy, but the specific contribution of PD-L1 in various cell compartments to antitumor immunity remains to be fully elucidated. Here we show that PD-L1 expression is significantly elevated in human and mouse mature adipocytes versus preadipocytes. When co-cultured with mouse splenocytes, adipocytes reduce αPD-L1 antibody-mediated CD8+T cell activation. Genetic ablation of adipose PD-L1 obliterates, while enforced PD-L1 expression in preadipocytes confers, the immune-inhibitory effect of adipocytes. Pharmacologic inhibition of adipogenesis by the PPARγ antagonist GW9662 reduces adipose PD-L1 expression and enhances the antitumor efficacy of αPD-L1 and αPD-1 immunotherapies in female mice bearing syngeneic melanoma or mammary tumors. Combo treatment with GW9662 and αPD-L1 increased antitumor lymphocytes infiltration versus control or single agent treatment. In diet-induced obese female mice, combo treatment elicited suppressed melanoma growth, although less effectively versus lean females. In contrast to females, melanomas in either lean or obese male mice exhibited no applicable response to combo treatment. More strikingly, obese males lost αPD-L1 single treatment response versus lean males, with lower CD45+and CD3+T cell infiltration in tumors. However, castration in lean males rescued efficacy of combo treatment. These data suggest an antagonistic effect of male hormones in this combination. The potential impact of sex and obesity warrants consideration in future development of immunotherapy-related combination therapy.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2019
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2020
    In:  The Journal of Immunology Vol. 204, No. 1_Supplement ( 2020-05-01), p. 165.24-165.24
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 204, No. 1_Supplement ( 2020-05-01), p. 165.24-165.24
    Abstract: PD-L1 has become a major target in anti-cancer immunotherapy, however the overall response rate still remains relatively low among most types of cancers, notably breast cancer. There is an unmet need to boost efficacy of immune checkpoint blockade therapies. While both tumor and host immune cell-derived PD-L1 are implicated in immune suppressive functions of PD-L1, the exact immunosuppressive contribution of PD-L1 from different host tissues is little studied. Here we show that PD-L1 expression is significantly higher in human breast adipose tissue versus stromal vascular fractions. In vitro adipogenesis of mouse pre-adipocytes significantly up-regulates PD-L1 versus pre-adipocytes. In vitro co-culture shows that adipocyte PD-L1 suppresses T cell activation and response to anti-PD-L1. In an adipocyte-specific knockout (KO) mouse model, we show that syngeneic mammary tumors grow slower in KO than wildtype hosts. Immunophenotyping shows that tumors grown in KO mice have higher CD8+and CD4+ T cell infiltration as well as more CD8+ Prf+ cytotoxic T cells. Tumor tissue RNA-seq analysis reveals that genetic ablation of adipocyte PD-L1 confers a distinct transcriptomic signature of T cell activation and tumor killing. Our current findings uncover a previously unappreciated source of immune suppressive PD-L1 in the breast cancer microenvironment and could inform novel therapeutic strategies through targeting tumor-surrounding adipose tissue for treating breast cancer.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2020
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Georg Thieme Verlag KG ; 2018
    In:  Experimental and Clinical Endocrinology & Diabetes Vol. 126, No. 04 ( 2018-04), p. 213-221
    In: Experimental and Clinical Endocrinology & Diabetes, Georg Thieme Verlag KG, Vol. 126, No. 04 ( 2018-04), p. 213-221
    Abstract: Tumor-associated fibroblasts are the most abundant population in tumor stroma and impact on tumor initiation and progression. However, the biological function of tumor-associated fibroblasts in pituitary adenomas has not been fully elucidated to date. So, this study aims to clarify the function and significance of primary cultured pituitary adenoma-derived tumor-associated fibroblasts on rat pituitary adenoma cells. We identified primary cultured tumor-associated fibroblasts and normal fibroblasts based on the expression of α-smooth muscle actin as well as morphology. Furthermore, we investigated cell biological influences on rat pituitary adenoma cells through indirectly co-culturing tumor-associated fibroblasts with GH3 cells and subcutaneous xenograft model. All sorts of fibroblasts showed positive staining for α-smooth muscle actin. But α-smooth muscle actin and vascular endothelial growth factor highly expressed in invasive pituitary adenoma-derived tumor-associated fibroblasts compared to non-invasive pituitary adenoma-derived tumor-associated fibroblasts and normal fibroblasts. Besides, invasive pituitary adenoma-derived tumor-associated fibroblasts promoted the proliferation of GH3 cells in vitro as well as tumor growth in vivo. Finally, vascular endothelial growth factor was highly expressed in tumor specimens co-injected with invasive pituitary adenoma-derived tumor-associated fibroblasts. Our results suggested that invasive pituitary adenoma-derived tumor-associated fibroblasts displayed apparent growth promotion effects on rat pituitary cells both in vitro and in vivo accompanied by over-expression of vascular endothelial growth factor in invasive pituitary adenoma-derived tumor-associated fibroblasts and tumor specimens.
    Type of Medium: Online Resource
    ISSN: 0947-7349 , 1439-3646
    RVK:
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2018
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. 1035-1035
    Abstract: 1035 Background: For human epidermal growth factor receptor 2 (HER2) positive advanced or metastatic breast cancer, the standard therapeutic strategy is HER2-targeted agents combined with a taxane. This multicenter, single-arm phase 2 trial was designed to assess the efficacy and safety of pyrotinib (a brand-new generation, irreversible anti-HER2 tyrosine kinase inhibitor) plus nab-paclitaxel in patients with HER2-positive advanced or metastatic breast cancer. Methods: This was a multicenter, single-arm, open-label phase 2 trial conducted at seven centers in China (ChiCTR1900023653). Women aged 18-75 years, with histologically or cytologically confirmed HER-2 positive (immunohistochemistry [IHC] 3+ or positive confirmed by fluorescence in situ hybridization) advanced or metastatic breast cancer and with Eastern Cooperative Oncology Group performance score (ECOG PS) of 0-1 were enrolled. Patients with primary resistance to trastuzumab and bone-only metastases were excluded. Eligible patients received pyrotinib (400 mg, po, qd) plus nab-paclitaxel (125 mg/m 2 , iv, day 1/8/15) for each 28-day cycle until disease progression, unacceptable toxicity, consent withdrawal or death. The primary endpoint was objective response rate (ORR), defined as the proportion of patients with complete response (CR) or partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. Secondary endpoints included progression-free survival (PFS), overall survival, safety and quality of life. Results: Between December 2019 and December 2021, 51 patients were enrolled. The median age was 55 years (range 35–72). Twenty-three patients (45.1%) had ECOG PS of 0. Ten patients (19.6%) with metastatic disease had previously received first-line treatment and 28 (54.9%) had received prior treatment with trastuzumab. More than half (29 of 51, 56.9%) had hormone receptor-positive disease. Visceral metastases occurred in 56.9% of the patients (29 of 51) and 26 patients (51.0%) were menopausal. The data cutoff for the present analysis was January 21, 2022. Among 38 evaluable patients, four patients (10.5%) had CR, 27 (71.1%) had PR, six (15.8%) had stable disease, and one (2.6%) had progressive disease. The confirmed ORR was 81.6% (95% CI 65.1-91.7%). The PFS data were still immature. The most common grade ≥3 treatment-emergent adverse events included neutropenia (14 of 51, 27.5%), diarrhea (10 of 51, 19.6%), fatigue (5 of 51, 9.8%) and peripheral neuropathy (4 of 51, 7.8%). Conclusions: Pyrotinib combined with nab-paclitaxel showed a promising antitumor activity with good tolerance in patients with HER2-positive advanced or metastatic breast cancer. Clinical trial information: ChiCTR1900023653.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    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...