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) ; 2020
    In:  Cancer Immunology Research Vol. 8, No. 4_Supplement ( 2020-04-01), p. B48-B48
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 4_Supplement ( 2020-04-01), p. B48-B48
    Abstract: PBRM1 is a subunit of a SWI/SNF chromatin remodeling complex, and more than 30% of clear cell RCCs show PBRM1 mutations. The function of PBRM1 in tumor immune microenvironment, tumor prognosis and response to immune checkpoint therapies still remain unclear. Here we found that Pbrm1 knockout in mouse Renca RCC cells decreased the activity of IFNγ-STAT1 signaling pathway and consequently the expression of chemokines that recruit effector CD8+ T cells. Pbrm1 deficient murine RCC tumors showed reduced infiltration of CD8+ T cells, which was associated with less PD-1 expression. Pbrm1 deficient tumors demonstrated longer latency but they were more resistant to anti-PD1 treatment. This study provided mechanistic insights and therapeutic guideline to manage ccRCC associated with PBRM1 inactivation. Citation Format: Xian-De Liu, Wen Kong, Anh Hoang, Xuesong Zhang, Lijun Zhou, Patrick G. Pilie, Sevinj Isgandrova, Margie M. Moczygemba, Eric Jonasch. PBRM1 loss promotes resistance to immunotherapy in RCC [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B48.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 11, No. 1 ( 2020-05-01)
    Abstract: A non-immunogenic tumor microenvironment (TME) is a significant barrier to immune checkpoint blockade (ICB) response. The impact of Polybromo-1 ( PBRM1 ) on TME and response to ICB in renal cell carcinoma (RCC) remains to be resolved. Here we show that PBRM1 / Pbrm1 deficiency reduces the binding of brahma-related gene 1 (BRG1) to the IFNγ receptor 2 ( Ifngr2 ) promoter, decreasing STAT1 phosphorylation and the subsequent expression of IFNγ target genes. An analysis of 3 independent patient cohorts and of murine pre-clinical models reveals that PBRM1 loss is associated with a less immunogenic TME and upregulated angiogenesis. Pbrm1 deficient Renca subcutaneous tumors in mice are more resistance to ICB, and a retrospective analysis of the IMmotion150 RCC study also suggests that PBRM1 mutation reduces benefit from ICB. Our study sheds light on the influence of PBRM1 mutations on IFNγ-STAT1 signaling and TME, and can inform additional preclinical and clinical studies in RCC.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 945-945
    Abstract: PBRM1 is a subunit of SWI/SNF complex that regulates chromatin accessibility for transcription factors, and more than 30% of clear cell renal cell carcinoma (RCC) contain PBRM1 mutations. Nivolumab (anti-PD-1) showed profound and prolonged response only in a subset of patients with advanced RCC. The impact of PBRM1 loss on response to immunotherapy in RCC patients still remains contradictory. It is important to develop immune competent murine RCC models that can precisely isolate the influence of PBRM1 loss on the tumor microenvironment. IFNγ-related gene expression profiles have been reported to predict response to anti-PD-1 blockade. We analyzed the effect of PBRM1 on the IFNγ STAT1 signaling pathway in isogenic murine RCC Renca cells. We observed that Pbrm1 knockout reduced IFNγ-induced STAT1 phosphorylation, and consequently the expression of downstream genes including Irf1, Cxc9 and Stat1 itself. Considering the critical role of CXCL9 in T cell recruitment, we found that Pbrm1 deficiency reduced accessibility to the Cxcl9 promoter by STAT1 and BRG1, the ATP-dependent enzyme subunit of the SWI/SNF complex. Furthermore, a bioinformatics analysis of the Cancer Genome Atlas of human RCC and our murine RCC RNAseq database showed that PBRM1inactivation was associated with reduced expression of an IFNγ-related mRNA profile, including checkpoint genes CTLA4 and PDCD1. These results revealed that PBRM1 loss reduced IFNγ signaling pathway activation and chromatin accessibility by the transcription factor STAT1. Since Pbrm1 deficiency reduced IFNγ-STAT1 activity and consequently Cxcl9 expression, we then compared the immune cell infiltration in both Pbrm1 proficient and deficient Renca tumors in BalB/C mice. As expected, Pbrm1 deficient tumors harbored fewer infiltrating CD3+ and CD8+ T cells, and expressed less PD-1. It has previously been reported that response to immune checkpoint therapy requires pre-existing T cells, and their intrinsic anti-tumor activity potentiated by blocking immune checkpoints. We found that Pbrm1 deficient tumors demonstrated longer latency but more resistance to immunotherapy with PD-1 blockade. We previously showed that antiangiogenic therapy increased T cell infiltration associated with PD-L1 upregulation, and proposed that antiangiogenic agents could promote the response to checkpoint blockade. Here we confirmed that antiangiogenic agent axitinib reduced Pbrm1 proficient tumor growth, and its combination with anti-PD-1 blockade exhibited synergistic efficacy. In contrast, neither axitinib monotherapy nor combination therapy with PD-L1 antibody exerted obvious control on Pbrm1 deficient tumor, indicating patients with PBRM1 functional mutation were more resistant to immunotherapy and antiangiogenic therapy. This study provides mechanistic insights into therapeutic response of PBRM1 mutated RCC. Citation Format: Xiande Liu, Wen Kong, Christine Peterson, Anh Hoang, Xuesong Zhang, Lijun Zhou, Patrick Pilie, Truong Lam, Haifeng Zhu, Sevinj Isgandrova, Margie Moczygemba, Eric Jonasch. PBRM1 loss reduces IFNγ-STAT1 activity and promotes resistance to immunotherapy and antiangiogenic therapy in renal cell carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 945.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...