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
    In: Genes & Development, Cold Spring Harbor Laboratory, Vol. 27, No. 19 ( 2013-10-01), p. 2086-2098
    Abstract: Serine and cysteine cathepsin (Cts) proteases are an important class of intracellular and pericellular enzymes mediating multiple aspects of tumor development. Emblematic of these is CtsB, reported to play functionally significant roles during pancreatic islet and mammary carcinogenesis. CtsC, on the other hand, while up-regulated during pancreatic islet carcinogenesis, lacks functional significance in mediating neoplastic progression in that organ. Given that protein expression and enzymatic activity of both CtsB and CtsC are increased in numerous tumors, we sought to understand how tissue specificity might factor into their functional significance. Thus, whereas others have reported that CtsB regulates metastasis of mammary carcinomas, we found that development of squamous carcinomas occurs independently of CtsB. In contrast to these findings, our studies found no significant role for CtsC during mammary carcinogenesis but revealed squamous carcinogenesis to be functionally dependent on CtsC. In this context, dermal/stromal fibroblasts and bone marrow-derived cells expressed increased levels of enzymatically active CtsC that regulated the complexity of infiltrating immune cells in neoplastic skin, development of angiogenic vasculature, and overt squamous cell carcinoma growth. These studies highlight the important contribution of tissue/microenvironment context to solid tumor development and indicate that tissue specificity defines functional significance for these two members of the cysteine protease family.
    Type of Medium: Online Resource
    ISSN: 0890-9369 , 1549-5477
    RVK:
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2013
    detail.hit.zdb_id: 1467414-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 6, No. 3 ( 2016-03-01), p. 270-285
    Abstract: Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell–dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ+ tumor–associated macrophages, resulting in TH2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a TH1 phenotype that fostered CD8+ T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. Significance: We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell–dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer. Cancer Discov; 6(3); 270–85. ©2015 AACR. See related commentary by Roghanian et al., p. 230. See related article by Pylayeva-Gupta et al., p. 247. See related article by Lee et al., p. 256. This article is highlighted in the In This Issue feature, p. 217
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Elsevier BV ; 2010
    In:  Cytokine & Growth Factor Reviews Vol. 21, No. 1 ( 2010-2), p. 3-10
    In: Cytokine & Growth Factor Reviews, Elsevier BV, Vol. 21, No. 1 ( 2010-2), p. 3-10
    Type of Medium: Online Resource
    ISSN: 1359-6101
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2025966-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Elsevier BV ; 2012
    In:  Trends in Immunology Vol. 33, No. 3 ( 2012-3), p. 119-126
    In: Trends in Immunology, Elsevier BV, Vol. 33, No. 3 ( 2012-3), p. 119-126
    Type of Medium: Online Resource
    ISSN: 1471-4906
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2012
    detail.hit.zdb_id: 2040190-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4391-4391
    Abstract: Using the K14-HPV16 mouse model of squamous carcinogenesis (SCC), we previously reported that B cells foster neoplastic progression through deposition of immunoglobulin complexes in premalignant tissue via Fcα receptor-dependent activation of recruited myeloid cells. Here we evaluated therapeutic interventions targeting these pathways in preclinical trials through administration of depleting αCD20 antibody and small molecule inhibitor of Syk kinase. Both approaches harbored efficacy in preventing premalignant progression to the dysplastic/carcinoma in situ state. Screening a diverse spectrum of human solid tumors revealed SCCs of the vulva, head and neck, as well as pancreatic ductal adenocarcinomas (PDAC) as scoring positively for “signatures” of B cell or plasma cell infiltration, i.e. Ig or CD20 mRNA expression, thereby identifying carcinomas potentially amenable to anti-B cell therapies. Accordingly, B cell-deficient mice failed to support growth of either transplantable orthotopic SCC or PDAC. While administration of αCD20 mAB as a single agent was inefficient in impeding growth of preexisting SCCs, when delivered in combination with cytotoxic chemotherapy, e.g., paclitaxel, carboplatin or cisplatin, αCD20 mAb significantly improved chemotherapeutic response and improved survival by a mechanism dependent on CD8+ T cells. These data reveal that blocking protumorigenic programs regulating by humoral immunity, in combination with chemotherapy, effectively reprograms the tumor immune microenvironment and improves outcome. The authors acknowledge generous support from the NIH/NCI (R01CA130980, R01CA13256, R01CA140943, R01CA15531), the Department of Defense (W81XWH-09-1-0342, W81XWH-10-BCRP-EOHS-EXP) and the Susan G Komen Foundation (KG111084) Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4391. doi:1538-7445.AM2012-4391
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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...