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
  • American Association for Cancer Research (AACR)  (4)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_1 ( 2015-12-01), p. PR01-PR01
    Abstract: Cancer-associated fibroblasts (CAFs) play a central role in tumor progression through the mechanical remodeling of the stroma. Indeed CAFs secrete a plethora of extracellular matrix (ECM) components and ECM modifiers which contribute to generate stiff and dense tumors. Increased tumor stiffness induces endothelial cell (EC) sprouting and tumor cell invasion. Moreover, excessive stiffness represents a critical barrier to therapy because it blocks perfusion thus preventing diffusion of drugs and favoring hypoxia. Tuning tumor stiffness has therefore the potential to contribute to improve the efficacy of conventional anti-cancer therapies. Our study aims at using unbiased proteomics approach to identify CAF proteins which alter the tumor stroma, and investigating their functional role. We have established mass spectrometry-based proteomics approach to accurately and in-depth analyze secretomes of cells in culture, and used it to compare cell lines of human mammary normal (iNF) and myofibroblasts-like cancer-associated (iCAF) fibroblasts. Unexpectedly, we detected the chloride intracellular channel protein 3 (CLIC3), which so far was known as an intracellular regulator of receptor trafficking in tumor cells, amongst the most up-regulated proteins in the ECM generated by iCAF. Highlighting the relevance of CLIC3 in a clinical context, high levels of CLIC3 were also measured in CAFs isolated from patient samples. Notably, CLIC3 silencing reduces the pro-angiogenic and pro-invasive activity of CAFs in vitro. To investigate the role of secreted CLIC3, we have generated the recombinant protein (rCLIC3) and the mutant for the active site (rCLIC3mut) and show that rCLIC3 induces angiogenesis in vitro and in vivo, and tumor cell invasion in vitro and that its active site is required for this activity. Furthermore, we show that rCLIC3 is used by iCAF to increase the stiffness of the ECM that they produce. Using gene silencing and specific inhibitors in combination with functional assays in vitro and in vivo, we show that rCLIC3 exerts its functions through the cooperation with the ECM remodeling enzyme transglutaminase 2 (TGM2). Importantly, TGM2 is highly abundant in many tumors. In a clinical context, CLIC3 staining of various TMAs shows that this protein is expressed in the stroma of some tumors and that it is particularly abundant in the stroma of ovarian cancers. Preliminary analysis of ovarian TMAs shows that the stroma of aggressive and highly vascularized tumors strongly stains for CLIC3. In conclusion, our work has discovered a completely unexpected function for CLIC3, which we have now characterized as a stromal protein which is secreted and alters the ECM stiffness cooperating with TGM2, thus enhancing the pro-angiogenic and pro-invasive properties of the CAFs. CLIC3 is therefore a promising candidate to be further investigated for its role in tumor progression and as a target to improve current anti-cancer therapy. Citation Format: Juan Ramon Hernández-Fernaud, Elena Ruengeler, Andrea Casazza, Lisa Neilson, Ellie Pulleine, Iain MacPherson, Karen Blyth, Huabing Yin, Massimiliano Mazzone, Jim Norman, Sara Zanivan. CLIC3 is secreted by CAFs and enhances angiogenesis and tumor cell invasion by cooperating with TGM2. [abstract]. In: Proceedings of the AACR Special Conference: Tumor Angiogenesis and Vascular Normalization: Bench to Bedside to Biomarkers; Mar 5-8, 2015; Orlando, FL. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl):Abstract nr PR01.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3398-3398
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3398-3398
    Abstract: Semaphorins comprise a wide family of secreted and membrane-bound signals, originally found in neural development, and then implicated in a range of functions: from angiogenesis, to the immune response, to cancer progression. We have previously shown that high-affinity semaphorin receptors are found in the families of the plexins and of the neuropilins. Moreover, additional molecules interact with semaphorin receptors in cell-context dependent manner, featuring a complex scenario of multiple potential signaling pathways. The role of semaphorin signaling in tumor progression is currently under close scrutiny; it is known that cancer cells release semaphorins regulating their own behavior as well as that of cells in the tumor microenvironment, such as endothelial cells and recruited leukocytes. In this work, we focused on the function of Semaphorin 3E (Sema3E) and its high affinity receptor PlexinD1, and on their potential role to regulate cancer progression. Here we found that Sema3E and PlexinD1 are highly expressed in human metastatic tumors. Moreover, we provided experimental evidences that Sema3E released by tumor cells is a double-faced signal, acting via two distinctive signaling pathways. In paracrine manner, Sema3E-PlexinD1 signaling can elicit endothelial cell repulsion, thereby reducing vessel density and tumor growth. Conversely, Sema3E autocrine signaling in tumor cells thrusts their invasiveness, trans-endothelial migration, and metastatic spreading, through the trans-activation of PlexinD1-associated oncogenic kinases. Notably, by silencing the endogenous expression of either Sema3E or PlexinD1 the metastatic potential of cancer cells is significantly reduced, identifying this pathway as a major driver of the metastatic spreading. In sum, Sema3E autocrine signaling in cancer cells is crucially implicated to induce a metastatic behavior, and it appears as a promising target for strategies aimed at blocking tumor metastasis. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3398.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 21, No. 4 ( 2022-04-01), p. 568-581
    Abstract: Clinical use of doxorubicin (Dox) is limited by cumulative myelo- and cardiotoxicity. This research focuses on the detailed characterization of PhAc-ALGP-Dox, a targeted tetrapeptide prodrug with a unique dual-step activation mechanism, designed to circumvent Dox-related toxicities and is ready for upcoming clinical investigation. Coupling Dox to a phosphonoacetyl (PhAc)-capped tetrapeptide forms the cell-impermeable, inactive compound, PhAc-ALGP-Dox. After extracellular cleavage by tumor-enriched thimet oligopeptidase-1 (THOP1), a cell-permeable but still biologically inactive dipeptide-conjugate is formed (GP-Dox), which is further processed intracellularly to Dox by fibroblast activation protein-alpha (FAPα) and/or dipeptidyl peptidase-4 (DPP4). In vitro, PhAc-ALGP-Dox is effective in various 2D- and 3D-cancer models, while showing improved safety toward normal epithelium, hematopoietic progenitors, and cardiomyocytes. In vivo, these results translate into a 10-fold higher tolerability and 5-fold greater retention of Dox in the tumor microenvironment compared with the parental drug. PhAc-ALGP-Dox demonstrates 63% to 96% tumor growth inhibition in preclinical models, an 8-fold improvement in efficacy in patient-derived xenograft (PDX) models, and reduced metastatic burden in a murine model of experimental lung metastasis, improving survival by 30%. The current findings highlight the potential clinical benefit of PhAc-ALGP-Dox, a targeted drug-conjugate with broad applicability, favorable tissue biodistribution, significantly improved tolerability, and tumor growth inhibition at primary and metastatic sites in numerous solid tumor models.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 180-180
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 180-180
    Abstract: Use of traditional chemotherapeutic drugs is restricted by severe side effects and lack of tumor specificity of these cytotoxic agents. Less toxic prodrug that can be selectively activated in tumor tissue have been explored in attempts to improve the therapeutic index. Some approaches to the development of tumor activating prodrugs take advantage of inherent properties of the tumor, for example, selective enzyme expression, hypoxia, or low extracellular pH in the vicinity of the tumor. CoBioRes makes use of capped, tetrapeptidic prodrug of existing chemotherapeutics. Doxorubicin (Dox) was the first chemotherapeutic that was linked to the tetrapeptide sequence and tested experimentally (PhAc-ALGP-Dox). This approach makes the prodrug impermeable to cell membranes of both normal and tumor cells. Furthermore, it remains stable in blood by the use of the capping group on the amino-terminal group of the peptide that prevents aspecific activation by circulating esopeptidases. The prodrug as such is thus not active. The peptide sequences have been developed to be sensitive to the hydrolytic action of a selected group of peptidases that are released within the tumor microenvironment. These peptidases, which accumulate in the tumors, also play an important role in cancer cell invasion and metastasis. What makes this approach unique is that it is not targeting a single enzyme, but requires, for its selectivity, a two-step activation based on enzymes with an increasing selectivity. In vivo activation starts when the tetrapeptide prodrug is exposed to endonucleases (CD10 and THOP1) leading to the formation of intermediates that are still inactive and poorly permeable through cell membranes. This leads to a tissue distribution that is determined by the expression of the endonucleases. Since these are known to be overexpressed in several tumors, and only present at lower level in some normal tissues, the prodrug will be preferentially located and activated in the vicinity of tumors. The second step of the activation is the cleavage at the prolyl by dipeptidases FAP and DPPIV, releasing the free doxorubicin. The simultaneous expression of all the enzymes involved in the prodrug activation was confirmed both in human triple negative breast cancer biopsies and in experimental triple negative breast cancer models. In vitro analysis of PhAc-ALGP-Dox reveals higher tumor cells tropism and higher cytotoxicity in cancer cells than in normal cells. In vivo PhAc-ALGP-Dox gives promising preliminary results in terms of systemic tolerance even at high concentration, and in vivo efficacy in xenograft mouse models compared to free doxorubicin. Taken together, these results provide a strong rationale for further investigation aimed at unleashing the potential clinical value of this compound. Citation Format: Andrea Casazza, Massimiliano Mazzone, Peter Pokreisz. PhAc-ALGP-Dox is a new tumor selective peptide prodrug of doxorubicin that shows improved efficacy and systemic tolerance in triple negative breast cancer models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 180. doi:10.1158/1538-7445.AM2017-180
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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...