Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    In: Molecular Therapy, Elsevier BV, Vol. 29, No. 10 ( 2021-10), p. 2963-2978
    Type of Medium: Online Resource
    ISSN: 1525-0016
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2001818-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: OncoImmunology, Informa UK Limited, Vol. 5, No. 11 ( 2016-11), p. e1234571-
    Type of Medium: Online Resource
    ISSN: 2162-402X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2016
    detail.hit.zdb_id: 2645309-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Tumori Journal, SAGE Publications, Vol. 75, No. 5 ( 1989-10), p. 429-434
    Abstract: The growth patterns of two murine and eight human tumors, bilaterally implanted into subcutaneous tissue of groups of recipient mice, were studied. A Gompertz equation was fitted to experimental data for each individual implant and the Gompertz parameters were utilized as quantitative growth characteristics. The relative roles of the tumor-implanted flank (right versus left), of the individual host and of the tumor were analyzed by the paired t-test, simple linear regression model, one-way and two-way analysis of variance. Sixty pairs of Gompertz curves were obtained in seventy animals. Heterogeneity was the main characteristic of the growth pattern in all tumors under study, with a wide variability among the Gompertz parameters. Statistical analysis of experimental data showed that only the tumor systematically influenced the growth characteristics, whereas neither the tumor-implanted flank nor the individual host played a significant role. These results have both theoretical and practical implications.
    Type of Medium: Online Resource
    ISSN: 0300-8916 , 2038-2529
    Language: English
    Publisher: SAGE Publications
    Publication Date: 1989
    detail.hit.zdb_id: 280962-X
    detail.hit.zdb_id: 2267832-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4719-4719
    Abstract: Deregulated c-Myc expression plays a critical role in the pathogenesis and progression of aggressive B-cell non-Hodgkin lymphomas (B-NHLs) and multiple myeloma. Chromosomal translocations juxtaposing c-Myc to immunoglobulin gene regulatory elements are virtually invariant, early transforming events in Burkitt's lymphoma (BL), thus constituting the prime example of c-Myc-driven oncogenesis. The key role of c-Myc-mediated epigenetic changes for its oncogenic gene expression reprogramming makes histone deacetylases (HDAC) inhibitors novel suitable drug candidates for the treatment of c-Myc-driven lymphomas. Furthermore, an increasing body of evidence is unraveling the ability of these inhibitors to down-regulate c-Myc expression itself in tumor cells. Therefore, we studied the potential antitumor activity of ITF2357, a new-generation pan-HDACs inhibitor, in human BL pre-clinical models. ITF2357 boosted histone and non-histone protein acetylation in Namalwa and Raji cells, and induced G1 cell-cycle arrest followed by apoptosis especially in the Namalwa model. Treated cells displayed a significant decrease of c-Myc protein but not mRNA levels. This effect was only partially explained by the significant up-regulation of let-7a and miR-26a - two microRNA that negatively control c-Myc expression - since it was not as persistent as c-Myc down-regulation. Given tumor cell addiction to cap-dependent translation for the constitutive expression of short-lived oncoproteins, including c-Myc, we investigated whether this mechanism was also affected by ITF2357. Akt and PIM kinases are the best-defined activators of cap-dependent protein translation initiation via 4E-BP1 phosphorylation and derepression of the translation factor eIF4E. We found that 4E-BP1 and eIF4E phosphorylation as well as phosphorylated-Akt and PIM kinases were significantly down-regulated in the treated cells. Since mTOR and PIM kinase inhibitors display only limited clinical efficacy against B-NHLs, ITF2357 treatment may thus exert a therapeutic advantage, given its ability to concurrently block the redundant oncogenic survival signals that control cap-dependent translation initiation, and potentially overcome the selection of chemoresistance mechanisms. In Raji-xenografted immunodeficient mice, ITF2357-mediated biomolecular effects were translated into 45% tumor shrinkage and complete disease eradication in 70% of the animals when the drug was used as maintenance management after its combination with low-dose cyclophosphamide. Taken as a whole, these findings provide the proof-of-concept for the clinical evaluation of ITF2357 in rational combinations with conventional and unconventional anticancer agents for the treatment of c-Myc-overexpressing and/or cap-translation-dependent NHLs. 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 4719. doi:1538-7445.AM2012-4719
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 65, No. 14 ( 2005-07-15), p. 6388-6393
    Abstract: CpG-oligodeoxynucleotides (CpG-ODN) exhibit potent immunostimulatory activity by binding with Toll-like receptor 9 (TLR9). Based on the finding that TLR9 is highly expressed and functional in pancreatic tissue, we evaluated the antitumor effects of chemotherapy combined with CpG-ODNs in the orthotopic mouse model of a human pancreatic tumor xenograft. Chemotherapy consisted of the maximum tolerated dose of gemcitabine (i.v., 100 mg/kg, q3dx4). CpG-ODNs were delivered (i.p., 20 μg/mouse), weekly, after the end of chemotherapy. CpG-ODNs alone had little effect on tumor growth, whereas gemcitabine alone significantly delayed the median time of disease onset (palpable i.p. tumor) and of bulky disease development (extensive peritoneal tumor burden), but did not enhance survival time. When the gemcitabine regimen was followed by administration of the immunostimulator, development of bulky disease was delayed, survival time was significantly improved (median survival time, 106 days; P & lt; 0.02 versus gemcitabine-treated mice). Autoptic examination showed that tumor spread in the peritoneal cavity was reduced to a greater extent than with gemcitabine alone. All treatment regimens were well-tolerated. The use of nude mice excluded a T cell–mediated immune response, whereas the high pancreatic expression of TLR9 might have contributed to the tumor response. The clear improvement of survival observed in an orthotopic murine model of human pancreatic cancer by the combined use of CpG-ODNs with chemotherapy suggests the promise of this therapeutic regimen in the clinical setting.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2005
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 22 ( 2006-11-15), p. 10976-10982
    Abstract: A series of water-soluble camptothecins obtained by linking a spermidine moiety to the 21-position of the open form through an amidic bond have been tested for their biochemical and biological activities. Growth inhibition assay on the human non–small cell lung cancer carcinoma NCI-H460 cell line revealed that the camptothecin analogues were less potent than topotecan and SN38 after 1 hour of treatment. The potency increased after 72 hours of exposure, being similar to that of reference camptothecins. The analysis of topoisomerase I–mediated DNA cleavage using the purified enzyme indicated that the novel camptothecin analogues retained ability to poison topoisomerase I and displayed the same cleavage pattern of SN38. Persistence of the DNA cleavage was comparable with that of SN38. Stabilization of the cleavable complex was not the result of hydrolysis of the N-C bond between polyamine and the drug because no free camptothecin was recovered at the end of DNA cleavage in presence of IDN5174, the analogue selected for detailed studies. IDN5174 exhibited an antitumor activity comparable with that of topotecan and irinotecan against NCI-H460 tumor xenograft. The pharmacokinetics in mice showed a favorable disposition in tumor tissue with low amount of camptothecin detectable in plasma and tumor (around 5-10%), thus supporting the efficacy of intact IDN5174. In conclusion, we found that IDN5174 maintained the biological and antitumor properties, in spite of lack of the closed E ring. The available results support the interpretation that the polyamine linked at the 21-position may allow a favorable drug interaction in the ternary complex. (Cancer Res 2006; 66(22): 10976-82)
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2006
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1106-1106
    Abstract: Background: Platinum-based treatment of locally advanced non-small cell lung cancer (NSCLC) provides some clinical benefit in controlling local disease but is ineffective on metastatic dissemination. Rising evidence suggests that chemotherapy can promote an inflammatory reaction supportive of metastasis growth.We previously demonstrated that cisplatin treatment of mouse xenografts enriches for the chemoresistant fraction of CD133+CXCR4+ lung cancer metastasis initiating cells (MICs), increasing distant metastasis development. We hypothesize here that the SDF-1/CXCR4 axis, implied in MICs migration and in stromal cells trafficking, could play a critical role in cisplatin-induced pro-metastatic effects. Results: To study the effects of cisplatin in promoting a pre-metastatic niche, tumor-free naïve SCID mice were treated with cisplatin plus/minus peptide R, a novel inhibitor of CXCR4. Cisplatin resulted in a rapid expansion of CCR2+CXCR4+Ly6Chigh inflammatory monocytes (IM) in the bone marrow, concomitantly with their recruitment to murine lungs guided by enhanced endothelial release of SFD-1. Tail-vein injection of H460 human lung cancer cells 72h after cisplatin administration resulted in augmented number of lung metastases enriched in CD133+CXCR4+ MICs. We proposed that the abundance of CXCR4+IM together with increased endothelial permeability caused by cisplatin favors tumor cells extravasations and the expansion of MICs through SDF-1/CXCR4 axis activation. Combination treatment with a CXCR4 inhibitor prevented IM recruitment, MICs expansion and consequently abolished metastasis overgrowth induced by cisplatin. Combination treatment of H460 subcutaneous xenografts in a regimen mimicking clinical setting revealed that cisplatin also caused tumor release of SDF-1 able to trigger local expansion of MICs subset and to recruit CXCR4+tumor associated macrophages involved in tumor cells intravasation. Stromal SDF-1 induced by cisplatin at distant site also co-recruited MICs and CCR2+CXCR4+IM, promoting spontaneous metastasis formation that can be counteracted by CXCR4 blockade. We confirmed in clinical setting that platinum-based neoadjuvant treatment of NSCLC patients significantly increases tumor SDF-1 expression. Moreover, an higher expression of tumor SDF-1 after cisplatin neo-adjuvant treatment was associated with patients' shorter DFS (p=0,0056; Hazards Ratio= 3,1) and poor OS (p=0,029; Hazards Ratio= 3,46). Conclusions: Our data reveal a paradoxical pro-metastatic effect of cisplatin that fosters MIC-IM recruitment and cross-talk via SDF-1/CXCR4 axis activation. A new combination strategy based on CXCR4 inhibition may disrupt these interactions, providing more effective results for lung cancer treatment Citation Format: Giulia Bertolini, Valeria Cancila, Orazio Fortunato, Giuseppe Lo Russo, Monica Tortoreto, Giovanni Centonze, Massimo Milione, Claudio Tripodo, Stefania Scala, Gabriella Sozzi, Luca Roz. Cisplatin-induced activation of SDF-1/CXCR4 axis sustains lung cancer metastasis by promoting co-recruitment of metastasis initiating cells and inflammatory monocytes [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1106.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6726-6726
    Abstract: Desmoplastic small round cell tumor (DSRCT) is a ultra-rare pediatric scarcoma with poor overall survival. This tumor is dependent on the continued expression and activity of its pathognomonic molecular lesion, the EWS-WT1 transcription factor. DSRCT is often treated with multimodal approach of chemotherapy, surgery, and radiotherapy. Given the rarity of the disease, there have not been clinical studies to establish an effective therapeutic regimen. Indeed, the development of fully characterized preclinical models, able to reproduce the molecular characteristics of clinical tumors, appears instrumental for testing novel therapeutic strategies and accelerating the translation of preclinical findings to the clinical practice. In this study we exploited a novel DSRCT patient-derived xenograft (PDX), which reproduces histomorphological, genomic (CNV) and transcriptomic characteristics of the paired clinical tumor, to comparatively assess the activity of cytotoxic and targeted anticancer agents. Anti-tumor effect was moderate for single-agent doxorubicin, pazopanib and larotrectenib [maximum tumor volume inhibition (max TVI): 55-66%], trabectedin had a higher effect (max TVI: 82%) while irinotecan and eribulin almost complete inhibited tumor growth (max TVI: 96% and 98%, respectively). Interestingly, combination of irinotecan with either eribulin or trabectedin resulted in complete responses which were maintained until the end of the experiment for irinotecan + trabectedin. The trabectedin + irinotecan combination markedly reduced the expression of anti-apoptotic proteins and caused caspase-3 cleavage, consistent with an apoptotic response, and also induced the accumulation of phospho-RIP1 (Ser166) and phospho-RIP3 (Ser227), indicating the occurrence of necroptosis, a type of programmed cell death with necrotic morphology. In line with these findings, transcriptomic profile analysis of ex-vivo tumor samples obtained from mice exposed to trabectedin ± irinotecan revealed a reduced expression of the biological pathways related to apoptosis and cell proliferation in tumor exposed to the drug combination. Mechanistically, we found that these effects were mediated, at least in part, by the down-regulation of EWS-WT1 chimeric protein and its downstream targets, as assessed by PCR and western blotting. Overall, this study emphasizes the importance of patient-derived pre-clinical models to explore new treatments in DSRCT and fosters clinical investigation in the activity of irinotecan plus trabectedin, providing a step forward for developing more effective trabectedin-based combinations for DSRCT to be tested in clinical trials. Citation Format: Valentina Zuco, Sandro Pasquali, Monica Tortoreto, Stefano Percio, Valentina Doldi, Marta Barisella, Paola Collini, Gianpaolo Dagrada, Silvia Brich, Patrizia Gasparini, Marco Fiore, Michela Casanova, Anna Maria Frezza, Alessandro Gronchi, Silvia Stacchiotti, Andrea Ferrari, Nadia Zaffaroni. Effectiveness of irinotecan plus trabectedin in a desmoplastic small round cell tumor patient-derived xenograft. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6726.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 6 ( 2011-03-15), p. 1382-1393
    Abstract: Purpose: Heparanase promotes myeloma growth, dissemination, and angiogenesis through modulation of the tumor microenvironment, thus highlighting the potential of therapeutically targeting this enzyme. SST0001, a nonanticoagulant heparin with antiheparanase activity, was examined for its inhibition of myeloma tumor growth in vivo and for its mechanism of action. Experimental Design: The ability of SST0001 to inhibit growth of myeloma tumors was assessed using multiple animal models and a diverse panel of human and murine myeloma cell lines. To investigate the mechanism of action of SST0001, pharmacodynamic markers of angiogenesis, heparanase activity, and pathways downstream of heparanase were monitored. The potential use of SST0001 as part of a combination therapy was also evaluated in vivo. Results: SST0001 effectively inhibited myeloma growth in vivo, even when confronted with an aggressively growing tumor within human bone. In addition, SST0001 treatment causes changes within tumors consistent with the compound's ability to inhibit heparanase, including downregulation of HGF, VEGF, and MMP-9 expression and suppressed angiogenesis. SST0001 also diminishes heparanase-induced shedding of syndecan-1, a heparan sulfate proteoglycan known to be a potent promoter of myeloma growth. SST0001 inhibited the heparanase-mediated degradation of syndecan-1 heparan sulfate chains, thus confirming the antiheparanase activity of this compound. In combination with dexamethasone, SST0001 blocked tumor growth in vivo presumably through dual targeting of the tumor and its microenvironment. Conclusions: These results provide mechanistic insight into the antitumor action of SST0001 and validate its use as a novel therapeutic tool for treating multiple myeloma. Clin Cancer Res; 17(6); 1382–93. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2089-2089
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2089-2089
    Abstract: An increasing number of experimental evidences show that microRNAs can have a causal role in breast cancer tumorigenesis as a novel class of oncogenes or tumor suppressor genes, depending on the targets they regulate. miR-205, down-modulated in breast cancer versus normal breast tissue, was originally associated to absence of vascular invasion and shown to be involved in the EMT. Moreover, we recently demonstrated that miR-205 directly targets HER3 receptor and impairs the Akt-mediated survival pathway, thus inhibiting clonogenic potential of SKBr3 cells and increasing the responsiveness to tyrosin-kinase inhibitors. Considering the striking evidences of a major role of miR-205 as oncosuppressor in breast cancer, we decided to investigate its possible involvement in another tumor subgroup, extremely aggressive and still lacking a specific therapy: triple negative breast tumors. Here we show that re-introduction of miR-205 in the triple negative breast cancer cell line MDA-MB-231 led to a dramatic reduction of both proliferation and clonogenic potential as well as migration capability. Notably, miR-205 also significantly inhibited the in vivo growth in xenograft models obtained injecting stable clones of MDA-MB-231 cells transfected with a miR-205-encoding plasmid or the empty vector. Investigating alternative pathways potentially regulated by miR-205 in this model, we noticed that many of the predicted targets are actually interconnected and related to adhesion and migration processes. Indeed, this microRNA seems to be involved in the control of cell-cell and cell-matrix adhesion regulating the expression of extracellular matrix-related molecules. Among them, we focused on LAMC1, reported to be involved in cell adhesion, differentiation, migration, signaling and metastasis. By Western Blot and Luciferase assay, we demonstrated that miR-205 directly targets LAMC1 in MDA-MB-231 cells. In conclusion, miR-205 exerts a role as oncosuppressor gene also in triple negative breast cancer: it exerts a dramatic inhibition of proliferation and migration in triple negative MDA-MB-231 cell line, at least partially through direct regulation of LAMC1. These evidences not only underline the consistent role of miR-205 in the biology of different breast cancer subgroups, but provide the rational bases for a therapeutic intervention miRs-based. 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 2089.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages