feed icon rss

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: The Oncologist, Oxford University Press (OUP), Vol. 23, No. 1 ( 2018-01-01), p. 62-70
    Abstract: Alveolar soft part sarcoma (ASPS) is an exceedingly rare and orphan disease, without active drugs approved in the front line. Pazopanib and trabectedin are licensed for sarcoma treatment from second-line, but very little and contradictory data are available on their activity in ASPS. Lacking ongoing and/or planned clinical trials, we conducted a multi-institutional study involving the reference sites for sarcoma in Europe, U.S., and Japan, within the World Sarcoma Network, to investigate the efficacy of pazopanib and trabectedin. Materials and Methods From May 2007, 14 of the 27 centers that were asked to retrospectively review their databases had identified 44 advanced ASPS patients treated with pazopanib and/or trabectedin. Response was evaluated by Response Evaluation Criteria in Solid Tumors 1.1. Progression-free survival (PFS) and overall survival (OS) were computed by Kaplan-Meier method. Results Among 30 patients who received pazopanib, 18 were pretreated (13 with other antiangiogenics). Response was evaluable in 29/30 patients. Best responses were 1 complete response, 7 partial response (PR), 17 stable disease (SD), and 4 progressions. At a 19-month median follow-up, median PFS was 13.6 months (range: 1.6–32.2+), with 59% of patients progression-free at 1 year. Median OS was not reached. Among 23 patients treated with trabectedin, all were pretreated and evaluable for response. Best responses were 1 PR, 13 SD, and 9 progressions. At a 27-month median follow-up, median PFS was 3.7 months (range: 0.7–109), with 13% of patients progression-free at 1 year. Median OS was 9.1 months. Conclusion The value of pazopanib in advanced ASPS is confirmed, with durable responses, whereas the value of trabectedin appears limited. These results are relevant to defining the best approach to advanced ASPS. Implications for Practice This retrospective study, conducted among the world reference centers for treatment of sarcoma, confirms the value of pazopanib in patients with advanced alveolar soft part sarcoma (ASPS), with dimensional and durable responses, whereas trabectedin shows a limited activity. Alveolar soft part sarcoma is resistant to conventional cytotoxic chemotherapy. Pazopanib and trabectedin are licensed for treatment of sarcoma from second line; in the lack of prospective clinical trials, these results are relevant to defining ASPS best management and strongly support initiatives aimed at obtaining the approval of pazopanib in the front line of the disease.
    Type of Medium: Online Resource
    ISSN: 1083-7159 , 1549-490X
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2018
    detail.hit.zdb_id: 2023829-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2016
    In:  Clinical Sarcoma Research Vol. 6, No. 1 ( 2016-12)
    In: Clinical Sarcoma Research, Springer Science and Business Media LLC, Vol. 6, No. 1 ( 2016-12)
    Type of Medium: Online Resource
    ISSN: 2045-3329
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2623217-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5321-5321
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 5321-5321
    Abstract: S100A4 is a protein linked to increased invasion and metastatic potential of cancer cells and the significance of S100A4 as a prognostic marker has been reported for different tumor types. It has no enzymatic activity and exerts its functions through interaction with other proteins, both intracellularly and in the extracellular space. Many functions and biological effects of S100A4 have been elucidated, but the detailed knowledge on receptor(s) affected and signaling pathways induced are not completely revealed. Induction of metastasis resembles changes occurring during epithelial-to-mesenchymal transition (EMT) and it has been suggested that several of the metastases promoting properties of S100A4 is caused by involvement in this process. In a previous study we identified a set of 136 genes affected by extracellular S100A4. Induced expression of some of these genes was validated and found to be dependent on NF-κB signaling. In the osteosarcoma cell line used, S100A4 activated NF-κB through IKK and IκBα phosphorylation, and the induced signal transduction was found independent of the previously suggested S100-receptor RAGE (receptor for advanced glycation end products). In the present study, tyrosine kinase arrays (PamGene) were utilized to screen for substrates phosphorylated by S100A4-activated kinases. Comparing levels of substrate phosphorylation, S100A4 stimulation significantly affected 32 substrates, including EGFR and PDGFR. EGFR and PDGFR phosphorylation has been validated using western immunoblotting and immunoprecipitation. The EGFR inhibitors AG 1478 and gefitinib reduced the level of basal and S100A4-induced EGFR phosphorylation. Interestingly, reduced levels of S100A4-induced IκBα phosphorylation and NF-κB activation were also observed. The mechanisms of EGFR activation are under further investigation. Of particular interest, EGFR could be activated by ligands cleaved from the cell membrane in a process involving ADAM activity, referred to as ectodomain shedding. Thus, we are in the process of testing the effect of ADAM inhibitors in our cell system and characterize the expression of specific ADAMs. Insight into the signaling mechanisms induced by S100A4 is important to understand how this protein is involved in the biological processes constituting the metastatic cascade and will provide valuable information for identification of novel therapeutic targets. 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 5321. doi:1538-7445.AM2012-5321
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2004-2004
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2004-2004
    Abstract: S100A4 is a Ca2+-binding protein promoting metastasis in several types of tumors. The protein possesses a wide variety of biological functions, and locates to different subcellular compartments including the nucleus, cytoplasm and extracellular space. Even though S100A4 is mainly localized in the latter two compartments, we have previosusly shown that the protein is localized to the nucleus in more aggressive forms of colorectal cancer (CRC) and that nuclear S100A4 has prognostic impact. This may suggest a function of nuclear S100A4 in the regulation of tumor progression and metastasis formation. The CRC cell line SW620 shows heterogeneity in nuclear S100A4 expression and in preliminary experiments we sorted cells into three distinct phases of the cell cycle using Fluorescent-activated cell sorting (FACS). When comparing the phases, the G2/M cells showed increased accumulation of nuclear S100A4 compared to cells in the G1 and S phase, respectively. Further validation was done using treatment with the cell cycle inhibitors nocodazole or thymidine, followed by subsequent release of cells. Protein expression of S100A4 at selected time points after release did also show accumulation in the nucleus around the G2/M transition making this subcellular translocation interesting for further investigation. Cancer cells are known to have a dysfunctional cell division apparatus and these results made it tempting to speculate whether S100A4 may be involved in cell cycle regulation. To further study the functional importance of the nuclear expression two different approaches were followed. First, CRC cells exclusively expressing S100A4 in the nucleus were made and used to identify downstream targets of nuclear S100A4. At the moment potential targets are being validated. Furthermore, we searched for factors known to be associated with the G2/M phase. Of great interest, the Proximity Ligation Assay (PLA) demonstrated that S100A4 and Cyclin B1, an important regulator of the G2/M transition, are within the same protein complex, and we are currently in the process of further characterizing this interaction. With clinical data linking nuclear expression of S100A4 with aggressive CRC, elucidation of the biological processes involved might unravel novel therapeutic possibilities. Citation Format: Eivind V. Egeland, Kjetil Boye, Kjersti Flatmark, Solveig J. Pettersen, Tove Oyjord, Gunhild M. Maelandsmo. S100A4 in colorectal cancer - biological function of nuclear localization. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2004. doi:10.1158/1538-7445.AM2014-2004
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 24, No. 2_Supplement ( 2018-01-15), p. A08-A08
    Abstract: The CircSarc study aims to provide new insights into the clinical utility of liquid biopsies in sarcomas. Today, mutational profiles of solid tumors are obtained from tissue biopsies or surgical specimens. Recent advances in technology now allows to use blood plasma as a “liquid biopsy,” examining circulating tumor DNA (ctDNA) shed by the tumor cells into peripheral blood. ctDNA in plasma carries tumor-specific alterations that can be used to monitor minimal residual disease, response to therapy, tumor burden and evolution throughout the course of the disease. In CircSarc, we have enrolled 30 high-grade soft-tissue sarcoma patients, with localized disease, who are being followed throughout the course of their disease. Plasma from each patient is collected longitudinally: before and after surgery, at each routine control, and before and after each treatment cycle. Patients' tumor and germline DNA were exome sequenced to identify tumor-specific mutations, and ctDNA is being sequenced using a comprehensive 900 cancer gene panel. The level of ctDNA in plasma, represented by the tumor-specific biomarkers, is then monitored throughout the course of the treatment, and acts as an indicator of tumor burden. In addition, we are analyzing plasma samples from 70 gastrointestinal stroma tumor (GIST) samples using targeted resequencing with Anchored Multiplex PCR (ArcherDX). The levels of KIT and PDGFRA mutations in ctDNA are being correlated with clinical and pathologic features. The established targeted resequencing protocols for ctDNA provide different levels of complexity and sensitivity. In the first screening, we have successfully detected somatic mutations in plasma at time of surgery in 70% of the samples analyzed. Mutations present in the tumor at an allele frequency larger than 20% can be robustly identified in plasma. In addition, analysis of plasma samples identified novel mutations not detected in the primary tumor, possibly reflecting intratumor heterogeneity. Similarly, analysis of plasma from GIST patients has identified ctDNA in over 50% of the cases analyzed. In selected cases, ctDNA sequencing has revealed tumor heterogeneity, which has been confirmed by spatial biopsies of the resected tumor. Our work provides new insights into the clinical significance of ctDNA in sarcomas. By repeated sampling of liquid biopsies, somatic mutations identified in ctDNA can be used as unique noninvasive tumor-specific biomarkers for monitoring tumor burden and disease evolution throughout the course of the disease. Citation Format: Heidi M. Namløs, Seyed Hossein Moosavi, Susanne Lorenz, Bodil Bjerkehagen, Olga Zaikova, Synnøve Granlien, Stine Næss, Eva Stratford, Else Munthe, Lars B. Aasheim, Sigve Nakken, Eivind Hovig, Nina L. Jebsen, Kirsten Sundby Hall, Skyler Mishkin, Brian Kudlow, Ola Myklebost, Kjetil Boye, Leonardo A. Meza-Zepeda. Disease monitoring by liquid biopsies in sarcomas [abstract]. In: Proceedings of the AACR Conference on Advances in Sarcomas: From Basic Science to Clinical Translation; May 16-19, 2017; Philadelphia, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(2_Suppl):Abstract nr A08.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 11 ( 2018-11-01), p. 2473-2480
    Abstract: Molecular analysis of circulating tumor DNA (ctDNA) has a large potential for clinical application by capturing tumor-specific aberrations through noninvasive sampling. In gastrointestinal stromal tumor (GIST), analysis of KIT and PDGFRA mutations is important for therapeutic decisions, but the invasiveness of traditional biopsies limits the possibilities for repeated sampling. Using targeted next-generation sequencing, we have analyzed circulating cell-free DNA from 50 GIST patients. Tumor-specific mutations were detected in 16 of 44 plasma samples (36%) from treatment-naïve patients and in three of six (50%) patients treated with tyrosine kinase inhibitors. A significant association between detection of ctDNA and the modified National Institutes of Health risk classification was found. All patients with metastatic disease had detectable ctDNA, and tumor burden was the most important detection determinant. Median tumor size was 13.4 cm for patients with detectable mutation in plasma compared with 4.4 cm in patients without detectable mutation (P = 0.006). ctDNA analysis of a patient with disease progression on imatinib revealed that multiple resistance mutations were synchronously present, and detailed analysis of tumor tissue showed that these were spatially distributed in the primary tumor. Plasma samples taken throughout the course of treatment demonstrated that clonal evolution can be monitored over time. In conclusion, we have shown that detection of GIST-specific mutations in plasma is particularly feasible for patients with high tumor burden. In such cases, we have demonstrated that mutational analysis by use of liquid biopsies can capture the molecular heterogeneity of the whole tumor, and may guide treatment decisions during progression. Mol Cancer Ther; 17(11); 2473–80. ©2018 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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. 74, No. 19_Supplement ( 2014-10-01), p. 1093-1093
    Abstract: The cysteine proteinase legumain is unique in cleaving substrates N-terminally of asparagine, and recent observations also revealed carboxypeptidase activity. This specificity combined with its up-regulation in a number of solid tumors prompts to take advantage of legumain for cancer therapy and as a prognostic biomarker. Traditionally legumain was thought to reside within the endo-lysosomes, but the proteinase has also been reported expressed elsewhere in the cell, and we recently described nuclear localization of proteolytically active legumain in colorectal cancer cells. Presently, trafficking of legumain in the cell remains enigmatic, particularly when not routed to the endo-lysosomes. Our recent analysis of legumain expression in tissue from primary colorectal cancer (n=277) revealed that the subcellular localization pattern in tumor cells was diversified between the cases. Some had mostly dispersed and others more granulated cytoplasmic expression, while nuclear localization could be observed in about one third of the patients. We have thus established the association of legumain expression with clinicopathological parameters and outcome, and found that high levels of total or nuclear legumain in colon cancer cells were significantly negative for overall survival. Although an overall lower expression, nuclear localized legumain was also observed in normal tissue while in cytoplasm it was found polarized primarily to the apical side of the epithelial layer. We have previously observed legumain in the extracellular media of cancer cells and are in the process of analyzing wether it can also be found in exosomes. The unexpected observations in cellular localization are presumably due to altered trafficking. We have therefore analyzed legumain transport in colorectal cancer cells treated with tunicamycin to block N-glycosylation of de novo synthesized legumain, which had a striking effect on the trafficking and activation of the proteinase. Regarding functionality, we hypothesize that nuclear legumain may be acting as a transcription factor and have made fusion constructs of legumain and HaloTag® to identify potential genomic targets. The fusion protein has so far been verified to be highly expressed in the nuclei and will be used for chromatin capture. While the unique features of legumain make it an attractive target to exploit for therapy and diagnosis of cancer, our results reveal that location and function of legumain in normal and cancerous tissue is not completely elucidated. Such factors may be of outmost importance in attempts to utilize the protease activity for diagnostic and/or therapeutic purposes and may also represent novel cancer treatment opportunities. Citation Format: Mads H. Haugen, Kjersti Flatmark, Ingrid Damgaard, Tripti Tamhane, Birgit Engesæter, Siri Tveito, Eivind V. Egeland, Rigmor Solberg, Harald T. Johansen, Solveig J. Pettersen, Kjetil Boye, Klaudia Brix, Gunhild M. Mælandsmo. Legumain in colorectal cancer: Unorthodox localization and trafficking. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1093. doi:10.1158/1538-7445.AM2014-1093
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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
    Online Resource
    Online Resource
    MDPI AG ; 2020
    In:  Cancers Vol. 12, No. 12 ( 2020-12-11), p. 3719-
    In: Cancers, MDPI AG, Vol. 12, No. 12 ( 2020-12-11), p. 3719-
    Abstract: Natural killer (NK) cells are innate lymphoid cells with potent antitumor activity. One of the most NK cell cytotoxicity-sensitive tumor types is sarcoma, an aggressive mesenchyme-derived neoplasm. While a combination of radical surgery and radio- and chemotherapy can successfully control local disease, patients with advanced sarcomas remain refractory to current treatment regimens, calling for novel therapeutic strategies. There is accumulating evidence for NK cell-mediated immunosurveillance of sarcoma cells during all stages of the disease, highlighting the potential of using NK cells as a therapeutic tool. However, sarcomas display multiple immunoevasion mechanisms that can suppress NK cell function leading to an uncontrolled tumor outgrowth. Here, we review the current evidence for NK cells’ role in immune surveillance of sarcoma during disease initiation, promotion, progression, and metastasis, as well as the molecular mechanisms behind sarcoma-mediated NK cell suppression. Further, we apply this basic understanding of NK–sarcoma crosstalk in order to identify and summarize the most promising candidates for NK cell-based sarcoma immunotherapy.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 14, No. 1 ( 2014-12)
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2014
    detail.hit.zdb_id: 2041352-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: BMC Cancer, Springer Science and Business Media LLC, Vol. 17, No. 1 ( 2017-12)
    Type of Medium: Online Resource
    ISSN: 1471-2407
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2041352-X
    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