Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: British Journal of Cancer, Springer Science and Business Media LLC, Vol. 115, No. 6 ( 2016-9), p. 674-681
    Type of Medium: Online Resource
    ISSN: 0007-0920 , 1532-1827
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2002452-6
    detail.hit.zdb_id: 80075-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Pathogens, MDPI AG, Vol. 10, No. 7 ( 2021-07-05), p. 845-
    Abstract: Nodding syndrome has been suggested to be triggered by neurotoxic leiomodin-1 auto-antibodies cross-reacting with Onchocerca volvulus. Here, we screened serum and CSF samples of persons with nodding syndrome and other forms of onchocerciasis-associated epilepsy (OAE) and African and European controls for leiomodin-1 antibodies by a cell-based assay (CBA) and Western blot (WB). These samples were also investigated for the presence of auto-antibodies cross-reacting with rat brain tissue by immunohistochemistry (IHC). Additionally, IHC was used to detect the leiomodin-1 protein in post-mortem brain samples of persons with OAE who died. Leiomodin-1 antibodies were detected by CBA in 6/52 (12%) and by WB in 23/54 (43%) persons with OAE compared to in 14/61 (23%) (p = 0.113) and 23/54 (43%) (p = 0.479) of controls without epilepsy. Multivariable exact logistic regression did not show an association between O. volvulus infection or epilepsy status and the presence of leiomodin-1. Leiomodin-1 antibodies were not detected in 12 CSF samples from persons with OAE or in 16 CSF samples from persons with acute-onset neurological conditions, as well as not being detected in serum from European controls. Moreover, the leiomodin-1 protein was only detected in capillary walls in post-mortem brain tissues and not in brain cells. IHC on rat brain slides with serum samples from persons with OAE or controls from persons with or without O. volvulus infection revealed no specific staining pattern. In conclusion, our data do not support OAE to be an autoimmune disorder caused by leiomodin-1 antibodies.
    Type of Medium: Online Resource
    ISSN: 2076-0817
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2695572-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: The Prostate, Wiley, Vol. 76, No. 10 ( 2016-07), p. 927-936
    Abstract: Resistance to docetaxel is common in metastatic castration‐resistant prostate cancer (mCRPC) and may be caused by sub‐therapeutic intratumoral drug concentrations. Cabazitaxel demonstrated survival benefit in docetaxel‐pretreated and docetaxel‐refractory patients. In this study, we investigated whether the superior antitumor activity of cabazitaxel in mCRPC is explained by higher intratumoral cabazitaxel levels. Since recent studies suggest a reduced efficacy of docetaxel following treatment with novel androgen receptor (AR)‐targeted agents, we also investigated taxane efficacy in an enzalutamide‐resistant tumor model. METHODS Intratumoral concentrations of docetaxel and cabazitaxel were correlated with antitumor activity in docetaxel‐naïve, docetaxel‐resistant, and enzalutamide‐resistant patient‐derived xenografts (PDXs) of prostate cancer. RESULTS Intratumoral drug levels were negatively related to intrinsic and acquired resistance to docetaxel. Also, the observed stronger antitumor activity of cabazitaxel was associated with increased cumulative exposure and higher intratumoral of cabazitaxel concentrations in all PDXs. CONCLUSIONS The superior antitumor activity of cabazitaxel in docetaxel‐ and enzalutamide‐resistant tumors can be partly attributed to higher intratumoral drug concentrations. Especially for patients who are intrinsically resistant to docetaxel resulting from suboptimal intratumoral docetaxel concentrations, cabazitaxel may be the preferred chemotherapeutic agent. Prostate 76:927–936, 2016 . © 2016 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 0270-4137 , 1097-0045
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 1494709-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: British Journal of Cancer, Springer Science and Business Media LLC, Vol. 123, No. 12 ( 2020-12-08), p. 1715-1719
    Abstract: Androgen receptor (AR) signalling drives neoplastic growth and therapy resistance in prostate cancer. Recent clinical data show that docetaxel combined with androgen deprivation therapy improves outcome in hormone-sensitive disease. We studied whether testosterone and AR signalling interferes with docetaxel treatment efficacy in castration-resistant prostate cancer (CRPC). We found that testosterone supplementation significantly impaired docetaxel tumour accumulation in a CRPC model, resulting in decreased tubulin stabilisation and antitumour activity. Furthermore, testosterone competed with docetaxel for uptake by the drug transporter OATP1B3. Irrespective of docetaxel-induced tubulin stabilisation, AR signalling by testosterone counteracted docetaxel efficacy. AR-pathway activation could also reverse long-term tumour regression by docetaxel treatment in vivo. These results indicate that to optimise docetaxel efficacy, androgen levels and AR signalling need to be suppressed. This study lends evidence for continued maximum suppression of AR signalling by combining targeted therapeutics with docetaxel in CRPC.
    Type of Medium: Online Resource
    ISSN: 0007-0920 , 1532-1827
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2002452-6
    detail.hit.zdb_id: 80075-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 157-157
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 157-157
    Abstract: Inactivation of the PTEN gene is a common event in PCa, leading to activation of the PI3K/AKT/mTOR pathway and promoting PCa progression. Inhibitors targeting this pathway are currently being developed as anticancer agents. While recent clinical trials with mTORC1 inhibitors have shown limited single-agent efficacy in castration-resistant prostate cancer (CRPC), it is still not known whether targeting alternative nodes of the PI3K/AKT/mTOR cascade may improve response. The current study aimed to characterize the response of PCa cell lines to multiple inhibitors targeting the PI3K, AKT and mTOR nodes of the cascade and identify possible mechanisms of resistance. The impact of the different PI3K/AKT/mTOR inhibitors on cell proliferation and apoptosis was measured by WST-1 and ApoTox-Glo assays, respectively, in a broad panel of established PCa cell lines and primary cell cultures generated from patient-derived xenografts. PTEN-negative PCa cell lines showed strong response to PI3Kβ/δ, AKT and mTOR inhibition, whereas PTEN-positive cells were resistant to PI3Kβ/δ and showed variable sensitivity to mTOR inhibitors. MTORC1 inhibitor showed low IC50 but reached plateau at about 70-80% growth inhibition. In contrast, mTORC1/2 inhibitor could reach complete growth inhibition at ~1μM concentration, in sensitive cell lines. Furthermore, treatment with PI3K or AKT inhibitor in combination with androgen depletion induced apoptosis in various PTEN-negative models, which is consistent with previously reported synergistic effect of PI3K/AKT and androgen receptor co-targeting. The effect of the inhibitors on phosphorylation of PI3K downstream targets (AKT, PRAS40, GSK3, S6K1 and 4EBP1) was analyzed by phospho-specific western blotting. Perturbation of different nodes of the PI3K/AKT/mTOR cascade resulted in differential phosphorylation of downstream targets, but there was no evident correlation with in vitro growth inhibition. In conclusion, the current study validates PI3Kβ and AKT as alternative targets in PTEN-negative PCa, whereas PTEN-positive models showed preferential yet variable responses towards mTOR inhibitors. Citation Format: Rute B. Marques, Ashraf Aghai, Wendy Stam, Wytske M. van Weerden. Targeting distinct nodes of the PI3K/AKT/mTOR cascade in prostate cancer cells: impact on cell proliferation, apoptosis and pathway signaling [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 157. doi:10.1158/1538-7445.AM2017-157
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Therapeutics Vol. 12, No. 11_Supplement ( 2013-11-01), p. B185-B185
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. B185-B185
    Abstract: Introduction and Objectives: A current challenge in prostate cancer (PCa) research is how to treat metastatic patients that have relapsed after androgen deprivation therapy. The PI3K/AKT pathway is frequently activated during PCa progression, with PTEN loss or mutation reported in up to 60% of advanced tumors, making it an interesting target for therapy. Recent reports have suggested a potential cross-talk between PI3K/AKT and androgen receptor (AR) pathway (1, 2). The present study aimed to assess the efficacy of combination therapy in PTEN-positive and PTEN-negative PCa models in vitro and in vivo and elucidate the mechanisms of interaction between both pathways. Material and Methods: The PI3K beta/delta inhibitor AZD8186 and AKT inhibitor AZD5363 were supplied by AstraZeneca (Macclesfield, UK). Growth response was tested in PTEN-positive (DU145 and VCAP) and PTEN-negative (LNCaP, PC346C, PC346Flu1 and PC346DCC) PCa cell lines by MTT assay. As response biomarkers, PI3K/AKT downstream targets phospho-AKT, phospho-PRAS40 and phospho-GSK3 were analyzed by western blotting. AR pathway activity was assessed by quantitative RT-PCR against AR, PSA, and TMPRSS2. Efficacy of both compounds was also tested in vivo in the PTEN-negative PC346C subcutaneous xenograft in combination with surgical castration. Results: AZD8186 and AZD5363 inhibited growth of all the PTEN-negative cell lines (LNCaP, PC346C, PC346Flu1, and PC346DCC) at nanomolar concentrations, while PTEN-positive cells were less sensitive (VCAP) or resistant (DU145). A dose dependent decrease in phosphorylation of AKT and its downstream targets was observed after treatment with AZD8186, which correlated with inhibition of cell growth. AZD5363 treatment resulted in increased AKT phosphorylation, as this compound stabilizes AKT in an inactive hyperphosphorylated state. Both AZD8186 and AZD5363 significantly inhibited in vivo growth of PC346C xenograft compared to that of placebo treated controls (respectively, 66% and 60% tumor growth inhibition after 28 days of treatment). Androgen depletion strongly increased the sensitivity to the PI3K/AKT inhibitors both in vitro and in vivo, with complete abrogation of tumor growth in castrated tumor-bearing mice. Additionally, up-regulation of AR signaling was observed after inhibition of the PI3K/AKT pathway, further lending support for cross-talk between both pathways. Conclusions: In conclusion, AZD8186 and AZD5363 efficiently targeted PI3K/AKT pathway and inhibited the growth of PTEN-negative PCa models in vitro and in vivo, particularly in combination with androgen depletion. These data suggest cross-talk between PI3K/AKT and AR pathways and indicate that for optimal antitumor efficacy inhibition of both pathways is required. The functional mechanisms underlying this cross-talk are the focus of our current research. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B185. Citation Format: Rute Marques, Ashraf Aghai, Debra Stuurman, Corrina de Ridder, Agnes Boer, Sander Hoeben, Jan Trapman, Wytske M. van Weerden. High efficacy of combination therapy of PI3K/AKT inhibitors with androgen deprivation in prostate cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B185.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: European Urology, Elsevier BV, Vol. 67, No. 6 ( 2015-06), p. 1177-1185
    Type of Medium: Online Resource
    ISSN: 0302-2838
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2015
    detail.hit.zdb_id: 1482253-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 255-255
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 255-255
    Abstract: Background Docetaxel is the first line of chemotherapy in metastatic castration resistant prostate cancer (mCRPC) patients. More than 50% of prostate cancer tumors express the solute carrier organic anion transporter family member 1B3 (SLCO1B3/OATP1B3), an uptake transporter that actively transports docetaxel into the cell. Interestingly, SLCO1B3 is also a transporter of endogenous substrates, such as testosterone. As the androgen receptor (AR) pathway is still active in mCRPC patients, we hypothesize that docetaxel can interact with the uptake of testosterone through SLCO1B3, which would result in decreased AR-pathway activation and subsequent tumor growth inhibition. Materials and Methods SLCO1B3 overexpressing cells (CHO-1B3) were used for uptake experiments with radioactively labeled hormones, such as estradiol-β-glucuronide (E2G). We stably transfected prostate cancers cells (PC346C-DCC and PC346C-DCC-G) with SLCO1B3 or with a green fluorescent protein (GFP) tag only. An androgen responsive element (ARE)-luciferase assay was created as a sensitive measure of AR pathway activation. ARE-activity and prostate specific antigen (PSA) production were analyzed with an ELISA assay after exposure to 1nM testosterone for 30 minutes with or without preincubation with docetaxel 10 μM in SLCO1B3 overexpressing and GFP- tagged control cells. Results SLCO1B3 overexpressing cells showed testosterone-mediated activation of the AR pathway: ARE-activity increased 6.4-fold and PSA production 7-fold in SLCO1B3-overexpressing cells as compared to control cells. Pre-incubation of CHO-1B3 cells with docetaxel resulted in reduced uptake of E2G to the level observed in control cells. Similarly, after exposure to testosterone and docetaxel ARE-activity was significantly decreased in SLCO1B3-overexpressing cells compared to that of controls. Conclusion: Docetaxel inhibits the activation of the AR pathway via SLCO1B3. This is a novel anti-androgenic effect exerted by docetaxel in addition to inhibition of the AR translocation that we demonstrated previously. Experiments are ongoing to investigate if testosterone uptake via SLCO1B3 is inhibited by docetaxel. Citation Format: Ellen S. de Morree, Ashraf Aghai, Alice A. Gibson, Ron H.J. Mathijssen, Eric A.C. Wiemer, Alex Sparreboom, Wytske M. van Weerden, Ronald de Wit. Docetaxel inhibits testosterone mediated activation of the AR pathway by SLCO1B3. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 255.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Molecular Cancer Therapeutics Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. C68-C68
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. C68-C68
    Abstract: Introduction: Castration resistant prostate cancer (CRPC) patients are treated with docetaxel as first-line chemotherapy. Unfortunately, response to docetaxel is highly variable among patients and intrinsic or acquired resistance is common. Understanding mechanisms of resistance may help to define biomarkers to select patients for the best therapy options and prevent treatments to which they may respond poorly. We established docetaxel-resistant xenografts from chemotherapy-naïve patient-derived xenografts (PDXs) in vivo. Next generation sequencing analysis of these chemotherapy-naïve and docetaxel-resistant PDXs revealed that SLCO1B3 expression was significantly downregulated in a subset of docetaxel-resistant tumors. As SLCO1B3 is also a known transporter of testosterone, the aim of the study was to further investigated its expression in relation with hormonal status of CRPC cells. Methods: Androgen responsive SLCO1B3 expressing PC346C cells were used to perform uptake assays with [14C]-labeled docetaxel and cabazitaxel after silencing of SLCO1B3 with siRNA. The androgen resistant PC346C-DCC-G clone, created by long-term culturing of PC346C cells in androgen deprived medium, did not express SLCO1B3. PC346C-DCC-G cells were stably transfected with SLCO1B3 or with a control construct containing green fluorescent protein (GFP). MTT cell proliferation assays were performed after 10 days of exposure to docetaxel or cabazitaxel in a dose range of 0-10nM. Prostate specific antigen (PSA) production was measured with an ELISA assay after exposure of 0.1nM testosterone for 30 minutes. Results: Silencing of SLCO1B3 in parental PC346C decreased uptake of docetaxel and cabazitaxel 2.0-fold (p = 0.01) and 2.1-fold (p = 0.0003), respectively. In line with this observation, SLCO1B3-transfected, overexpressing PC346C-DCC-G cells were more sensitive to docetaxel and cabazitaxel, resulting in reduced IC50-values of 1.9-fold for docetaxel and 3.5-fold for cabazitaxel compared to control transfected cells. Moreover, PSA production in SLCO1B3-overexpressing PC346C-DCC-G cells increased 2-fold as compared to control (p = 0.05) after testosterone exposure. Conclusion: Prostate cancer cells that overexpress SLCO1B3 are more sensitive to docetaxel and cabazitaxel treatment, which could be linked to increased uptake of both taxanes. Further studies are needed to clarify the role of SLCO1B3 in the uptake of cabazitaxel into the cell. Moreover, SLCO1B3 expression affects hormonal status of prostate cancer cells as reflected by PSA production. Research is ongoing to further elucidate the role of SLCO1B3 in prostate cancer and its impact on taxane efficacy and response. Citation Format: Ellen S. de Morree, Rene Bottcher, Robert J. van Soest, Ashraf Aghai, Corrina M. de Ridder, Alice A. Gibson, Ron HJ Mathijssen, Herman Burger, Erik AC Wiemer, Alex Sparreboom, Wytske M. van Weerden, Ronald de Wit. SLCO1B3 influences taxane-response in prostate cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C68.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: The Prostate, Wiley, Vol. 78, No. 13 ( 2018-09), p. 1013-1023
    Abstract: Prostate cancer is recognized as a heterogeneous disease demanding appropriate preclinical models that reflect tumor complexity. Previously, we established the PSA‐Cre ; PtenLoxP/LoxP genetic engineered mouse model (GEMM) for prostate cancer reflecting the various stages of tumor development. Prostate tumors in this Pten KO model slowly develop, requiring more than 10 months. In order to enhance its practical utility, we established a syngeneic panel of cell lines derived from PSA‐Cre targeted Pten KO tumors, designated the mouse prostate cancer (MuCap) model. Methods Four different MuCaP epithelial cell lines were established from three independent primary Pten KO mouse prostate tumors. Tumorigenic capacity of the MuCaP cell lines was determined by subcutaneous inoculation of these cell lines in immunocompetent mice. Response to PI3K‐targeted therapy was validated in ex vivo tissue slices of the established MuCaP tumors. Results The MuCaP cell lines were all tumorigenic in immunocompetent mice after subcutaneous inoculation. Interestingly, these syngrafted tumors represented different tumor growth rates and morphologies. Treatment with the specific PI3K inhibitor GDC0941 resulted in responses very similar between syngeneic MuCaP and primary Pten KO prostate tumors. Finally, immunoprofiling of the different syngeneic MuCaP tumors demonstrated differential numbers of tumor infiltrating lymphocytes and distinct immune gene profiles with expression of CD8, INFy, and PD1 being inversely related to tumor aggressiveness. Conclusions Collectively, we present here a well‐defined MuCaP platform of in vitro and in vivo mouse prostate cancer models that may support preclinical assessment of (immune)‐therapies for prostate cancer.
    Type of Medium: Online Resource
    ISSN: 0270-4137 , 1097-0045
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2018
    detail.hit.zdb_id: 1494709-2
    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