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
  • Johnson, Michael D.  (4)
  • 2010-2014  (4)
Material
Person/Organisation
Language
Years
  • 2010-2014  (4)
Year
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2293-2293
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2293-2293
    Abstract: Endocrine therapy remains the cornerstone of treatment of estrogen-receptor positive breast cancer, with aromatase inhibitors (AIs) the current front-line therapy in post-menopausal women. Despite extending disease-free survival, 15-20% of patients receiving adjuvant AIs will relapse within 5 years. Long term estrogen deprivation (LTED) of breast cancer cells in culture has been successfully used to mimic AI-induced estrogen depletion to dissect mechanisms of AI resistance, implicating several pathways in estrogen-independent growth. However, patients treated with the AI anastrozole on the ATAC trial were reported to have average serum estrogen (E2) concentrations of 3pM. Importantly, in vitro growth assays with MCF-7 cells show that 3pM E2 is sufficient to induce growth. Further, 5α-androstane-3β,17β-diol (3βAdiol) is an androgen metabolite generated independent of aromatase, and thus is insensitive to AI therapy. We have demonstrated that 3βAdiol is an ERα agonist and can induce the growth of breast cancer cells. We sought to expand upon established LTED models to account for these observations. We hypothesize that weak or partial agonism of ERα during LTED by low concentrations of E2 or by 3βAdiol may result in the development of estrogen-independence different to that seen during complete estrogen deprivation. To model the effects of long term low concentrations of E2 or 3βAdiol, MCF-7 cells were grown in medium with charcoal stripped serum supplemented with defined hormone concentrations. Cells were selected in the EC10 and EC90 of E2 (1pM (LE) and 50pM (HE), respectively) or 3βAdiol (50pM (L3) and 1nM (H3), respectively), or estrogen deprived (ED – with 0.05% ethanol vehicle) for 7 months. Estrogen-independence was evaluated throughout selection by assessing cell growth in the absence or presence of E2 or 3βAdiol. ED cells developed estrogen-independence at 9-12 weeks, and are minimally responsive to E2 or 3βAdiol. H3, L3 and LE cells developed the ability to grow in estrogen-free conditions by 3-4 months, 4-5 months, and 6-7 months, respectively. L3 and LE cells are minimally responsive to E2/3βAdiol, while H3 cells remain fully responsive to E2/3βAdiol compared to parental MCF-7. HE cells continue to mimic parental cells after selection. Estrogen-free growth is blocked by anti-estrogen in LE and L3 cells; ED and H3 cells are anti-estrogen resistant. Western blot analysis demonstrated that LE and L3 cells maintain ERα activation (via ERα phosphorylation) in the absence of hormone, correlating with activated MAPK and Akt. cDNA expression analysis suggests distinct mechanisms of estrogen-independence in ED versus H3 cells. The presence of low concentrations of E2 or 3βAdiol selected unique mechanisms of estrogen-independence versus traditional LTED. These long term selections may represent novel and clinically relevant models of AI resistance. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2293. doi:10.1158/1538-7445.AM2011-2293
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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 ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5568-5568
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 5568-5568
    Abstract: The cytochrome P450 17A1 (CYP17A1) inhibitor abiraterone, which blocks synthesis of all steroid hormones including androgens and estrogens, is an effective treatment for castrate resistant prostate cancer. Its utility for treating hormone receptor-positive breast cancer has yet to be established. In prostate cancer, the anti-proliferative effects of abiraterone are mediated by inhibiting CYP17A1 activity resulting in a significant reduction in circulating concentrations of the sex steroids dehydroepiandrosterone (DHEA), androstenedione, testosterone, and dihydrotestosterone (DHT). Recently, it has been proposed that the reduction in aromatizable androgens by abiraterone may make this drug useful for the management of hormone-dependent breast cancer. However, data from our lab suggests that abiraterone has estrogenic properties and can induce proliferation in breast cancer cells expressing the estrogen receptor (ER). Crystal violet assays were used to assess abiraterone-induced proliferation in the ER-positive, estrogen-dependent breast cancer cell lines MCF-7 and T47D. We show that abiraterone induced a dose dependent increase in cell proliferation with an IC50 of 3.7 μM and maximal stimulation (200%) observed at 8μM, compared to vehicle treated cells. Abiraterone also induced the expression of the ER response gene, GREB1. Abiraterone-induced proliferation and gene expression was blocked by increasing doses of the selective estrogen receptor down-regulator (SERD) fulvestrant. Abiraterone induced the dose-dependent expression of luciferase in MCF-7 cells transfected with an estrogen responsive luciferase reporter construct. In conclusion, our data suggest that abiraterone can induce ER-positive breast cancer cell proliferation in vitro by acting as a weak ER agonist. Further studies are underway to determine whether estrogenic effects of abiraterone are also observed using in vivo models of ER response. If abiraterone exhibits estrogenic effects in vivo then these data would suggest that abiraterone should be combined with an ER antagonist if used for the clinical management of women with ER+ tumors. Citation Format: Cameron P. Capper, Michael D. Johnson, José M. Larios, James M. Rae. CYP17A1 and abiraterone: Implications for breast cancer endocrine therapy. [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 5568. doi:10.1158/1538-7445.AM2014-5568
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4593-4593
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 4593-4593
    Abstract: Previously we have shown that androgens induce the growth of estrogen-dependent, estrogen receptor (ER)-positive breast cancer cell lines via metabolism to estrogen-like steroids, independent of CYP19 aromatase. One pathway involves the 3β-hydroxysteroid dehydrogenase (3β-HSD) mediated metabolism of 5α-dihydrotestosterone (DHT) to 5α-androstane-3β,17β-diol (3βAdiol). 3βAdiol induces the growth of ER+ breast cancer cells by directly activating ER. However, in MCF-7 cells, maximal growth induction by 3βAdiol was only ∼75% of maximal growth induced by 17β-estradiol (E2), suggesting that 3βAdiol acts as a weak or partial agonist of ERα. In this study, we characterize 3βAdiol as a partial agonist of ERα using cell growth assays and an in vitro receptor binding assay. We use cDNA microarrays to examine gene expression induced by the partial agonist 3βAdiol vs. the full agonist E2. We tested if 3βAdiol as a partial agonist could antagonize E2-induced breast cancer cell growth. MCF-7 cells in estrogen-free conditions were treated with 100pM E2 (∼EC90) and increasing concentrations of 3βAdiol. Increasing 3βAdiol suppressed growth induced by E2 to the maximal level induced by 3βAdiol alone. These data confirm that 3βAdiol acts as a partial agonist of ERα. Additionally, we performed an in vitro, florescence polarization-based receptor binding assay to determine the relative affinities of 3βAdiol and E2 for ERα. Based on their ability to compete with a labeled ligand for binding to ERα, 3βAdiol bound ERα with an approximately 10-fold lower affinity than E2. This decreased affinity of 3βAdiol for ERα is consistent with the reduced potency of 3βAdiol versus E2 observed in growth assays. To compare changes in gene expression induced by either the full agonist E2 or partial agonist 3βAdiol, samples were analyzed with Affymetrix HG-U133 Plus 2.0 microarrays. MCF-7 cells were treated for 24 hours with 10nM of steroid or vehicle control. To evaluate overall changes in gene expression induced by the steroids, probe sets were selected that had & gt;2-fold change in expression versus the control array, with at least one sample having an expression value & gt;26. 1273 probe sets were differentially expressed between the E2 and control arrays, and 1318 between the 3βAdiol and control arrays, however, 1030 of these were common to both steroids. Genes in canonical estrogen signaling pathways were similarly regulated by both E2 and 3βAdiol, including GREB1, Bcl-2, TFF1 and CXCL12. Only 45 probe sets were differentially expressed between the E2 and 3βAdiol arrays. These data demonstrate that 3βAdiol is a partial agonist of ERα in our breast cancer cell model. The majority of changes in gene expression are similar between the full and partial agonist. These data suggest that the differential growth induction by the full and partial agonist are likely due to differential levels or time courses of gene induction, rather than the induction of a novel set of genes. 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 4593.
    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 ...
  • 4
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2012
    In:  Breast Cancer Research and Treatment Vol. 134, No. 3 ( 2012-8), p. 1027-1039
    In: Breast Cancer Research and Treatment, Springer Science and Business Media LLC, Vol. 134, No. 3 ( 2012-8), p. 1027-1039
    Type of Medium: Online Resource
    ISSN: 0167-6806 , 1573-7217
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2004077-5
    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...