GLORIA

GEOMAR Library Ocean Research Information Access

Sprache
Bevorzugter Suchindex
Ergebnisse pro Seite
Sortieren nach
Sortierung
Anzahl gespeicherter Suchen in der Suchhistorie
E-Mail-Adresse
Voreingestelltes Exportformat
Voreingestellte Zeichencodierung für Export
Anordnung der Filter
Maximale Anzahl angezeigter Filter
Autovervollständigung
Themen (Es wird nur nach Zeitschriften und Artikeln gesucht, die zu einem oder mehreren der ausgewählten Themen gehören)
Feed-Format
Anzahl der Ergebnisse pro Feed

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (10)
  • 2010-2014  (10)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (10)
Sprache
Erscheinungszeitraum
  • 2010-2014  (10)
Jahr
Fachgebiete(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 6 ( 2011-03-15), p. 2193-2202
    Kurzfassung: PrLZ/PC-1 is a newly identified, prostate-specific and androgen-inducible gene. Our previous study showed that PrLZ can enhance the proliferation and invasive capability of LNCaP cells, contributing to the development of prostate cancer. However, its potential role in androgen-independent processes remains elusive. In this study, we showed that PrLZ enhanced in vitro growth and colony formation of prostate cancer cells on androgen deprivation as well as tumorigenicity in castrated nude mice. In addition, PrLZ stabilized mitochondrial transmembrane potential, prevented release of cytochrome c from mitochondria to cytoplasm, and inhibited intrinsic apoptosis induced by androgen depletion. Mechanistically, PrLZ elevated the phosphorylation of Akt and Stat3 and upregulated Bcl-2 expression. Our data indicate that PrLZ protects prostate cancer cells from apoptosis and promotes tumor progression following androgen deprivation. In summary, we propose that PrLZ is a novel antiapoptotic gene that is specifically activated in prostate cancer cells escaping androgen deprivation may offer an appealing therapeutic target to prevent or treat advanced prostate malignancy. Cancer Res; 71(6); 2193–202. ©2011 AACR.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2011
    In:  Clinical Cancer Research Vol. 17, No. 8 ( 2011-04-15), p. 2159-2169
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 8 ( 2011-04-15), p. 2159-2169
    Kurzfassung: Purpose: Prostate tumor cells frequently show the features of osteoblasts, which are differentiated from bone marrow mesenchymal stem cells. We examined human prostate cancer cell lines and clinical prostate cancer specimens for additional bone marrow mesenchymal stem cell properties. Experimental Design: Prostate cancer cell lines were induced for osteoblastogenic and adipogenic differentiation, detected by standard staining methods and confirmed by lineage-specific marker expression. Abnormal expression of the markers was then assessed in clinical prostate cancer specimens. Results: After osteoblastogenic induction, cells of the LNCaP lineage, PC-3 lineage, and DU145 displayed osteoblastic features. Upon adipogenic induction, PC-3 lineage and DU145 cells differentiated into adipocyte-like cells. The adipocyte-like cancer cells expressed brown adipocyte-specific markers, suggesting differentiation along the brown adipocyte lineage. The adipogenic differentiation was accompanied by growth inhibition, and most of the adipocyte-like cancer cells were committed to apoptotic death. During cyclic treatments with adipogenic differentiation medium and then with control medium, the cancer cells could commit to repeated adipogenic differentiation and retrodifferentiation. In clinical prostate cancer specimens, the expression of uncoupling protein 1 (UCP1), a brown fat-specific marker, was enhanced with the level of expression correlated to disease progression from primary to bone metastatic cancers. Conclusions: This study thus revealed that prostate cancer cells harbor the stem cell properties of bone marrow mesenchymal stem cells. The abnormally expressed adipogenic UCP1 protein may serve as a unique marker, while adipogenic induction can be explored as a differentiation therapy for prostate cancer progression and bone metastasis. Clin Cancer Res; 17(8); 2159–69. ©2011 AACR.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 17 ( 2014-09-01), p. 4636-4646
    Kurzfassung: Purpose: miR-409-3p/-5p is a miRNA expressed by embryonic stem cells, and its role in cancer biology and metastasis is unknown. Our pilot studies demonstrated elevated miR-409-3p/-5p expression in human prostate cancer bone metastatic cell lines; therefore, we defined the biologic impact of manipulation of miR-409-3p/-5p on prostate cancer progression and correlated the levels of its expression with clinical human prostate cancer bone metastatic specimens. Experimental Design: miRNA profiling of a prostate cancer bone metastatic epithelial-to-mesenchymal transition (EMT) cell line model was performed. A Gleason score human tissue array was probed for validation of specific miRNAs. In addition, genetic manipulation of miR-409-3p/-5p was performed to determine its role in tumor growth, EMT, and bone metastasis in mouse models. Results: Elevated expression of miR-409-3p/-5p was observed in bone metastatic prostate cancer cell lines and human prostate cancer tissues with higher Gleason scores. Elevated miR-409-3p expression levels correlated with progression-free survival of patients with prostate cancer. Orthotopic delivery of miR-409-3p/-5p in the murine prostate gland induced tumors where the tumors expressed EMT and stemness markers. Intracardiac inoculation (to mimic systemic dissemination) of miR-409-5p inhibitor–treated bone metastatic ARCaPM prostate cancer cells in mice led to decreased bone metastasis and increased survival compared with control vehicle–treated cells. Conclusion: miR-409-3p/-5p plays an important role in prostate cancer biology by facilitating tumor growth, EMT, and bone metastasis. This finding bears particular translational importance as miR-409-3p/-5p appears to be an attractive biomarker and/or possibly a therapeutic target to treat bone metastatic prostate cancer. Clin Cancer Res; 20(17); 4636–46. ©2014 AACR.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2014
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 24 ( 2014-12-15), p. 6559-6569
    Kurzfassung: Purpose: MicroRNAs in the delta-like 1 homolog–deiodinase, iodothyronine 3 (DLK1-DIO3) cluster have been shown to be critical for embryonic development and epithelial to mesenchymal transition (EMT). DLK1-DIO3 cluster miRNAs are elevated in the serum of patients with metastatic cancer. However, the biologic functions of these miRNAs in the EMT and metastasis of cancer cells are poorly understood. We previously demonstrated the oncogenic and metastatic role of miR-409-3p/5p, a member of this cluster, in prostate cancer. In this study, we defined the role of miR-154* and miR-379, two key members of this cluster, in prostate cancer progression and bone metastasis in both cell line models and clinical specimens. Experimental Design: Genetic manipulation of miR-154* and miR-379 was performed to determine their role in tumor growth, EMT, and bone metastasis in mouse models. We determined the expression of miR-154* in prostate cancer clinical samples and bone metastasis samples using in situ hybridization and quantum dot labeling. Results: Elevated expression of miR-154* and miR-379 was observed in bone metastatic prostate cancer cell lines and tissues, and miR-379 expression correlated with progression-free survival of patients with prostate cancer. Intracardiac inoculation (to mimic systemic dissemination) of miR-154* inhibitor-treated bone metastatic ARCaPM prostate cancer cells in mice led to decreased bone metastasis and increased survival. Conclusion: miR-154* and miR-379 play important roles in prostate cancer biology by facilitating tumor growth, EMT, and bone metastasis. This finding has particular translational importance because miRNAs in the DLK1-DIO3 cluster can be attractive biomarkers and possible therapeutic targets to treat bone metastatic prostate cancer. Clin Cancer Res; 20(24); 6559–69. ©2014 AACR.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2014
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 3942-3942
    Kurzfassung: RANKL-elicited RANK activity plays critical roles in many biological and pathological conditions, including osteoclast differentiation/bone remodeling, lymph node/thymic development, central thermoregulation and progesterone-driven mammary gland maturation, differentiation and carcinogenesis. RANKL can be derived from osteoblasts, infiltrating inflammatory cells and stromal fibroblasts. We previously showed that malignant prostate cancer (PCa) cells expressed RANKL and that its expression was correlated with clinical PCa progression and bone metastasis. The present study examined whether PCa-derived RANKL acts on RANK+ PCa cells to promote PCa bone metastasis. We demonstrated several interesting key findings for RANKL-RANK signaling in PCa cells: 1) RANKL overexpression in human PCa LNCaP and ARCaPE cells results in increased RANKL-RANK signaling within PCa cells and confers PCa bone and soft tissue metastases in a tumor cell RANK-dependent manner since RANK knockdown in RANKL-expressing PCa cells fails to induce bone colonization or metastasis. 2) RANKL amplifies downstream signaling by activating RANKL and c-Met expression through a common transcription factor complex, c-Myc/Max, which was identified by site-directed mutagenesis and transcription factor deletion/interference assays. 3) Even a few RANKL+ PCa cells are sufficient to initiate the in vivo metastatic cascade by recruiting non-tumorigenic RANKL− PCa cells to participate in the metastatic process via downstream signaling amplification. This is supported by the observation that recombinant RANKL protein alone is sufficient to induce bone colonization and growth of RANKL− and non-metastatic PCa cells. 4) RANKL also promotes EMT and confers stem and neuroendocrine (NE) cell properties on participating cancer cells determined by changes in their specific markers. 5) In support of the roles of RANKL-RANK signaling in PCa bone metastasis, RANKL expression at the single cell level in primary PCa specimens predicts PCa patient survival. Collectively, these results demonstrated that autocrine/paracrine RANKL-RANK signaling in PCa cells establishes a premetastatic niche through a “vicious cycle," inducing RANKL and c-Met expression via activation of c-Myc/Max, and this promotes PCa EMT progression, stem and NE cell properties, and PCa bone and soft tissue metastases. RANKL expression status therefore offers new insights for dissecting the mechanism by which PCa cells exhibit propensity for bone colonization/metastasis. (Funding supported in part by R01 CA122602 and P01 CA098912 grants) Citation Format: Gina C.Y. Chu, Haiyen E. Zhau, Ruoxiang Wang, Andre Rogatko, Xu Feng, Majd Zayzafoon, Leland W.K. Chung. Autocrine/paracrine RANKL-RANK signaling promotes cancer bone metastasis and establishes premetastatic niche recruiting bystander cancer cells to participate in the metastatic process. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3942. doi:10.1158/1538-7445.AM2013-3942
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2013
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2010
    In:  Clinical Cancer Research Vol. 16, No. 10 ( 2010-05-15), p. 2833-2844
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 16, No. 10 ( 2010-05-15), p. 2833-2844
    Kurzfassung: Purpose: Near-IR fluorescence imaging has great potential for noninvasive in vivo imaging of tumors. In this study, we show the preferential uptake and retention of two hepatamethine cyanine dyes, IR-783 and MHI-148, in tumor cells and tissues. Experimental Design: IR-783 and MHI-148 were investigated for their ability to accumulate in human cancer cells, tumor xenografts, and spontaneous mouse tumors in transgenic animals. Time- and concentration-dependent dye uptake and retention in normal and cancer cells and tissues were compared, and subcellular localization of the dyes and mechanisms of the dye uptake and retention in tumor cells were evaluated using organelle-specific tracking dyes and bromosulfophthalein, a competitive inhibitor of organic anion transporting peptides. These dyes were used to detect human cancer metastases in a mouse model and differentiate cancer cells from normal cells in blood. Results: These near-IR hepatamethine cyanine dyes were retained in cancer cells but not normal cells, in tumor xenografts, and in spontaneous tumors in transgenic mice. They can be used to detect cancer metastasis and cancer cells in blood with a high degree of sensitivity. The dyes were found to concentrate in the mitochondria and lysosomes of cancer cells, probably through organic anion transporting peptides, because the dye uptake and retention in cancer cells can be blocked completely by bromosulfophthalein. These dyes, when injected to mice, did not cause systemic toxicity. Conclusions: These two heptamethine cyanine dyes are promising imaging agents for human cancers and can be further exploited to improve cancer detection, prognosis, and treatment. Clin Cancer Res; 16(10); 2833–44. ©2010 AACR.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2010
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 561-561
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 561-561
    Kurzfassung: Prostate stromal cells may play binary roles in the process of prostate cancer development. Being the first to be encountered by infiltrating prostate cancer cells, prostate stromal cells form the first defense line against prostate cancer progression and metastasis. On the other hand, interaction between prostate cancer and stromal cells may facilitate the formation of a tumor microenvironment favoring cancer cell growth and survival. To establish an experimental system for studying the interaction between cancer and stromal cells, we isolated three matched pairs of normal and cancer-associated human prostate stromal cells. The specimens were from three prostate cancer patients who underwent radical prostatectomy in the Department of Urology, Emory University School of Medicine. From each prostate, a cube of prostate tissue was dissected from a histologically normal region distal to the tumor, and another cube was dissected from a cancer-affected zone. Live cells in single cell preparation were cultured in low density for isolation of stromal clones. In contrast to the LNCaP prostate cancer cells, the isolated prostate stromal clones exhibited large fibroblastic morphology with slow growth rate. Growth and survival of the stromal clones were not affected by androgens and were highly resistant to serum starvation. In contrast to the normal counterparts, the cancer-associated stromal clones exhibited differentiated survival ability, as determined by colony formation assay following serum starvation. In direct co-culture, the prostate stromal cells inhibited growth of the LNCaP cells and made the production of Prostate Specific Antigen (PSA) less sensitive to androgen deprivation. Importantly in co-culture experiments, the stromal cells protected some LNCaP prostate cancer cells from death by serum starvation, and cancer-associated stromal clones showed more protection. The surviving LNCaP clones showed features of androgen-independent PSA expression similar to the lineage-related androgen-independent C4-2 cells, suggesting that cancer cells acquired increased malignancy through interaction with the stromal cells. The results from this study indicate that matched stromal cell pairs may serve as models for comparative analysis of molecular changes in the tumor microenvironment. These cells can also be used in co-culture with prostate cancer cells to simulate cancer-stromal interaction in the tumor microenvironment in order to define the role of prostate stromal cells in prostate cancer progression and metastasis. 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 561.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2010
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 3913-3913
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 3913-3913
    Kurzfassung: Near-infrared (NIR) fluorescence imaging has great potential for noninvasive tumor detection. In this study, we describe a novel hypoxia-mediated mechanism underlying a group of heptamethine cyanine dyes, MHI-148 and IR-783, which we developed previously that show tumor-specific targeting properties in cancer cells and living animals (Yang et al., Clin Cancer Res 2010, 16:2833-44 and J Urol 2013, in press). Hypoxia is a common condition found in a wide range of tumors, and mediates many aspects of cancer progression at both tissue and single-cell levels. Using a hypoxia-sensitive luciferase reporter construct (5HREp-ODD-luc) to assess dynamic and real-time tumor hypoxia, in vivo and ex vivo dual-modality imaging studies of human prostate cancer (PCa) growth in nude mice indicate superimposed distribution of NIR signals with hypoxia in tumors. Hypoxia and hypoxic mimickers augment NIR dye uptake in multiple human and murine cell lines representing different types of cancer. We determined that the increased dye uptake under hypoxic conditions was mediated by hypoxia-inducible factor 1α (HIF1α) both in vitro and in vivo. Using cDNA microarray analysis, we identified a group of organic anion-transporting polypeptide (OATP) genes (e.g. OATP1B3, OATP2B1 and OATP5A1), which were highly inducible by hypoxia/HIF1α in PCa cells. We further showed increased co-expression of HIF1α and a representative OATP1B3 gene upon PCa progression in clinical specimens. Pharmacological inhibition or genetic silencing of the OATP1B3 gene reduced the uptake of NIR dyes in tumor cells and tissues. Moreover, hypoxia/HIF1α up-regulates the transcriptional and translational expression of OATP1B3 in PCa, and HIF1α directly binds to a hypoxia response element in the OATP1B3 promoter determined by chromatin immunoprecipitation assays. In addition, we applied NIR dyes for clinical detection of renal cell carcinomas, which shows consistent results with clinical diagnostic tests, and importantly, the HIF1α/OATP1B3 signaling was manifested in these tumors. Together, we provide for the first time the molecular mechanisms of specific NIR dye uptake in cancer via tumor hypoxia and HIF1α/OATPs signals, and present clinical evidence for its potential future applications in cancer detection, prognosis, and therapy. Citation Format: Jason Boyang Wu, Chen Shao, Xiangyan Li, Peizhen Hu, Yi-Ting Chen, Xiaoliang Dou, Divya Sahu, Wei Li, Hiroshi Harada, Ruoxiang Wang, Haiyen E. Zhau, Leland W.K. Chung. Hypoxia-mediated cancer imaging by a novel class of near-infrared (NIR) heptamethine cyanine dyes. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3913. doi:10.1158/1538-7445.AM2013-3913
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2013
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2011
    In:  Molecular Cancer Therapeutics Vol. 10, No. 11_Supplement ( 2011-11-12), p. A233-A233
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 11_Supplement ( 2011-11-12), p. A233-A233
    Kurzfassung: Background: Prostate cancer bone metastases are associated with high levels of MET expression, and both HGF, the only known ligand for MET, and VEGF appear to direct crosstalk between tumor cells, osteoblasts (OBs), and osteoclasts (OCs). Cabozantinib (cabo), a dual MET-VEGFR2 inhibitor, has shown clinical activity in patients with metastatic castration-resistant prostate cancer (CRPC), where a complete or partial resolution of lesions on bone scans, reduced pain, and soft tissue tumor regression were observed in the majority of patients studied. Therefore, the preclinical effects of cabo were studied in the human CRPC bone xenograft model ARCaPM, which expresses both MET and the VEGF co-receptor neuropilin-1. In addition, the effects of cabo on the differentiation and activity of human OCs and mouse OBs were studied in vitro. Methods: ARCaPM cells were injected bilaterally into the tibiae of nude mice on Day 1, and on Day 31 animals received either cabo at 10 or 30 mg/kg (mpk) or vehicle once-daily for 7 weeks (wks). Animals (n=10 for each dose group) were sacrificed at the end of the treatment period and X-ray images of the tibiae were taken. Five representative tibiae per group were also scanned and analyzed by a Scanco 40 micro-CT instrument. In addition, one tibia from each mouse was fixed, decalcified and embedded for histology and histomorphometry analyzes. The OC culture system consisted of CD34+ cells derived from human bone marrow that were cultured on bovine bone slices in the presence of growth factors including M-CSF and RANK-L. The OB culture system used mouse KS483 cells that differentiate into OBs capable of forming mineralized bone nodules. Results: X-ray and micro-CT imaging of tibiae harvested after the 7 wk treatment period indicated that cabo treatment blocked both the osteoblastic and osteolytic progression of ARCaPM xenograft tumors in bone, with concomitant increases in bone mineral density. Histomorphometry data indicated that tumors were present in 70% (7/10) of the tibiae in the control group but only 30% (3/10) and 20% (2/10) in the 10 mpk and 30 mpk cabo groups, respectively. Consistent with its anti-tumor effect in the bone, cabo treatment decreased the ratio of tumor to tissue area and increased the bone area relative to the tissue area in the analyzed tibia sections compared to vehicle. Histological analyzes indicated an increase in the number of OBs and no change in the number of OCs along the trabecular bone perimeter of tibiae from cabo-treated animals relative to vehicle controls. In the in vitro studies, cabo treatment resulted in reduced OC differentiation in a dose dependent manner but did not affect the ability of mature OCs to resorb bone. In contrast, in the OB studies cabo treatment resulted in a biphasic effect inducing OB differentiation and bone forming activity at the lower doses while reducing these parameters at higher doses. Conclusions: Cabo treatment resulted in diverse effects on the differentiation and function of OBs and OCs in vitro, and blocked both osteoblastic and osteolytic progression of ARCaPM xenograft tumors in bone. These data demonstrate a substantial impact of cabo on tumor cells, OBs, and OCs consistent with the observed clinical results in patients with CRPC. Studies to characterize the molecular mechanisms underlying these effects are underway. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A233.
    Materialart: Online-Ressource
    ISSN: 1535-7163 , 1538-8514
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 2062135-8
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 7 ( 2011-04-01), p. 2600-2610
    Kurzfassung: Bone metastasis is one of the predominant causes of cancer lethality. This study demonstrates for the first time how β2-microglobulin (β2-M) supports lethal metastasis in vivo in human prostate, breast, lung, and renal cancer cells. β2-M mediates this process by activating epithelial to mesenchymal transition (EMT) to promote lethal bone and soft tissue metastases in host mice. β2-M interacts with its receptor, hemochromatosis (HFE) protein, to modulate iron responsive pathways in cancer cells. Inhibition of either β2-M or HFE results in reversion of EMT. These results demonstrate the role of β2-M in cancer metastasis and lethality. Thus, β2-M and its downstream signaling pathways are promising prognostic markers of cancer metastases and novel therapeutic targets for cancer therapy. Cancer Res; 71(7); 2600–10. ©2011 AACR.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...