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
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. PD7-05-PD7-05
    Abstract: Background: Modulation of estrogen activity and/or synthesis is a mainstay therapeutic strategy for ER+ breast cancer (BC). However, despite the effectiveness of available endocrine therapies, many patients ultimately relapse or develop resistance to these agents via estrogen-dependent and estrogen-independent mechanisms, including mutations in ESR1 affecting the ER ligand-binding domain, that drive ER-dependent transcription and proliferation in the absence of estrogen. ER antagonists that robustly suppress ER signaling in both ESR1 wild-type and mutant tumors may be of substantial therapeutic benefit. GDC-9545 is a novel, potent, non-steroidal, orally bioavailable, selective ER antagonist/ER degrader (SERD) that induces tumor regression in ER+ BC patient-derived xenograft models. Methods: A phase I dose escalation study with 3+3 design was conducted in postmenopausal women with ER+ (HER2−) advanced or metastatic BC (mBC). Patients were eligible if they had advanced or mBC that had progressed while being treated with adjuvant endocrine therapy for ≥ 24 months and/or endocrine therapy in the advanced or mBC setting for ≥ 6 months. No more than 2 prior treatments for advanced or mBC. Primary objective was to determine the safety, pharmacokinetics (PK) and the recommended Phase 2 dose (RP2D) of GDC-9545. Pharmacodynamic (PD) activity was assessed with [18F] -fluoroestradiol (FES)-PET scans and, when feasible, by performing biomarker analysis of pre- and on-study tumor biopsies. Plasma samples were collected for PK analyses and imaging for response assessment (CT and bone scans) were repeated every 8 weeks. Results: From November 27 2017 to February 1 2019 patients (n = 29) with a median age of 58 years (range 42-75) and a median number of prior therapies for mBC of 2 (range 1-4) were enrolled at 4 total daily dose levels (10, 30, 90 and 250 mg) given once daily (QD) while fasting. Increases in GDC-9545 exposure were approximately dose proportional and t1/2 was ~30 hours. Treatment related adverse events (AEs) were all Grade 1 or 2. The most common treatment-related AEs in ≥10% of patients were fatigue (21%, n=6), nausea (21%, n=6), hot flushes (17%, n=5), diarrhea (17%, n=5), arthralgia (17%, n=5), constipation (10%, n=3) and dyspepsia (10%, n=3). No dose-limiting toxicities, treatment-related serious adverse events (SAEs), Grade ≥ 3 events, or fatal events have been reported. Treatment interruption was required for 3 patients: one patient for a short episode of dyspepsia, vomiting, diarrhea, and dizziness; and two patients due to unrelated SAEs. In 14 patients with FES-PET avid disease at baseline and post-baseline scans, 11 (78%) showed complete or near complete ( & gt; 90%) suppression of FES uptake to background levels, including patients with ESR1 mutations. Evidence of reduced ER, PR, and Ki67 IHC values and an ER activity signature was observed in paired pre- and on-treatment biopsies (n = 7). Twelve of 27 patients (10 mg [n=2], 30 mg [4] , 90 mg [4], 250 mg [2] ) had either confirmed partial responses (n = 3) or were on study ≥ 24 weeks (CBR = 44%), including patients pretreated with CDK4/6i, fulvestrant or harboring baseline ESR1 mutations. The R2PD was 100 mg QD and was selected for dose-expansion in post- and pre/peri-menopausal women with and without palbociclib, which is ongoing. Updated results will be presented. Conclusions: GDC-9545 is well-tolerated to date with PK exposure supporting QD dosing, evidence of robust PD target engagement, and encouraging anti-tumor activity in patients with advanced or ER+ mBC, including patients with ESR1 mutations. Dose expansion cohorts of GDC-9545 with or without palbociclib in post-menopausal and pre/peri-menopausal women with mBC are ongoing. Citation Format: Komal Jhaveri, Eric Paul Winer, Elgene Lim, Jose AlejandroPerez Fidalgo, Meritxell Bellet, Ingrid Alina Mayer, Valentina Boni, Jaymin M Patel, Aditya Bardia, Jose Manuel Garcia, Peter Kabos, Mary Gates, Ya-Chi Chen, Jill Fredrickson, Xiaojing Wang, Lori Sickels Friedman, Jill Spoerke, Steven Gendreau, Ciara Metcalfe, Lichuan Liu, Ching-Wei Chang, Sharareh Monemi, Monica Gonzalez, Ursula McCurry, Sandra Milan, Eric William Humke, Sherene Loi. A first-in-human phase I study to evaluate the oral selective estrogen receptor degrader (SERD), GDC-9545, in postmenopausal women with estrogen receptor-positive (ER+) HER2-negative (HER2-) metastatic breast cancer [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr PD7-05.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. P2-11-05-P2-11-05
    Abstract: Background: Estrogen (ER) and progesterone (PR) receptor staining with immunohistochemistry (IHC) forms the basis for endocrine therapy (ET) eligibility. However, ER protein level does not necessarily reflect ER functionality and is insufficient as a predictor of response or as a pharmacodynamic (PD) biomarker to assess the impact of ET on ER activity. Furthermore, assessing PD modulation based on a single ER target gene, PR, lacks robustness and cannot be utilized in PR- tumors. We established a transcriptional signature that more comprehensively reflects ER pathway activity. We showcase, in vitro, in vivo, and in patients enrolled in a Phase I trial of GDC-9545 -a novel, potent, orally bioavailable, selective estrogen receptor antagonist / degrader- that this signature may have utility as a predictive and PD biomarker. Methods: A signature of 21 estradiol (E2)-induced and 17 E2-repressed genes was experimentally derived by transcriptional profiling of 8 E2-stimulated ER+ breast cancer (BC) cell lines in vitro, and refined by leveraging TCGA RNA-seq data. The effect of ER inhibition by GDC-9545 on cellular proliferation was determined in an 8 day in vitro viability assay, and correlated with pre-treatment ER pathway activity as determined by the signature. The signature was evaluated in an in vivo efficacy study with GDC-9545 (0.1, 1 or 10 mg/kg) in PDX breast model HCI-013, measured using a Fluidigm® panel. Paired pre- and on-treatment tumor biopsies from 7 patients enrolled in the Phase I dose escalation study of GDC-9545 (10, 30 or 90 mg) in ER+/HER2- advanced or metastatic BC were collected to retrospectively assess ER pathway modulation. ER, PR and Ki67 protein levels were analyzed by IHC, while gene expression analysis from FFPE tissue was performed using Illumina’s RNA Access protocol and HiSeq. The ER pathway activity score was defined as the difference in average expression of the E2-induced versus E2-repressed genes, relative to ER pathway activity in a reference collection of 139 procured ER+/HER2- breast tumors. Results: A panel of 14 ER+/HER2- BC cell lines exhibited a spectrum of in vitro responses to GDC-9545 that strongly correlated with the pre-treatment ER pathway activity score. GDC-9545 had little anti-proliferative effect in cell lines with lowest scores, while having a considerably greater impact on cell lines exhibiting higher scores. Besides potential applicability as a predictive biomarker, we explored the utility of the ER activity score in vivo as a PD biomarker. We observed enhanced transcriptional pathway suppression with increased dose of GDC-9545, and a concomitant greater impact on Ki67 expression, in the in vivo HCI-013 PDX breast model. The ER signature was further evaluated in pre- and on-treatment tumor biopsy pairs from 7 patients enrolled in the GDC-9545 Phase I trial. ER activity scores of tumors from these patients were benchmarked against a cohort of 139 procured ER+/HER2- breast tumors, revealing a range in ER pathway activity prior to treatment with GDC-9545. Two tumors exhibiting profound Ki67 responses (post-treatment IHC & lt;1%, 10 or 30 mg) were amongst the 3 with highest pre-treatment ER activity scores, and additionally exhibited robust decreases in ER pathway activity following treatment with GDC-9545. An on-treatment decrease in ER pathway activity was observed at all dose levels in 6 patients total, covering both PR+ and PR- BC. The degree of pathway suppression was largely concordant with decreases in ER and PR (for PR+ tumors) protein levels, though ER IHC levels remained high in two patients with a low on-treatment ER activity score. Conclusions: The ER pathway activity signature introduced herein may have utility as a biomarker of drug-induced PD response and as a predictive biomarker to better identify patients who have the greatest potential to benefit from endocrine therapy, versus who may need combination therapy. Citation Format: Anneleen Daemen, Jill M Spoerke, Wei Zhou, Jane Guan, Ellen Ingalla, Amy Young, Marc Hafner, Junko Aimi, Ching-Wei Chang, Jennifer M Giltnane, Mary Gates, Ingrid A Mayer, Analia Azaro, Eric P Winer, Sherene Loi, Komal Jhaveri, Jennifer Lauchle, Steven Gendreau, Eric W Humke, Ciara Metcalfe. ER pathway activity signature as a biomarker for endocrine agent GDC-9545 [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P2-11-05.
    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
    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. 3977-3977
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3977-3977
    Abstract: Triple-negative breast cancer is a subtype of breast cancer with strong aggressiveness of tumor behavior and distinct patterns of metastasis. This unique phenotype of breast cancer is given by its clinically negative expression of estrogen receptor (ER), progesterone receptors (PR), and HER2 protein. Triple-negative breast cancer patients often suffer from ineffective hormone therapies, high recurrence rate, and a predilection for visceral metastasis. An accurate gene-expression signature specifically associated with triple-negative breast cancer will facilitate the identification of the existence of this disease and provide insights of its etiology. DNA microarray analysis was used to evaluate gene-expression profiles of 181 Asian breast cancer patients. There were 48 breast cancer patients who were triple-negative, and 133 of them were non-triple-negative. Several clinical information were matched in each group, including lymphovascular invasion, age, stage, grade, tumor size, tubule formation, nuclear pleomorphism, and mitotic count. After all 181 microarray data were normalized with quantile normalization, significantly expressed genes differentiated from the triple-negative group and non-triple-negative group were extracted using SAM (Significant Analysis of Microarray) and T-test. A panel of 50 genes was selected as the gene-expression signature for triple-negative breast cancer for Asian ethnicity, based on their unique patterns of gene-expression data using hierarchical clustering. Pathway analysis of the signature genes for triple-negative breast cancer was then performed in the lab. Genetic interactions of significantly expressed genes among different pathways found in this study will be shown and elaborated on the poster. 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 3977.
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 16 ( 2023-08-15), p. 2763-2774
    Abstract: Systemic targeted therapy in prostate cancer is primarily focused on ablating androgen signaling. Androgen deprivation therapy and second-generation androgen receptor (AR)–targeted therapy selectively favor the development of treatment-resistant subtypes of metastatic castration-resistant prostate cancer (mCRPC), defined by AR and neuroendocrine (NE) markers. Molecular drivers of double-negative (AR−/NE−) mCRPC are poorly defined. In this study, we comprehensively characterized treatment-emergent mCRPC by integrating matched RNA sequencing, whole-genome sequencing, and whole-genome bisulfite sequencing from 210 tumors. AR−/NE− tumors were clinically and molecularly distinct from other mCRPC subtypes, with the shortest survival, amplification of the chromatin remodeler CHD7, and PTEN loss. Methylation changes in CHD7 candidate enhancers were linked to elevated CHD7 expression in AR−/NE+ tumors. Genome-wide methylation analysis nominated Krüppel-like factor 5 (KLF5) as a driver of the AR−/NE− phenotype, and KLF5 activity was linked to RB1 loss. These observations reveal the aggressiveness of AR−/NE− mCRPC and could facilitate the identification of therapeutic targets in this highly aggressive disease. Significance: Comprehensive characterization of the five subtypes of metastatic castration-resistant prostate cancer identified transcription factors that drive each subtype and showed that the double-negative subtype has the worst prognosis.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. LB-304-LB-304
    Abstract: Hypoxia-inducible transcription factors (HIF) are key regulators of cellular adaptation to hypoxia in solid tumors, and HIF-1α controls the expression of genes involved in anaerobic metabolism, angiogenesis, cell growth and survival. Hydrocarbon cross-linked alpha helical peptides (Stapled Peptides) are a breakthrough approach to create new class of drugs that modulate intracellular protein-protein interactions. Here, we have mimicked the structure and function of the CITED2 protein, an endogenous negative regulator of the interaction between HIF-1α and p300 proteins, by designing a Stapled Peptide derived from CITED2 to generate the first example of a potent and selective Stapled Peptide inhibitor of HIF-1α-dependent transcription. ATSP-9172 bound to the CH1 domain of p300 and disrupted the HIF-1α C-TAD/CH1 interaction in biochemical assays and inhibited HIF-dependent reporter gene activity in ME-180 cells. Examination of endogenous transcript levels in ME-180 cells revealed that ATSP-9172 down-regulated the transcription of HIF-1α target genes, such as adolase C, angiopoietin-like 4, and carbonic anhydrase 9 in a dose-dependent manner, but did not affect the expression of non-HIF target genes, verifying a specific and on-target mechanism of action. ATSP-9172 exhibited a dramatic improvement in solubility and plasma stability profile relative to the linear peptide, and demonstrated favorable pharmacokinetic properties in mice by providing high systemic exposure, low plasma clearance and long elimination half-life. Finally, intravenous administration of ATSP-9172 on an every other day schedule significantly inhibited tumor growth in a PC-3 human prostate tumor xenograft model (p & lt; 0.05); this inhibition was found to be dose-dependent. Our results demonstrate that a Stapled Peptide mimicking the HIF inhibitory function of the native CITED2 protein provides a novel and specific strategy to suppress HIF-1α-dependent gene expression for cancer therapy. Citation Format: Kaiming Sun, Steven J. DeMarco, Vincent Guerlavais, Aditi Mukherjee, Sean Irwin, Eric Shi, Hongliang Cai, Krzysztof Darlak, Solimar Santiago, Jessica Pero, Karen A. Olson, Huw M. Nash, Yong Chang. ATSP-9172, a novel Stapled Peptide inhibitor of HIF-dependent transcriptional activity with in vivo antitumor efficacy in a preclinical model of prostate cancer. [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 LB-304. doi:10.1158/1538-7445.AM2013-LB-304
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 2490-2490
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 2490-2490
    Abstract: Background: Germline mutations in the NF1 gene are responsible for neurofibromatosis type 1, which is the world's most common genetic disorder. NF1 is also a key tumor suppressor gene that is frequently somatically mutated in a wide range of cancers. Approximately 80% of breast cancer is driven by the estrogen receptor α (ER), encoded by the ESR1 gene, and ER-positive (ER+) breast cancer can be treated by endocrine therapy targeting the ER transcriptional pathway. NF1 encodes neurofibromin, which is best known as a GTPase Activating Protein (GAP) for repressing Ras signaling. However, in a recent study we presented evidence supporting the model that NF1 has a GAP-independent activity by also acting as a transcriptional co-repressor for ER. NF1 loss enhanced ER transcription causing resistance to tamoxifen and aromatase inhibition. Approach and Results: In this study, we examined patient data from TCGA cohort and found that low NF1 mRNA levels associated with recurrence in luminal breast cancer, particularly in the luminal B subtype. Using purified components, we showed that full-length NF1 can directly bind ER. The ESR1-pE380Q mutation is a recurrent event in metastatic ER+ breast cancer. Our two-hybrid data showed that NF1 interacted less with the ER-E380Q than wild type ER, which agrees with structural analysis predicting co-repressors binding to be mediated by the ER-E380 residue. To assess how NF1-loss impacts ER-dependent gene expression in ER+ breast cancer cells, our ER ChIP-seq data showed that in the presence of estradiol, NF1-depletion promoted global ER recruitment to estrogen response elements (EREs) on chromatin. Expression of ERE-bound genes showed concordant expression changes by RNA-seq, confirming genome-wide transcriptional dysregulation of ER targeted genes by NF1 loss. ER+ NF1-depleted breast cancer cells responded initially to a selective ER degrader (SERD), such as fulvestrant and an oral SERD AZD9496, but acquired resistance with prolonged treatment. Resistance may be dependent on CDK4/6, a common growth pathway controlled by both Ras and ER. We showed that fulvestrant together with a CDK4/6 inhibitor Palbociclib can efficiently inhibit the growth of ER+ NF1-depleted breast cancer leading to tumor regression in a patient derived xenograft model. Conclusion: The loss of the full length NF1 can stimulate both ER and Ras signaling, and it is possible to efficiently treat ER+ NF1-depleted breast cancer by a SERD, in combination with CDK4/6 inhibitor. Citation Format: Zeyi Zheng, Jonathan T. Lei, Meenakshi Anurag, Long Feng, Purba Singh, Hilda Kennedy, Jin Cao, Xi Chen, Matthew J. Ellis, Eric C. Chang. Optimizing treatment strategy for NF1-depleted estrogen receptor positive breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2490.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 19 ( 2011-10-01), p. 6240-6249
    Abstract: Triple-negative breast cancers are an aggressive subtype of breast cancer with poor survival, but there remains little known about the etiologic factors that promote its initiation and development. Commonly inherited breast cancer risk factors identified through genome-wide association studies display heterogeneity of effect among breast cancer subtypes as defined by the status of estrogen and progesterone receptors. In the Triple Negative Breast Cancer Consortium (TNBCC), 22 common breast cancer susceptibility variants were investigated in 2,980 Caucasian women with triple-negative breast cancer and 4,978 healthy controls. We identified six single-nucleotide polymorphisms, including rs2046210 (ESR1), rs12662670 (ESR1), rs3803662 (TOX3), rs999737 (RAD51L1), rs8170 (19p13.1), and rs8100241 (19p13.1), significantly associated with the risk of triple-negative breast cancer. Together, our results provide convincing evidence of genetic susceptibility for triple-negative breast cancer. Cancer Res; 71(19); 6240–9. ©2011 AACR.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1810-1810
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1810-1810
    Abstract: Targeted therapeutics, such as those abrogating hypoxia inducible factor (HIF)/VEGF signaling, are initially effective against kidney cancer (or renal cell carcinoma, RCC); however, drug resistance frequently occurs via subsequent activation of alternative pathways. Through genome-scale integrated analysis of the HIF-α network, we identified the major protein kinase C substrate MARCKS (myristoylated alanine-rich C kinase substrate) as a potential target molecule for kidney cancer. In a screen of nephrectomy samples from 56 patients with RCC, we found that MARCKS expression and its phosphorylation are increased and positively correlate with tumor grade. Genetic and pharmacologic suppression of MARCKS in high grade RCC cell lines in vitro led to a decrease in cell proliferation and migration. We further demonstrated that higher MARCKS expression promotes growth and angiogenesis in vivo in an RCC xenograft tumor. MARCKS acted upstream of the AKT/mTOR pathway, activating HIF-target genes, notably VEGF-A. Following knockdown of MARCKS in RCC cells, the IC50 of the multi-kinase inhibitor regorafenib was reduced. Surprisingly, attenuation of MARCKS using the MPS peptide synergistically interacted with regorafenib treatment and decreased survival of kidney cancer cells through inactivation of AKT and mTOR. Our data suggest a major contribution of MARCKS to kidney cancer growth and provide an alternative therapeutic strategy of improving the efficacy of multi-kinase inhibitors. Citation Format: Ching-Hsien Chen, Wen-Hsin Chang, Eric Yu, Muhammad S. Arif, Reen Wu, Robert H. Weiss. Dysregulation of myristoylated alanine-rich C kinase substrate (MARCKS): A novel therapeutic target in renal cell carcinoma [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 1810. doi:10.1158/1538-7445.AM2017-1810
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1961-1961
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1961-1961
    Abstract: The epithelial-mesenchymal transition (EMT) is a developmental program that is aberrantly activated in cancer cells, producing an invasive phenotype that can lead to metastases. Inducers of EMT are largely known and have been widely studied. However, the mechanisms that regulate the link between extracellular stimuli and EMT phenotypes remain poorly understood. Preliminary evidence from our laboratory suggests that downregulation of proteasome activity may be responsible, in part, for driving EMT. Using immortalized human mammary epithelial (HMLE) cells as a model, we show that (1) EMT is associated with decreased proteasome activity and increased polyubiquitinated substrates, (2) pharmacologic inhibition of proteasome activity leads to increased EMT phenotypes and functionality, and (3) pharmacologic inhibition of proteasome activity leads to increased EMT via stabilization of the TGF-β signaling pathway. Together, these data suggest that proteasome activity may be an unappreciated regulator of EMT. Citation Format: Daniel A. Garcia, Asoka Banno, Eric D. Van Baarsel, Patrick J. Metz, Christella E. Widjaja, Stephanie H. Kim, Jack D. Bui, Jing Yang, John T. Chang. Downregulation of proteasome activity promotes epithelial-mesenchymal transition [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 1961. doi:10.1158/1538-7445.AM2017-1961
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4534-4534
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4534-4534
    Abstract: Treatment of lung cancer with anti-EGFR antibodies or tyrosine kinase inhibitors (TKIs) fails when KRAS2 cancer gene is activated by mutation. Mutation of the KRAS2 12th codon from the wild type GGU that encodes glycine (G) to a mutant GAU that encodes aspartate (D) occurs in some lung cancers. For this reason, we hypothesize that determining KRAS2 mutation status in vivo by external mRNA PET imaging will enable physicians to decide on alternatives to EGFR-directed therapy. To test the hypothesis, we designed wild type and mutant KRAS2 hybridization agents based on peptide nucleic acid (PNA), due to its strong, specific basepairing, and resistance to nucleases and proteases. At the C-terminus, we included a cyclized tetrapeptide based on insulin-like growth factor 1 (IGF1) to enable IGF1R-mediated cellular uptake. We included an N-terminal N2S2 chelator in order to bind 64Cu. We then administered the 64Cu-labeled agents by tail vein into mutant KRAS2 transgenic mice that develop spontaneous lung tumors. We demonstrated sequence-specific positron emission tomography (PET) imaging and quantitation of mutant KRAS2 G12D mRNA in the mice. The wild type control agent, differing by one base in the middle of the 12th codon, showed neglible images in the same mice. Wobble base agents with a hypoxanthine to detect either G12D or G12V KRAS2 mutants are under investigation. Supported by NIH CA148565; IP owned by EW/MLT, licensed to MTTI. Citation Format: Eric Wickstrom, Chang-Po Chen, Bishnuhari Paudyal, Matthew E. Wampole, Kaijun Zhang, Antican Wang, Bo Lu, Mathew L. Thakur. Mutant KRAS2 mRNA PET imaging in spontaneous transgenic lung cancers. [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 4534. doi:10.1158/1538-7445.AM2013-4534
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...