Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 15_suppl ( 2020-05-20), p. 1066-1066
    Abstract: 1066 Background: The cyclin-dependent kinase 4/6 inhibitors (CDK4/6i), with an anti-estrogen, are the standard of care for HR+/HER2- MBC. Insights from patient biopsies and preclinical analysis suggest that AKT1 activation can provoke CDK4/6i resistance. We hypothesized that targeting AKT1 following CDK4/6i progression may provide clinical benefit. Methods: TAKTIC is an open-label phase Ib trial exploring the combination of the AKT1 inhibitor, ipatasertib (ipat), with an aromatase inhibitor (Arm A), fulvestrant (Arm B), or the triplet combination (Arm C) of fulvestrant + ipat + palbociclib (palbo). The primary objective is to evaluate the safety and tolerability of ipat in combination with endocrine therapy +/- CDK4/6i. Key inclusion criteria include unresectable HR+/HER2- MBC; at least 1 prior therapy for MBC including any CDK4/6i; up to 2 prior lines of chemotherapy for MBC (no limit on prior endocrine therapy). Here, we present an interim analysis from the triplet combination (Arm C). Results: As of 1/31/2020, 25 pts have enrolled, including 12 on Arm C, all of whom received prior CDK4/6i (median no of prior lines = 5.5, range 2-7). Along with fulvestrant, 3 pts received ipat at 200mg + 125mg palbo, 7 pts received 300mg + 125mg palbo, and 2 pts received 400mg + 100mg palbo. To date, 8/12 pts remain on treatment including 2 with partial response, 3 with stable disease, 3 with restaging studies pending and 4 with progressive disease. The triplet combination was well tolerated. Grade 3 toxicities included reduced WBC (8/12), reduced neutrophil count (11/12), reduced lymphocyte count (2/12) and single instances of transaminitis, rash, and reduced platelet count. The only grade 4 toxicity was reduced neutrophil count (4/12). There were no DLTs observed and no discontinuations due to toxicity. Mean steady state pharmacokinetic parameters for ipat were similar to historical data from single agent trials suggesting that combined treatment with palbo + fulvestrant did not affect the pharmacokinetics of ipat. Updated analysis will be presented at the meeting. Conclusions: The triplet combination of endocrine therapy with CDK 4/6i and AKTi appears to be well tolerated in heavily pre-treated pts, with a subset demonstrating signs of clinical benefit. The trial demonstrates how insights into the molecular mechanisms of CDK4/6i resistance could be leveraged into actionable therapeutic regimens for HR+/HER2- MBC. Clinical trial information: NCT03959891 .
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Breast Cancer Research, Springer Science and Business Media LLC, Vol. 14, No. 4 ( 2012-8)
    Type of Medium: Online Resource
    ISSN: 1465-542X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2041618-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Genome Biology, Springer Science and Business Media LLC, Vol. 21, No. 1 ( 2020-12)
    Abstract: Quiescence (G0) is a transient, cell cycle-arrested state. By entering G0, cancer cells survive unfavorable conditions such as chemotherapy and cause relapse. While G0 cells have been studied at the transcriptome level, how post-transcriptional regulation contributes to their chemoresistance remains unknown. Results We induce chemoresistant and G0 leukemic cells by serum starvation or chemotherapy treatment. To study post-transcriptional regulation in G0 leukemic cells, we systematically analyzed their transcriptome, translatome, and proteome. We find that our resistant G0 cells recapitulate gene expression profiles of in vivo chemoresistant leukemic and G0 models. In G0 cells, canonical translation initiation is inhibited; yet we find that inflammatory genes are highly translated, indicating alternative post-transcriptional regulation. Importantly, AU-rich elements (AREs) are significantly enriched in the upregulated G0 translatome and transcriptome. Mechanistically, we find the stress-responsive p38 MAPK-MK2 signaling pathway stabilizes ARE mRNAs by phosphorylation and inactivation of mRNA decay factor, Tristetraprolin (TTP) in G0. This permits expression of ARE mRNAs that promote chemoresistance. Conversely, inhibition of TTP phosphorylation by p38 MAPK inhibitors and non-phosphorylatable TTP mutant decreases ARE-bearing TNFα and DUSP1 mRNAs and sensitizes leukemic cells to chemotherapy. Furthermore, co-inhibiting p38 MAPK and TNFα prior to or along with chemotherapy substantially reduces chemoresistance in primary leukemic cells ex vivo and in vivo. Conclusions These studies uncover post-transcriptional regulation underlying chemoresistance in leukemia. Our data reveal the p38 MAPK-MK2-TTP axis as a key regulator of expression of ARE-bearing mRNAs that promote chemoresistance. By disrupting this pathway, we develop an effective combination therapy against chemosurvival.
    Type of Medium: Online Resource
    ISSN: 1474-760X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2040529-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society for Clinical Investigation ; 2012
    In:  Journal of Clinical Investigation Vol. 122, No. 5 ( 2012-5-1), p. 1895-1906
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 122, No. 5 ( 2012-5-1), p. 1895-1906
    Type of Medium: Online Resource
    ISSN: 0021-9738
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2012
    detail.hit.zdb_id: 2018375-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: JCO Precision Oncology, American Society of Clinical Oncology (ASCO), , No. 7 ( 2023-05)
    Abstract: For patients with hormone receptor–positive (HR+), human epidermal growth factor receptor 2–negative (HER2–) metastatic breast cancer (MBC), first-line treatment is endocrine therapy (ET) plus cyclin-dependent kinase 4/6 inhibition (CDK4/6i). After disease progression, which often comes with ESR1 resistance mutations (ESR1-MUT), which therapies to use next and for which patients are open questions. An active area of exploration is treatment with further CDK4/6i, particularly abemaciclib, which has distinct pharmacokinetic and pharmacodynamic properties compared with the other approved CDK4/6 inhibitors, palbociclib and ribociclib. We investigated a gene panel to prognosticate abemaciclib susceptibility in patients with ESR1-MUT MBC after palbociclib progression. METHODS We examined a multicenter retrospective cohort of patients with ESR1-MUT MBC who received abemaciclib after disease progression on ET plus palbociclib. We generated a panel of CDK4/6i resistance genes and compared abemaciclib progression-free survival (PFS) in patients without versus with mutations in this panel (CDKi-R[–] v CDKi-R[+] ). We studied how ESR1-MUT and CDKi-R mutations affect abemaciclib sensitivity of immortalized breast cancer cells and patient-derived circulating tumor cell lines in culture. RESULTS In ESR1-MUT MBC with disease progression on ET plus palbociclib, the median PFS was 7.0 months for CDKi-R(–) (n = 17) versus 3.5 months for CDKi-R(+) (n = 11), with a hazard ratio of 2.8 ( P = .03). In vitro, CDKi-R alterations but not ESR1-MUT induced abemaciclib resistance in immortalized breast cancer cells and were associated with resistance in circulating tumor cells. CONCLUSION For ESR1-MUT MBC with resistance to ET and palbociclib, PFS on abemaciclib is longer for patients with CDKi-R(–) than CDKi-R(+). Although a small and retrospective data set, this is the first demonstration of a genomic panel associated with abemaciclib sensitivity in the postpalbociclib setting. Future directions include testing and improving this panel in additional data sets, to guide therapy selection for patients with HR+/HER2– MBC.
    Type of Medium: Online Resource
    ISSN: 2473-4284
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    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:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 782-782
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 782-782
    Abstract: The p53 tumor suppressor is a critical regulator of genomic stability. P53 is known as “the guardian of the genome” because it plays a crucial role in inducing apoptosis and cell cycle arrest in response to DNA damage. P53 is mutated in approximately 50% of all human tumors and is downregulated in a large majority of the remainder. An important regulator of p53 is the E3 ubiquitin ligase, MDM2, which acts to ubiquitinate p53 and target it for degradation through the proteasome. There is currently a large effort in the field directed toward developing targeted therapies that inhibit MDM2′s ability to interact with p53, thus allowing p53 to be stabilized. One such drug that is already undergoing clinical trials is Nutlin-3a. Our preliminary data demonstrate that the homeobox containing transcription factor, Six1, decreases the p53 response to DNA damage in a proteasome independent manner, suggesting that it may downregulate the p53 response even in the presence of Nutlins. Six1, is an important developmental regulator as it increases cell migration, invasion, and proliferation, however, it is not expressed in most normal adult tissues. It is, however, re-expressed in cancerous cells, including human breast lesions. As we are currently developing drugs to target Six1, it is essential to understand mechanistically how Six1 downregulates p53 and whether or not inhibition of Six1 may re-sensitize cells to Nutlin therapies. Here we show that Six1 downregulates p53, not on the mRNA level, but on the protein level, and that this downregulation leads to a decrease in p53 function. We have shown that Six1 can target p53 in a variety of different cell culture systems, including both breast and colon cancer cell lines, as well as normal breast and embryonic kidney cell lines. We have begun to examine the mechanism by which Six1 downregulates p53, and have shown that it is able to target p53 independent of MDM2, as siRNA knock down of MDM2 is not able to rescue p53 levels in the presence of Six1. Furthermore, Six1 does not affect the half life of p53, and congruently Six1 is able to downregulte p53 even in the presence of a proteosome inhibitor. Together, these results suggest that MDM2’s ability to target p53 for proteasomal degradation is dispensable for the ability of Six1 to decrease p53 levels. Furthermore, our preliminary data suggests that Six1 regulates p53 in a post-transcriptional mechanism, potentially through the use of the p53 3’UTR region. Six1 overexpressing lines are more resistant to Nutlin therapies in-vitro, demonstrating a significant increase in viability in the presence of Nutlins when compared to control cell lines. Thus, high levels of Six1 may correlate with decreased response to Nutlins clinically, and targeting of Six1 may be a novel approach to re-sensitize tumors expressing high levels of Six1 to Nutlin therapy. Citation Format: Christina N. Garlington, Douglas S. Micalizzi, Heide L. Ford. The Six1 homeoprotein downregulates p53, via a novel, MDM2 independent mechanism. [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 782. doi:10.1158/1538-7445.AM2013-782
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Wiley ; 2004
    In:  ChemInform Vol. 35, No. 8 ( 2004-02-24)
    In: ChemInform, Wiley, Vol. 35, No. 8 ( 2004-02-24)
    Type of Medium: Online Resource
    ISSN: 0931-7597 , 1522-2667
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2004
    detail.hit.zdb_id: 2110203-X
    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) ; 2008
    In:  Cancer Research Vol. 68, No. 7 ( 2008-04-01), p. 2204-2213
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 7 ( 2008-04-01), p. 2204-2213
    Abstract: Homeoproteins are transcription factors that act as master regulators of development and are frequently dysregulated in cancers. During embryogenesis, the Six1 homeoprotein is essential for the expansion of precursor cell populations that give rise to muscle and kidney, among other organs. Six1 overexpression is observed in numerous cancers, resulting in increased proliferation, survival, and metastasis. Here, we investigate whether Six1 can play a causal role in mammary tumor initiation. We show that Six1 overexpression in MCF12A mammary epithelial cells promotes multiple properties associated with malignant transformation, including increased proliferation, genomic instability, and anchorage-independent growth. We further show that this transformation is dependent on up-regulation of its transcriptional target, cyclin A1, which is normally expressed in the embryonic mammary gland but dramatically reduced in the adult gland. Six1-transformed MCF12A cells are tumorigenic in nude mice, forming aggressive tumors that are locally invasive and exhibit peritumoral lymphovascular invasion. In human breast carcinomas, expression of Six1 and cyclin A1 mRNA correlate strongly with each other (P & lt; 0.0001), and expression of Six1 and cyclin A1 each correlate with Ki67, a marker of proliferation (P & lt; 0.0001 and P = 0.014, respectively). Together, our data indicate that Six1 overexpression is sufficient for malignant transformation of immortalized, nontumorigenic mammary epithelial cells, and suggest that the mechanism of this transformation involves inappropriate reexpression of cyclin A1 in the adult mammary gland. [Cancer Res 2008;68(7):2204–13]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    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) ; 2012
    In:  Cancer Research Vol. 72, No. 2_Supplement ( 2012-01-08), p. A20-A20
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 2_Supplement ( 2012-01-08), p. A20-A20
    Abstract: Recent studies have highlighted the developmental transcription factor Six1 as an important mediator of breast cancer progression and metastasis. Six1 is overexpressed in a striking 90% of metastatic breast cancer lesions, and patients whose tumors overexpress Six1 have a decreased time to metastasis and relapse, as well as an overall decrease in survival. Six1 mediates metastasis via multiple mechanisms, including its ability to induce an epithelial-to-mesenchymal transition (EMT) and tumor initiating cell (TIC) characteristics, both of which are dependent on an upregulation of TGF-β signaling. Indeed, the Six1-induced increase in TGF-β signaling is critical for the ability of Six1 to induce late stage metastasis. Interestingly, Six1 not only activates TGF-β signaling, but it also enables the switch of TGF-β signaling from tumor suppressive to tumor promotional, a phenomenon of considerable importance in cancer pathogenesis. This event, coined the “TGF-β paradox,” has been an area of extensive research, but remains largely elusive. To further investigate the mechanism by which Six1 mediates the switch in TGF-β signaling, we performed a miRNA microarray screen and identified a cluster of miRNAs, the miR-106b-25 cluster, that is upregulated in response to Six1 overexpression. The miR-106b-25 cluster consists of three miRNAs, miR-106b, miR-93, and miR-25, which reside together in the intron of the MCM7 gene. Importantly, overexpression and knockdown experiments demonstrate that Six1 regulates all three miRNAs within the cluster. Interestingly, these miRNA have previously been implicated in the impairment of TGF-β-mediated growth suppression through repression of the cell cycle inhibitor, p21, and pro-apoptotic factor, Bim. These data suggest that Six1-induced upregulation of these miRNA may mediate the switch in TGF-β signaling from tumor suppressive to tumor promotional. Surprinsingly, bioinformatic analysis revealed that the miR-106b-25 cluster may also contribute to the activation of TGFβ signaling through repression of the TGF-β signaling inhibitor, Smad7, which mediates the degradation of TβRI. Indeed, overexpression of the miR-106b-25 cluster results in repression of the Smad7 protein, with concominant upregulation of TβRI. Furthermore, activation of TGF-β signaling is observed with miR-106b-25 overexpression, as demonstrated by an increase in phosphorylation of the downstream effector of the TGF-β pathway, Smad3, and by an upregulation of downstream TGF-β transcriptional targets. miRNA inhibition also demonstrates that miR-106b and miR-93 are necessary for Six1 induced TGF-β activation. Additionally, like Six1, the miR-106b-25 cluster is sufficient to induce features of EMT, including a redistribution of E-cadherin, and increased β-catenin transcriptional activation. Furthermore, the miR-106b-25 cluster is also sufficient to increase the TIC population as measured by flow cytometry, mammosphere formation, and in vivo serial dilution assays. Finally, we demonstrate a significant correlation between miR-106b, Six1, and activated TGF-β signaling in human breast cancers, and further show that high levels of miR-106b and miR-93 in breast tumors significantly predicts shortened time to relapse. These findings expand the spectrum of oncogenic functions of miR-106b-25, and may provide a novel molecular explanation, through the Six1 regulated miR-106b-25 cluster, by which TGF-β signaling shifts from tumor suppressive to tumor promotional. Citation Format: Anna L. Smith, Ritsuko Iwanaga, David J. Drasin, Douglas S. Micalizzi, Rebecca L. Vartuli, Heide L. Ford. The Six1-regulated miR-106b-25 cluster is a mediator of the tumor promotional effects of TGF-β signaling in human breast cancer [abstract]. In: Proceedings of the AACR Special Conference on Noncoding RNAs and Cancer; 2012 Jan 8-11; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Res 2012;72(2 Suppl):Abstract nr A20.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 18 ( 2020-09-15), p. 4852-4862
    Abstract: Plasma genotyping may identify mutations in potentially “actionable” cancer genes, such as BRCA1/2, but their clinical significance is not well-defined. We evaluated the characteristics of somatically acquired BRCA1/2 mutations in patients with metastatic breast cancer (MBC). Experimental Design: Patients with MBC undergoing routine cell-free DNA (cfDNA) next-generation sequencing (73-gene panel) before starting a new therapy were included. Somatic BRCA1/2 mutations were classified as known germline pathogenic mutations or novel variants, and linked to clinicopathologic characteristics. The effect of the PARP inhibitor, olaparib, was assessed in vitro, using cultured circulating tumor cells (CTCs) from a patient with a somatically acquired BRCA1 mutation and a second patient with an acquired BRCA2 mutation. Results: Among 215 patients with MBC, 29 (13.5%) had somatic cfDNA BRCA1/2 mutations [nine (4%) known germline pathogenic and rest (9%) novel variants]. Known g ermline pathogenic BRCA1/2 mutations were common in younger patients (P = 0.008), those with triple-negative disease (P = 0.022), and they were more likely to be protein-truncating alterations and be associated with TP53 mutations. Functional analysis of a CTC culture harboring a somatic BRCA1 mutation demonstrated high sensitivity to PARP inhibition, while another CTC culture harboring a somatic BRCA2 mutation showed no differential sensitivity. Across the entire cohort, APOBEC mutational signatures (COSMIC Signatures 2 and 13) and the “BRCA” mutational signature (COSMIC Signature 3) were present in BRCA1/2-mutant and wild-type cases, demonstrating the high mutational burden associated with advanced MBC. Conclusions: Somatic BRCA1/2 mutations are readily detectable in MBC by cfDNA analysis, and may be present as both known germline pathogenic and novel variants.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages