GLORIA

GEOMAR Library Ocean Research Information Access

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
  • 1
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2019-12-09)
    Kurzfassung: FMS-like tyrosine kinase 3 (FLT3) is a key driver of acute myeloid leukemia (AML). Several tyrosine kinase inhibitors (TKIs) targeting FLT3 have been evaluated clinically, but their effects are limited when used in monotherapy due to the emergence of drug-resistance. Thus, a better understanding of drug-resistance pathways could be a good strategy to explore and evaluate new combinational therapies for AML. Here, we used phosphoproteomics to identify differentially-phosphorylated proteins in patients with AML and TKI resistance. We then studied resistance mechanisms in vitro and evaluated the efficacy and safety of rational combinational therapy in vitro , ex vivo and in vivo in mice. Proteomic and immunohistochemical studies showed the sustained activation of ERK1/2 in bone marrow samples of patients with AML after developing resistance to FLT3 inhibitors, which was identified as a common resistance pathway. We examined the concomitant inhibition of MEK-ERK1/2 and FLT3 as a strategy to overcome drug-resistance, finding that the MEK inhibitor trametinib remained potent in TKI-resistant cells and exerted strong synergy when combined with the TKI midostaurin in cells with mutated and wild-type FLT3 . Importantly, this combination was not toxic to CD34+ cells from healthy donors, but produced survival improvements in vivo when compared with single therapy groups. Thus, our data point to trametinib plus midostaurin as a potentially beneficial therapy in patients with AML.
    Materialart: Online-Ressource
    ISSN: 2045-2322
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2019
    ZDB Id: 2615211-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1581-1581
    Kurzfassung: Acute Myeloid Leukemia (AML) is the most common type of acute leukemia in adults and the second in children. The overall survival is less than 35% and 60% for adults and children respectively. Activating mutations of FLT3 are now recognized as the most common molecular abnormality in this disease, and the poor prognosis of patients harboring these mutations renders FLT3 an obvious target of therapy. Although different tyrosine kinase inhibitors (TKI) have been used for this purpose, the ability of these drugs to extend progression-free and overall survival in this patient population is limited by drug resistance. This strategy could be improved by rationally combining TKIs with other agents. In this work, we have explored by phosphoproteomics the alternative pathways activated after TKI treatment in vitro and ex vivo. The phosphoproteome profile of the bone marrow from a FLT3-AML patient before and after TKI treatment, studied by LC-MSMS after IMAC enrichment, suggested the activation of Ras-Raf-MEK-ERK1/2 pathway as a possible mechanism for TKI resistance, which could be avoided by dual inhibition using the MEK inhibitor trametinib. Therefore, we characterized the effect of trametinib in combination with the TIK pazopanib and sorafenib by the in vitro cell viability assay using WST8in the FLT3-ITD AML cell line MOLM13. As it is presented in figure 1a, trametinib showed an IC50 value in the low-nanomolar range (5.4 nM) and this MEKI produced a strong synergy (0.5 〈 Ci) with the two TKI tested (figure1b). Moreover, when we analized the activation of the three main pathways downstream the FLT3 receptor by western blot (figure 1c), we observed that the combination of trametinib with both pazopanib and sorafenib showed a complete inhibition of phospho-ERK1/2 compared to the DMSO control (P≤0.0001). Interestingly, we also observed some differences between the two combinations: while trametinib in combination with sorafenib inhibited STAT5 phosphorilation (P≤0.05), the MEKI combination with pazopanib produced a significant decrease on phospho-AKT levels (P≤0.01). In conclusion, we provide preclinical evidence that combining a TKI, such as sorafenib or pazopanib, with a MEKI, such as trametinib, is a rational and efficacious treatment regimen for AML. As trametinib has previously shown good results when combined with pazopanib in clinical trials for other kind of tumors, we expect similar results in AML. On the other hand, trametinib+sorafenib could offer an optimal combination in those patients with elevated levels of phospho-STAT5, as it has been described for patients which present FLT3-ITD mutations. M.L. holds a postdoctoral Fellowship of the Spanish Ministry of Economy and Competitiveness (FPDI-2013-16409) y ML.M. holds a Fellowship of the Spanish Ministry of Education, Culture and Sport (REF-91442). Disclosures Martínez-López: Novartis: Honoraria, Speakers Bureau.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2016
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    Online-Ressource
    Online-Ressource
    American Society of Clinical Oncology (ASCO) ; 2016
    In:  Journal of Clinical Oncology Vol. 34, No. 15_suppl ( 2016-05-20), p. 1071-1071
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 34, No. 15_suppl ( 2016-05-20), p. 1071-1071
    Materialart: Online-Ressource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Clinical Oncology (ASCO)
    Publikationsdatum: 2016
    ZDB Id: 2005181-5
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1221-1221
    Kurzfassung: Background: We hypothesized that the biphasic relapse pattern of TNBC could be explained by a limited number of activation patterns of signaling nodes. In addition, we sought to determine whether the hyperactive signaling nodes, distinguishing the cases with favorable vs adverse outcome, could be potential targets. Methods: Training set of 34 frozen tumor samples divided in two sets, (A) 13 patients, relapsed in & lt;4 years; (B) 21 patients relapse-free & gt;12 years, (mimicking the percentage and relapse patterns of unselected TNBC, but paired for T, N, G and Ki67). TNBC cell lines: 7 indolent (no metastases in 60 weeks) and 3 aggressive, develop metastases and kill recipient mice in & lt;4 months. Shotgun phosphoproteomics and TiO2-IMAC phosphopeptide enrichment coupled with mass spectrometry runs in a Orbitrap Elite Mass Spectrometer was performed. Spectra were processed with MaxQuant software. Differentially expressed phosphopeptides were obtained by applying linear models R limma package. Differential kinase activation driving the profiles segregating cured vs. relapsing cases was done using linear sequence motif analysis. The hyperactivated kinases were validated in an independent set of 113 consecutive TNBC cases with 12+ years of follow-up spotted in TMAs by using an in-house algorithm for immunohistochemistry coupled with computer-aided quantitation using an Ariol scanning, we took the kinases in the upper quartile (high activity). Survival analysis was performed with KM curves and log rank test; and Cox proportionate hazards model was used for multivariate models. Results: 11405 phosphopeptides were identified and quantified in the training set. Supervised clustering of relapsed vs. cured cases showed that 161 and 541 peptides were significantly up-regulated in the A and B groups, respectively (FDR & lt;0.15). After kinase-to-kinase co-linearity was ruled-out , gathering the high activity (upper quartile) of six kinases (a combined variable, herein Var1) showed statistically significant association with relapse, being these: PRKCE, pERK, c-KIT, CDK6, pP70S6K and pPNKP. Cox proportional hazards model of any of the six probes high (var1) vs rest: 9.9 vs. not reached years (P & lt;0.001). Patients that had any of the 6 kinases high have 47% of chance to relapse (only 2 out of 42 relapsed patients have 0/6 active kinases) vs patients with Var1 negative, 7% of chance (29 patients out of 72 have 0/6 active kinases) we also observed constants patterns of activations in the different sets expressions of kinases. We considered the kinases at Var1 as a potentially targets and we developed a pharmacological in vitro assay, testing pairs inhibitors on 10 TNBC cell lines; 99.3% of the combinations were supra-additive. Conclusion: High throughput p-proteomics allows a parsimonious segregation of early TNBC cases, easily detecting the cases with long-term cure vs the remaining while identifying potential therapeutic solutions for the patients falling in the adverse prognostic subgroups. Citation Format: Sara Fernandez Gaitero, Ivana Zagorac, Jose Francisco Lopez-Acosta, Gonzalo Gomez-Lopez, David González Pisano, Javier Muñoz Peralta, Luis Manso, Soledad Alonso, Renske Penning, Maarten Altelaar, Albert JR Heck, Miguel Quintela-Fandino. Triple-negative breast cancer (TNBC) phosphoproteomics [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 1221. doi:10.1158/1538-7445.AM2017-1221
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2017
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Cell Reports, Elsevier BV, Vol. 15, No. 12 ( 2016-06), p. 2705-2718
    Materialart: Online-Ressource
    ISSN: 2211-1247
    Sprache: Englisch
    Verlag: Elsevier BV
    Publikationsdatum: 2016
    ZDB Id: 2649101-1
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. B9-B9
    Kurzfassung: BACKGROUND: Triple-negative breast cancer (TNBC) is a breast cancer subtype that is not defined by targetable molecular markers. Since all the aberrations that exist and ultimately contribute to the tumor phenotype converge, from a functional point of view, in the final status of the phosphorylation of the proteome in a given moment, we sought to interrogate the phosphoproteome with two aims: 1) establish a taxonomy of TNBC based on measurable markers that predict clinical course; 2) reduce the phosphoproteome that characterizes the bad- from good-prognosis cases to its targetable, driving kinases, in order to define rational therapeutic approaches in TNBC. We chose mass spectrometry as a discovery approach, because of its coverage, sensitivity, dynamic range and specificity. Here we report the part of phosphoproteomic analysis used for establishing a new taxonomy of TNBC. METHODS: We performed quantitative phosphoproteomics from a discovery set of 34 frozen tumor samples divided in two sets paired by classic prognosis factors: 1, with 13 patients relapsed in less than 3 years; 2, 21 patients, no relapse in 10 years follow up. Raw data were processed with Maxquant and cases clustered using a supervised hierarchical approach. Kinases (predicted with linear domain consensus analysis tools using a PHOSIDA) and phospho-sites discriminating the set 1 from 2 were validated by immunohistochemistry in 113 consecutive TNBC cases, spotted in TMAs, with 14 years follow up. RESULTS: Overall, 15000 unique phosphopeptides were identified. Supervised clustering of subset 1 vs. 2 showed that 161 and 541 peptides were significantly more phosphorylated in the subset 1 and 2, respectively (FDR & lt;0.15). Consensus domain analysis predicted kinases CDK1,2,4,6, CLK1, DAPK3/ZIPK, PAK2, RSK1, p70S6K, AKT, PKCEpsilon and PIM1 were driving the profile of subset 1. Those kinases, plus 9 phosphosites against which antibodies were available, were interrogated in the validation set. Kinase validation was performed directing an antibody against the active kinase form, if available; if not, against the total levels. The staining of kinases and the candidate phosphosites was analyzed with an Ariol, scored with a continue value from 0 to 3, and divided in quartiles for each staining probe. So far, 12 probes have been quantified. The prognosis of patients with upper quartile staining vs. the remainder was analyzed with the log-rank test and cox proportionate hazards model for each probe. Five of them showed statistically significant association with relapse (pSTAT3Tyr705, 6.1 vs. 10.1 years, p = 0.024; CYCB1, 7 vs. 10.3 years, p = 0.027; CDK6, 7 vs. 10.3 years, p = 0.013; pp70S6KThr389, 7.2 vs. 10 years, p = 0.042 and PRKCEpsilon, 7.1 vs. 10.2 years, p = 0.021). A combined variable integrating upper-quartile staining from either of those 5 probes occurring in 67.4% patients, identified almost all patients that experienced relapse: 48.3% of them relapsed (median time 7.6 years) vs. the reminder 32.6% of which only 13.8% relapsed (median time 11.9 years), log rank p = 0.001 INTERPRETATION: High-throughput mass-spectrometry is a powerful tool for generating a disease taxonomy, and can be translated to routine techniques like IHC. The signature constituted by pSTAT3Tyr705, CYCB1, CDK6, pp70S6K and PRKCEpsilon constitutes the most parsimonious signature to date able to detect all the early TNBC patients that relapse in the long term. Citation Format: Ivana Zagorac, Tamara Mondejar Tevar, Jesús Sánchez Ruíz, Albert Heck, Maarten Altelaar, Renske Penning, Harm Post, Gonzalo Gómez López, David Pisano, Javier Muñoz Peralta, Javier Muñoz Peralta, Manuel Morente, Luis Manso, Miguel Quintela-Fandiño. A phosphoproteomic portrait of triple - negative breast cancer: functional taxonomy. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B9.
    Materialart: Online-Ressource
    ISSN: 1535-7163 , 1538-8514
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2015
    ZDB Id: 2062135-8
    SSG: 12
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2018-08-29)
    Kurzfassung: Triple-negative breast cancer (TNBC) lacks prognostic and predictive markers. Here, we use high-throughput phosphoproteomics to build a functional TNBC taxonomy. A cluster of 159 phosphosites is upregulated in relapsed cases of a training set ( n  = 34 patients), with 11 hyperactive kinases accounting for this phosphoprofile. A mass-spectrometry-to-immunohistochemistry translation step, assessing 2 independent validation sets, reveals 6 kinases with preserved independent prognostic value. The kinases split the validation set into two patterns: one without hyperactive kinases being associated with a 〉 90% relapse-free rate, and the other one showing ≥1 hyperactive kinase and being associated with an up to 9.5-fold higher relapse risk. Each kinase pattern encompasses different mutational patterns, simplifying mutation-based taxonomy. Drug regimens designed based on these 6 kinases show promising antitumour activity in TNBC cell lines and patient-derived xenografts. In summary, the present study elucidates phosphosites and kinases implicated in TNBC and suggests a target-based clinical classification system for TNBC.
    Materialart: Online-Ressource
    ISSN: 2041-1723
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2018
    ZDB Id: 2553671-0
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...