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: Nature Communications, Springer Science and Business Media LLC, Vol. 3, No. 1 ( 2012-03-06)
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2012
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature, Springer Science and Business Media LLC, Vol. 520, No. 7547 ( 2015-4), p. 368-372
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 15 ( 2011-08-01), p. 5067-5074
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 15 ( 2011-08-01), p. 5067-5074
    Abstract: V600EB-RAF mutation is found in 50% to 60% of melanomas, and the novel agents PLX4032/vemurafenib and GSK2118436 that inhibit the V600EB-RAF kinase achieve a remarkable clinical response rate. However, as might be expected, acquired clinical resistance to these agents arises in most melanoma patients. PLX4032/vemurafenib resistance that arises in vivo in tumor matched short-term cultures or in vitro in melanoma cell lines is not caused by acquisition of secondary mutations in V600EB-RAF but rather is caused by upregulating platelet-derived growth factor receptor β (PDGFRβ) or N-RAS which results in resistance or sensitivity to mitogen-activated protein (MAP)/extracellular signal-regulated (ERK; MEK) kinase inhibitors, respectively. In this study, we define a targeted combinatorial strategy to overcome PLX4032/vemurafenib resistance in melanoma cell lines or short-term culture where the resistance is driven by PDGFRβ upregulation, achieving synergistic growth inhibition and cytotoxicity. PDGFRβ-upregulated, PLX4032-resistant (PPRM) cell lines show dual phospho (p)-ERK and p-AKT upregulation, and their growth inhibitory responses to specific small molecule inhibitors correlated with p-ERK, p-AKT, and p-p70S6K levels. Coordinate inhibition of V600EB-RAF inhibition and the RTK–PI3K–AKT–mTORC axis led to functionally significant rebound signaling, illustrating a robust and dynamic network connectivity. Combined B-RAF, phosphoinositide 3-kinase (PI3K), and mTORC1/2 inhibition suppressed both immediate early and delayed compensatory signaling, resulting in a highly synergistic growth inhibitory response but less efficient cytotoxic response. In contrast, the combination of MEK1/2, PI3K, and mTORC1/2 inhibitors consistently triggered apoptosis in a highly efficient manner. Together, our findings offer a rational strategy to guide clinical testing in preidentified subsets of patients who relapse during treatment with V600EB-RAF inhibitors. Cancer Res; 71(15); 5067–74. ©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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 634-634
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 634-634
    Abstract: V600EB-RAF mutation is found in 50-60% of melanomas, and its in-human “druggability” has recently been demonstrated using the novel agent PLX4032, which can achieve an unprecedented ∼80% on-target, anti-melanoma response rate. Acquired resistance to PLX4032, however, has been observed in the majority of patients. We have shown previously that melanoma tumors in patients and cell lines in vitro acquire resistance to PLX4032 not by developing secondary mutations in V600EB-RAF but instead by upregulating PDGFRβ (showing MEK inhibitor insensitivity) or N-RAS (showing MEK inhibitor sensitivity). Here we used distinct subsets of melanoma cell lines (resistance derived in vitro) and short-term cultures (resistance derived in vivo) with hitherto characterized mechanisms of acquired resistance to PLX4032 to define the optimal combination of molecular target inhibition necessary to achieve efficient growth inhibition and cytotoxicity. We focused on a set of molecular targets downstream of overexpressed PDGFRβ or mutant N-RAS in each subset and used specific small molecule inhibitors, singly or in combination, to correlate growth inhibitory responses with p-ERK, p-AKT, p-p70S6K levels and apoptotic responses. We found that single inhibitor treatment led to functionally significant compensatory activation or rebound activities, consistent with dynamic network connectivity at key nodes of signaling. In PDGFRβ-upregulated, PLX4032-resistant melanoma cell lines, combined B-RAF, PI3K and TORC1/2 inhibition led to the strongest synergistic growth inhibitory and cytotoxic responses. In N-RAS-upregulated, PLX4032-resistant cell lines, either MEK or dual TORC1/2 inhibition sufficed to yield significant growth inhibition, and combined MEK and TORC1/2 inhibition did not synergize in growth inhibitory or cytotoxic responses. Together, these observations could serve to rationally guide animal model studies and urgently needed clinical testing in pre-identified subsets of patients relapsing on V600EB-RAF inhibitors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 634. doi:10.1158/1538-7445.AM2011-634
    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 ...
  • 5
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 2, No. 5 ( 2012-05-01), p. 414-424
    Abstract: BRAF inhibitors (BRAFi) induce antitumor responses in nearly 60% of patients with advanced V600E/KBRAF melanomas. Somatic activating MEK1 mutations are thought to be rare in melanomas, but their potential concurrence with V600E/KBRAF may be selected for by BRAFi. We sequenced MEK1/2 exon 3 in melanomas at baseline and upon disease progression. Of 31 baseline V600E/KBRAF melanomas, 5 (16%) carried concurrent somatic BRAF/MEK1 activating mutations. Three of 5 patients with BRAF/MEK1 double-mutant baseline melanomas showed objective tumor responses, consistent with the overall 60% frequency. No MEK1 mutation was found in disease progression melanomas, except when it was already identified at baseline. MEK1-mutant expression in V600E/KBRAF melanoma cell lines resulted in no significant alterations in p-ERK1/2 levels or growth-inhibitory sensitivities to BRAFi, MEK1/2 inhibitor (MEKi), or their combination. Thus, activating MEK1 exon 3 mutations identified herein and concurrent with V600E/KBRAF do not cause BRAFi resistance in melanoma. Cancer Discov; 2(5); 414–24. ©2012 AACR. Significance: As BRAF inhibitors gain widespread use for treatment of advanced melanoma, biomarkers for drug sensitivity or resistance are urgently needed. We identify here concurrent activating mutations in BRAF and MEK1 in melanomas and show that the presence of a downstream mutation in MEK1 does not necessarily make BRAF–mutant melanomas resistant to BRAF inhibitors. Read the Commentary on this article by Paraiso and Smalley, p. 390. This article is highlighted in the In This Issue feature, p. 377.
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 4, No. 1 ( 2014-01-01), p. 69-79
    Abstract: BRAF inhibitor (BRAFi) therapy leads to remarkable anti melanoma responses, but the initial tumor shrinkage is commonly incomplete, providing a nidus for subsequent disease progression. Adaptive signaling may underlie early BRAFi resistance and influence the selection pattern for genetic variants, causing late, acquired resistance. We show here that BRAFi (or BRAFi + MEKi) therapy in patients frequently led to rebound phosphorylated AKT (p-AKT) levels in their melanomas early on-treatment. In cell lines, BRAFi treatment led to rebound levels of receptor tyrosine kinases (RTK; including PDGFRβ), phosphatidyl (3,4,5)-triphosphate (PIP3), pleckstrin homology domain recruitment, and p-AKT. PTEN expression limited this BRAFi-elicited PI3K–AKT signaling, which could be rescued by the introduction of a mutant AKT1 (Q79K) known to confer acquired BRAFi resistance. Functionally, AKT1Q79K conferred BRAFi resistance via amplification of BRAFi-elicited PI3K–AKT signaling. In addition, mitogen-activated protein kinase pathway inhibition enhanced clonogenic growth dependency on PI3K or AKT. Thus, adaptive or genetic upregulation of AKT critically participates in melanoma survival during BRAFi therapy. Significance: This study provides a mechanistic link between early, adaptive and late, acquired BRAF inhibitor resistance in melanoma, with early BRAFi-induced signaling alterations shaping the subsequent evolutionary selective pressure. These findings argue for upfront, combined targeting of the mutant BRAF genotype and a pervasive drug-adaptive, AKT-dependent tumor response. Cancer Discov; 4(1); 69–79. ©2013 AACR. See related commentary by Solit and Rosen, p. 27 This article is highlighted in the In This Issue feature, p. 1
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Nature, Springer Science and Business Media LLC, Vol. 480, No. 7377 ( 2011-12-15), p. 387-390
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. LB-428-LB-428
    Abstract: Over 50% of melanomas harbor activating mutations in BRAF, most commonly BRAF(V600E). Profound clinical activity has been observed with RAF inhibitors, including vemurafenib, which is now FDA approved for the treatment of patients with advanced melanomas whose tumors harbor a BRAF(V600E) mutation. RAF inhibitors affect ERK signaling in a mutation-specific manner: they inhibit ERK signaling in cells with BRAF(V600E), but paradoxically activate ERK signaling in cells with wild-type BRAF. We recently elucidated the mechanism of this phenomenon: activation of RAS promotes the dimerization of members of the RAF family. At non-saturating concentrations, binding of an ATP-competitive RAF inhibitor to one member of the dimer inhibits it, while also causing its transition to the activated state. This is associated with the allosteric transactivation of the other, non-drug bound member of the dimer. This leads to an overall increase in RAF specific activity and induction of ERK signaling. In BRAF(V600E) melanomas, RAS-GTP levels are low, BRAF(V600E) is found primarily as a monomer and RAF inhibitors effectively inhibit active BRAF(V600E) monomers. This model of RAF transactivation by RAF inhibitors predicts that any molecular lesion that enhances RAF dimerization will promote resistance to RAF inhibitors. To investigate mechanisms of acquired resistance to RAF inhibitors, we treated a sensitive BRAF(V600E) expressing melanoma cell line (SKME239) with the RAF inhibitor vemurafenib for 8 weeks and selected for resistant clones. We found that a subset of cells resistant to vemurafenib expressed a splicing variant form of BRAF(V600E) that lacked exons 4-8, a region that encompasses the RAS-binding domain. This form of BRAF(V600E) had a size of approximately 61KD (p61BRAF(V600E)). p61BRAF(V600E) exhibited enhanced dimerization as compared to full length BRAF(V600E) in cells with low levels of RAS-GTP. Ectopic expression of p61BRAF(V600E) conferred resistance to the RAF inhibitor. Moreover, a mutation that disrupts dimerization of p61BRAF(V600E) restored its sensitivity to vemurafenib. In tumors from patients that relapsed on vemurafenib we identified various splicing variants of BRAF(V600E), all of them lacking the RAS-binding domain (6/19). Disease progression samples obtained from a mutually exclusive subset (4/19) of the same group of patients harbored activating mutation in NRAS. We report the first RAF-inhibitor resistance mechanism that involves a structural change in BRAF and the first kinase inhibitor resistance mechanism that involves expression of aberrant splicing variants of the drug target. Tumors resistant to RAF inhibitors resulting from increased RAF dimerization retain sensitivity to inhibitors of downstream effectors of RAF such as MEK. Therefore, MEK inhibitors, if used in combination with RAF inhibitors, may delay or prevent the onset of this mechanism of resistance. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-428. doi:1538-7445.AM2012-LB-428
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Nature, Springer Science and Business Media LLC, Vol. 468, No. 7326 ( 2010-12), p. 973-977
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2010
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 4, No. 1 ( 2014-01-01), p. 80-93
    Abstract: BRAF inhibitors elicit rapid antitumor responses in the majority of patients with BRAFV600-mutant melanoma, but acquired drug resistance is almost universal. We sought to identify the core resistance pathways and the extent of tumor heterogeneity during disease progression. We show that mitogen-activated protein kinase reactivation mechanisms were detected among 70% of disease-progressive tissues, with RAS mutations, mutant BRAF amplification, and alternative splicing being most common. We also detected PI3K–PTEN–AKT–upregulating genetic alterations among 22% of progressive melanomas. Distinct molecular lesions in both core drug escape pathways were commonly detected concurrently in the same tumor or among multiple tumors from the same patient. Beyond harboring extensively heterogeneous resistance mechanisms, melanoma regrowth emerging from BRAF inhibitor selection displayed branched evolution marked by altered mutational spectra/signatures and increased fitness. Thus, melanoma genomic heterogeneity contributes significantly to BRAF inhibitor treatment failure, implying upfront, cotargeting of two core pathways as an essential strategy for durable responses. Significance: This study provides critical insights into how human BRAF-mutant melanoma, a malignancy with marked mutational burden, escapes from BRAF inhibitors. Understanding the core resistance pathways as well as tumor heterogeneity, fitness, and mutational patterns, which emerge under drug selection, lays a foundation to rationalize clinical studies and investigate mechanisms of disease progression. Cancer Discov; 4(1); 80–93. ©2013 AACR. See related commentary by Solit and Rosen, p. 27 This article is highlighted in the In This Issue feature, p. 1
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2607892-2
    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...