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
    Online-Ressource
    Online-Ressource
    BMJ ; 2012
    In:  Thorax Vol. 67, No. 6 ( 2012-06), p. 477-487
    In: Thorax, BMJ, Vol. 67, No. 6 ( 2012-06), p. 477-487
    Materialart: Online-Ressource
    ISSN: 0040-6376 , 1468-3296
    Sprache: Englisch
    Verlag: BMJ
    Publikationsdatum: 2012
    ZDB Id: 1481491-2
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Microvascular Research, Elsevier BV, Vol. 147 ( 2023-05), p. 104479-
    Materialart: Online-Ressource
    ISSN: 0026-2862
    RVK:
    Sprache: Englisch
    Verlag: Elsevier BV
    Publikationsdatum: 2023
    ZDB Id: 1471172-2
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2021
    In:  Molecular Cancer Therapeutics Vol. 20, No. 12_Supplement ( 2021-12-01), p. P076-P076
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 20, No. 12_Supplement ( 2021-12-01), p. P076-P076
    Kurzfassung: ROS1 tyrosine kinase inhibitors (TKIs) entrectinib and crizotinib have significantly improved outcomes for patients with ROS1 gene fusion positive (ROS1+) lung adenocarcinoma. However, drug resistance inevitably develops, leading to disease progression. Approximately one-third of patients resistant to ROS1 TKIs demonstrate on-target ROS1 kinase domain (KD) mutations (Dziadziuszko et al. ESMO 2019), but the mechanism of resistance in the majority of patients remain unknown or poorly characterized. The purpose of this study was to model and characterize mechanisms of acquired ROS1 TKI resistance. To investigate resistance to ROS1 TKIs, we used patient-derived cell lines CUTO28 (TPM3-ROS1) and CUTO37 (CD74-ROS1) to generate drug-resistant derivatives to entrectinib (CUTO28-ER, CUTO37-ER) or crizotinib (CUTO28-CR,CUTO37-CR) in vitro. We utilized several techniques to probe the mechanisms driving resistance, including DNA and RNA sequencing, fluorescence in-situ hybridization (FISH), cell proliferation assays, and western blotting. DNA sequencing of the ROS1 KD in all resistant cell lines did not reveal any mutations. CUTO28-ER cells displayed sensitivity to MET-selective TKIs in cell proliferation assays. Consistent with MET dependency, MAPK and Akt pathways were inhibited by treatment with a MET-selective, but not a ROS1-selective, TKI. Western blot demonstrated MET overexpression and interphase FISH confirmed MET gene amplification (MET:CEP7 ratio 4.2) compared to parental cells (MET:CEP7 ratio 1.0). Notably, metaphase FISH revealed extrachromosomal DNA (ecDNA) amplification of MET. CUTO28-CR cells displayed sensitivity to Src family kinase (SFK) TKIs, and addition of pan-HER TKI afatinib resulted in synergistic inhibition of proliferation and downstream MAPK signaling. CUTO37-ER cells displayed sensitivity via proliferation and signaling to pan-HER inhibitor afatinib, and phosphorylation of EGFR Y845 consistent with Src-mediated activation of EGFR. Parental CUTO28 and CUTO37 cells were not sensitive to SFK or HER inhibition. Interphase FISH revealed modest EGFR gene amplification in CUTO28-CR cells—(EGFR:CEP7 ratio 2.1) compared to parental (EGFR:CEP7 ratio 1.0)—but not in CUTO37-ER or CUTO37-CR cells. HER2 gene amplification was not identified in any of the cell lines. Stimulation with epidermal growth factor (EGF) markedly enhanced MAPK signaling in CUTO28-CR, CUTO37-ER, and CUTO37-CR cells to a greater extent than in corresponding parental cells, consistent with EGFR or HER2 dependence. In conclusion, we demonstrated novel mechanisms of resistance to ROS1 TKIs entrectinib and crizotinib. These include ecDNA amplification of MET as an alternate oncogene driver and Src/EGFR axis as a driver of bypass signaling. Combination strategies with existing TKIs could be explored in patients with ROS1 TKI resistance. The discovery of ecDNA MET amplification is particularly intriguing; ecDNA is associated with more aggressive cancers and next generation sequencing analyses do not typically involve amplicon reconstruction to detect ecDNA. Citation Format: Logan C. Tyler, Anh T. Le, Hala Nijmeh, Liming Bao, Kristen Turner, Jason Christianson, Robert C. Doebele. Novel mechanisms of acquired TKI resistance in ROS1+ NSCLC [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr P076.
    Materialart: Online-Ressource
    ISSN: 1535-7163 , 1538-8514
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2062135-8
    SSG: 12
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Vascular Cell, Publiverse Online S.R.L, Vol. 3, No. 1 ( 2011), p. 4-
    Materialart: Online-Ressource
    ISSN: 2045-824X
    Sprache: Englisch
    Verlag: Publiverse Online S.R.L
    Publikationsdatum: 2011
    ZDB Id: 2595417-9
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: SSRN Electronic Journal, Elsevier BV
    Materialart: Online-Ressource
    ISSN: 1556-5068
    Sprache: Englisch
    Verlag: Elsevier BV
    Publikationsdatum: 2022
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3003-3003
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3003-3003
    Kurzfassung: ROS1 gene fusions are predominantly found in non-small cell lung cancer (NSCLC), and ROS1 gene fusion-positive (ROS1+) tumors comprise 1-2% of all diagnosed lung adenocarcinomas. Currently, there are two ROS1 tyrosine kinase inhibitors (TKIs) which are US FDA approved, crizotinib and entrectinib, both of which generate tumor response rates in excess of 70% and demonstrate prolonged disease control. However, acquired resistance to TKIs is inevitable, and has been reported in ROS1+ NSCLC tumors or cancer cell models by our group and others and include ROS1 kinase domain mutations (e.g., L1951R, S1986Y/F, F2004V, L2026M, and G2032R), activation of bypass signaling pathways (e.g., EGFR, RAS, or KIT), or phenotypic transformation (such as epithelial to mesenchymal transition). In order to better understand mechanisms of acquired TKI resistance in ROS1 NSCLC, our lab utilized primary, patient-derived cancer cell models harboring these rare oncogenic kinase rearrangements. Herein, using a patient-derived NSCLC cancer cell line (CUTO28) harboring a TPM3-ROS1 fusion, we derived an entrectinib-resistant cell line (CUTO28-ER) through in vitro culture under drug selection. In these CUTO28-ER cells, we identified: (1) MET-mediated bypass signaling as an acquired resistance mechanism to entrectinib, (2) upregulation of MET signaling is accomplished via MET gene amplification, and (3) resistance could be overcome by the dual ROS1/MET inhibitor crizotinib. To our knowledge, this is the first reported case of MET-mediated acquired resistance in a ROS1+ cancer. This cell line model finding was supported by another primary, patient-derived cell line (CUTO38) harboring a CD74-ROS1 fusion; this cell line was derived from a NSCLC patient following disease progression on entrectenib in the clinic. CUTO38 is resistant to entrectinib in vitro and displays elevated MET expression, MET activation with increased phosphorylation, and dependence on MET for cell proliferation and survival. Notably, MET activation in CUTO38 was not generated by gene amplification, MET exon 14 splicing, MET gene fusion, or autocrine HGF expression, suggesting alternate means of MET activation. These findings suggest that interrogation of MET gene amplification or other MET biomarkers should be performed in ROS1+ NSCLC patients who experience disease progression on entrectinib or other ROS1 TKIs that do not inhibit MET, since there are clinically available MET inhibitors, some of which are dual ROS1/MET inhibitors, that may overcome drug resistance in these patients. Citation Format: Logan Tyler, Anh Le, Hala Nijmeh, Robert Doebele. MET mediates entrectinib resistance in ROS1 gene fusion positiveNSCLC [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3003.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1103-1103
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1103-1103
    Kurzfassung: ROS1 TKIs entrectinib and crizotinib have significantly improved outcomes for ROS1+ lung adenocarcinoma patients. However, drug resistance inevitably develops, leading to disease progression. Approximately 1/3 of patients resistant to ROS1 TKIs demonstrate ROS1 kinase domain (KD) mutations (Dziadziuszko et al. ESMO 2019), but the mechanism of resistance in most patients is unknown or poorly characterized. To model and characterize acquired resistance to ROS1 TKIs, we used patient-derived cell line CUTO28 (TPM3-ROS1) to generate an entrectinib-resistant derivative (CUTO28-ER) in vitro. We utilized several techniques to probe the mechanisms driving resistance: DNA and RNA sequencing (seq), fluorescence in-situ hybridization (FISH), cell proliferation assays, and western blotting. DNA seq of the ROS1 KD in CUTO28-ER failed to reveal mutations. CUTO28-ER cells displayed sensitivity to MET-selective TKIs in proliferation assays, and MAPK and AKT pathways were inhibited only with MET-selective TKI. RNA seq and western blot showed MET overexpression, and interphase FISH confirmed MET amplification compared to parental cells (MET:CEP7 ratio 4.2 vs 1.0). We substantiated in vitro findings in patient tissue, utilizing tumor samples at 2 different points of a single CD74-ROS1 NSCLC patient’s tumor progression. The first tumor sample did not display MET amplification while on ROS1 TKI (MET:CEP7 ratio 0.9, 3.4 copies of MET). However, the second tumor sample collected 5 months later displayed a MET:CEP7 ratio of 2.5 (9.5 copies of MET), indicating progression on ROS1 TKI was likely MET-driven. To determine prevalence of MET amplification in entrectinib-resistant ROS1+ NSCLC, we analyzed circulating tumor DNA (ctDNA) by FoundationOne Liquid CDx from patients with ROS1+ NSCLC in the STARTRK-2 entrectinib trial. Of 105 ROS1+ NSCLC patients with ctDNA analysis both at enrollment and progression, 2 (1.9%) displayed copy number amplification (CNA) of MET. Of these, 1 patient had no detectible CNA at study baseline but MET CNA gain by day 166 of entrectinib therapy and the other had detectable MET CNA gains both at baseline and at progression only 28 days later. Both received 3 lines of therapy prior to entrectinib, none of which targeted ROS1 or MET. In conclusion, we demonstrated MET gene amplification as a potential mechanism of resistance to ROS1 TKI entrectinib. The prevalence of MET amplification at resistance was 2 of 105, but may be greater than was detected in STARTRK-2 due to the sensitivity of ctDNA assays and challenges of measuring CNA in ctDNA. Utilization of the ROS1/MET TKI crizotinib or combination of entrectinib with capmatinib should be explored in patients with ROS1+ NSCLC that display MET CNA to overcome MET-driven resistance following entrectinib. Citation Format: Logan C. Tyler, Anh T. Le, Hala Nijmeh, Liming Bao, Timothy R. Wilson, Brian Simmons, David Chen, Robert C. Doebele. MET gene amplification is a mechanism of resistance to entrectinib in ROS1+ NSCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1103.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2022
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    In: American Journal of Physiology-Cell Physiology, American Physiological Society, Vol. 300, No. 2 ( 2011-02), p. C266-C275
    Kurzfassung: Extracellular ATP and ADP have been shown to exhibit potent angiogenic effects on pulmonary artery adventitial vasa vasorum endothelial cells (VVEC). However, the molecular signaling mechanisms of extracellular nucleotide-mediated angiogenesis remain not fully elucidated. Since elevation of intracellular Ca 2+ concentration ([Ca 2+ ] i ) is required for cell proliferation and occurs in response to extracellular nucleotides, this study was undertaken to delineate the purinergic receptor subtypes involved in Ca 2+ signaling and extracellular nucleotide-mediated mitogenic responses in VVEC. Our data indicate that stimulation of VVEC with extracellular ATP resulted in the elevation of [Ca 2+ ] i via Ca 2+ influx through plasma membrane channels as well as Ca 2+ mobilization from intracellular stores. Moreover, extracellular ATP induced simultaneous Ca 2+ responses in both cytosolic and nuclear compartments. An increase in [Ca 2+ ] i was observed in response to a wide range of purinergic receptor agonists, including ATP, ADP, ATPγS, ADPβS, UTP, UDP, 2-methylthio-ATP (MeSATP), 2-methylthio-ADP (MeSADP), and BzATP, but not adenosine, AMP, diadenosine tetraphosphate, αβMeATP, and βγMeATP. Using RT-PCR, we identified mRNA for the P2Y1, P2Y2, P2Y4, P2Y13, P2Y14, P2X2, P2X5, P2X7, A1, A2b, and A3 purinergic receptors in VVEC. Preincubation of VVEC with the P2Y1 selective antagonist MRS2179 and the P2Y13 selective antagonist MRS2211, as well as with pertussis toxin, attenuated at varying degrees agonist-induced intracellular Ca 2+ responses and activation of ERK1/2, Akt, and S6 ribosomal protein, indicating that P2Y1 and P2Y13 receptors play a major role in VVEC growth responses. Considering the broad physiological implications of purinergic signaling in the regulation of angiogenesis and vascular homeostasis, our findings suggest that P2Y1 and P2Y13 receptors may represent novel and specific targets for treatment of pathological vascular remodeling involving vasa vasorum expansion.
    Materialart: Online-Ressource
    ISSN: 0363-6143 , 1522-1563
    Sprache: Englisch
    Verlag: American Physiological Society
    Publikationsdatum: 2011
    ZDB Id: 1477334-X
    SSG: 12
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    Online-Ressource
    Online-Ressource
    American Physiological Society ; 2014
    In:  American Journal of Physiology-Lung Cellular and Molecular Physiology Vol. 306, No. 7 ( 2014-04-01), p. L661-L671
    In: American Journal of Physiology-Lung Cellular and Molecular Physiology, American Physiological Society, Vol. 306, No. 7 ( 2014-04-01), p. L661-L671
    Kurzfassung: Angiogenic expansion of the vasa vasorum (VV) is an important contributor to pulmonary vascular remodeling in the pathogenesis of pulmonary hypertension (PH). High proliferative potential endothelial progenitor-like cells have been described in vascular remodeling and angiogenesis in both systemic and pulmonary circulations. However, their role in hypoxia-induced pulmonary artery (PA) VV expansion in PH is not known. We hypothesized that profound PA VV neovascularization observed in a neonatal calf model of hypoxia-induced PH is due to increased numbers of subsets of high proliferative cells within the PA adventitial VV endothelial cells (VVEC). Using a single cell clonogenic assay, we found that high proliferative potential colony-forming cells (HPP-CFC) comprise a markedly higher percentage in VVEC populations isolated from the PA of hypoxic (VVEC-Hx) compared with control (VVEC-Co) calves. VVEC-Hx populations that comprised higher numbers of HPP-CFC also demonstrated markedly higher expression levels of CD31, CD105, and c-kit than VVEC-Co. In addition, significantly higher expression of CD31, CD105, and c-kit was observed in HPP-CFC vs. the VVEC of the control but not of hypoxic animals. HPP-CFC exhibited migratory and tube formation capabilities, two important attributes of angiogenic phenotype. Furthermore, HPP-CFC-Co and some HPP-CFC-Hx exhibited elevated telomerase activity, consistent with their high replicative potential, whereas a number of HPP-CFC-Hx exhibited impaired telomerase activity, suggestive of their senescence state. In conclusion, our data suggest that hypoxia-induced VV expansion involves an emergence of HPP-CFC populations of a distinct phenotype with increased angiogenic capabilities. These cells may serve as a potential target for regulating VVEC neovascularization.
    Materialart: Online-Ressource
    ISSN: 1040-0605 , 1522-1504
    Sprache: Englisch
    Verlag: American Physiological Society
    Publikationsdatum: 2014
    ZDB Id: 1477300-4
    SSG: 12
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    In: npj Precision Oncology, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2023-01-23)
    Kurzfassung: Patients with metastatic NSCLC bearing a ROS1 gene fusion usually experience prolonged disease control with ROS1-targeting tyrosine kinase inhibitors (TKI), but significant clinical heterogeneity exists in part due to the presence of co-occurring genomic alterations. Here, we report on a patient with metastatic NSCLC with a concurrent ROS1 fusion and KRAS p.G12C mutation at diagnosis who experienced a short duration of disease control on entrectinib, a ROS1 TKI. At progression, the patient continued entrectinib and started sotorasib, a small molecule inhibitor of KRAS p.G12C. A patient-derived cell line generated at progression on entrectinib demonstrated improved TKI responsiveness when treated with entrectinib and sotorasib. Cell-line growth dependence on both ROS1 and KRAS p.G12C was further reflected in the distinct downstream signaling pathways activated by each driver. Clinical benefit was not observed with combined therapy of entrectinib and sotorasib possibly related to an evolving KRAS p.G12C amplification identified on repeated molecular testing. This case supports the need for broad molecular profiling in patients with metastatic NSCLC for potential therapeutic and prognostic information.
    Materialart: Online-Ressource
    ISSN: 2397-768X
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2023
    ZDB Id: 2891458-2
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie auf den KOBV Seiten zum Datenschutz