Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Head & Neck, Wiley, Vol. 25, No. 9 ( 2003-09), p. 732-740
    Type of Medium: Online Resource
    ISSN: 1043-3074 , 1097-0347
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2003
    detail.hit.zdb_id: 2001440-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cellular Signalling, Elsevier BV, Vol. 16, No. 11 ( 2004-11), p. 1319-1327
    Type of Medium: Online Resource
    ISSN: 0898-6568
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2004
    detail.hit.zdb_id: 1496718-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancers, MDPI AG, Vol. 15, No. 16 ( 2023-08-09), p. 4034-
    Abstract: Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. Despite decades of clinical trials, the overall survival rate for patients with relapsed and metastatic disease remains below 30%, underscoring the need for novel treatments. FGFR4, a receptor tyrosine kinase that is overexpressed in RMS and mutationally activated in 10% of cases, is a promising target for treatment. Here, we show that futibatinib, an irreversible pan-FGFR inhibitor, inhibits the growth of RMS cell lines in vitro by inhibiting phosphorylation of FGFR4 and its downstream targets. Moreover, we provide evidence that the combination of futibatinib with currently used chemotherapies such as irinotecan and vincristine has a synergistic effect against RMS in vitro. However, in RMS xenograft models, futibatinib monotherapy and combination treatment have limited efficacy in delaying tumor growth and prolonging survival. Moreover, limited efficacy is only observed in a PAX3-FOXO1 fusion-negative (FN) RMS cell line with mutationally activated FGFR4, whereas little or no efficacy is observed in PAX3-FOXO1 fusion-positive (FP) RMS cell lines with FGFR4 overexpression. Alternative treatment modalities such as combining futibatinib with other kinase inhibitors or targeting FGFR4 with CAR T cells or antibody-drug conjugate may be more effective than the approaches tested in this study.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2023
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 8_Supplement ( 2023-04-14), p. CT192-CT192
    Abstract: Background: Futibatinib, an irreversible FGFR1-4 inhibitor, is indicated for the treatment of adult patients with previously treated, unresectable, locally advanced or metastatic intrahepatic cholangiocarcinoma harboring FGFR2 gene fusions or other rearrangements. As the primary elimination pathway for futibatinib is hepatic metabolism, we conducted a phase 1 study to evaluate the effect of hepatic impairment (HI) on futibatinib pharmacokinetics (PK) and safety in healthy adults. Methods: A single oral dose of 20 mg futibatinib was administered to adult subjects with mild (Child-Pugh score, 5-6), moderate (7-9), or severe (10-15) HI. Control healthy subjects were matched to each HI cohort according to age, body mass index, and sex. Intensive PK samples were collected up to 72 hours post-dose. Exposure measures (AUC0-inf, AUC0-t, and Cmax) in subjects with HI were compared with matching control cohorts and with the overall healthy-control cohort. Relationships between plasma PK and HI were examined graphically via scatter/regression plots of PK parameters versus baseline Child-Pugh score, bilirubin, albumin, international normalized ratio, and aspartate aminotransferase. Results: Overall, 38 subjects were enrolled (mild HI, n = 8; moderate HI, n = 8; severe HI, n = 6; healthy controls, n = 16). Following the administration of futibatinib, no trend was observed between the severity of HI and the extent of futibatinib exposure increase. Compared with matched controls, AUC0-inf increased by 21%, 20% and 18%, and Cmax by 43%, 15%, and 10% in subjects with mild, moderate, and severe HI, respectively. Changes in exposure were not considered clinically relevant as geometric mean ratios were within 80-125% bioequivalence limits, except for Cmax in subjects with mild HI (43%). Futibatinib PK parameters and HI measures did not appear to be associated based on visual inspection or statistical evaluation of regression plots (p-values all & gt; 0.05). No subjects discontinued from the study due to treatment-emergent adverse events (TEAEs). Overall, two (12.5%) subjects in the healthy-control cohort reported one Grade 1 TEAE each (dyspepsia and headache) and two (25.0 %) subjects in the mild HI cohort each reported one Grade 1 TEAE (toothache and headache). All TEAEs were considered related to treatment. No subjects with moderate/severe HI reported TEAEs. Conclusions: No clinically meaningful differences in the systemic plasma exposure of futibatinib were observed based on the severity of HI. Single oral doses of futibatinib were well tolerated among subjects with varying degrees of HI and matched healthy adult subjects in this study. The data suggest that dose adjustment may not be necessary in patients with HI receiving futibatinib 20 mg QD for its approved indication. Citation Format: Ling Gao, Ikuo Yamamiya, Mark Pinti, Juan Carlos Rondón, Thomas Marbury, Gareth Tomlinson, Lukas Makris, Nanae Hangai, Volker Wacheck. Phase 1, open-label, single-dose study to evaluate the effect of hepatic impairment on the pharmacokinetics and safety of futibatinib in adult subjects [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT192.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Molecular Cancer Therapeutics Vol. 8, No. 12_Supplement ( 2009-12-10), p. A124-A124
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 8, No. 12_Supplement ( 2009-12-10), p. A124-A124
    Abstract: Background: ERCC1 has been linked to resistance against platin-based chemotherapy. Cetuximab sensitizes colorectal cancer cells to oxaliplatin. This sensitization coincides with ERCC1 downregulation, but the link between EGFR and ERCC1 is not fully understood. It is known that treatment with cetuximab inhibits nuclear transport of EGFR and DNA repair in response to irradiation. Moreover, cisplatin treatment results in phosphorylation at Y845 and nuclear transport of EGFR. We intended to investigate the regulation of ERCC1 by cisplatin, cetuximab and dasatinib, a src inhibitor stabilizing EGFR in the membrane, its relevance for chemosensitivity and its correlation with EGFR and its phosphorylation at Y845 in head and neck cancer cells. Material and Methods: In UM-SCC 14c head and neck cancer cell line, ERCC1, EGFR and EGFR (pY845) expression was determined by western blot and FACS analysis. Cell viability, cell cycle and apoptosis induction was assessed by cell titer blue assay, propidium iodide and caspase 3 staining, respectively. Results: In cell viability assay, combination of cisplatin with cetuximab led to a significant (p & lt;0.05) and dose dependent sensitization to cisplatin. Cisplatin increased ERCC1 expression dose dependently. In contrast to previous reports, combination of cisplatin with cetuximab increased ERCC1 expression even stronger than the respective monotherapies. Mirroring the ERCC1 increase, we found a dose dependent increase of EGFR (pY845) after cisplatin and cetuximab treatment and the combination treatment led to an increased phosphorylation compared to monotherapy. Dasatinib decreased EGFR (pY845) but clearly increased EGFR and ERCC1 in parallel. To establish whether the findings after cisplatin and cetuximab treatment were EGF-receptor dependent we treated peripheral blood leukocytes, lacking the EGFR. No dose dependent regulation was observed by any monotherapy or combination of cetuximab and cisplatin. Conclusion: 14c cells are sensitized to cisplatin treatment by adding cetuximab. ERCC1 expression correlated with phosphorylation status of EGFR (pY845) following treatment with cisplatin and cetuximab. However, treatment with the src inhibitor dasatinib before adding cisplatin and cetuximab reduced phosphorylation of EGFR (Y845), but increased ERCC1 and EGFR expression. Whether the increase of ERCC1 is a late effect due to transcriptional upregulation of EGFR after dasatinib treatment remains to be determined. However, the dose-effect relationship and the lack of regulation of ERCC1 expression in peripheral blood leukocytes after treatment with cisplatin and cetuximab support the notion that ERCC1 expression is regulated by EGFR in 14c cells. Further studies elucidating the subcellular transport of EGFR and its transcriptional regulation after dasatinib treatment are ongoing and will be presented. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A124.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 2618-2618
    Abstract: Purpose: Genomic aberrations in fibroblast growth factor receptor (FGFR) are oncogenic drivers in several cancers. FGFR inhibitors (FGFRi) have demonstrated antitumor activity in cholangiocarcinoma (CCA) with FGFR2 fusions/rearrangements, though acquired resistance remains a therapeutic challenge and has been linked to FGFR2 mutations other than fusions/rearrangements. We sought to investigate mechanisms of acquired resistance to FGFRi and approaches to overcome resistance. Methods: Longitudinal plasma samples were collected from patients with FGFR pathway alterations enrolled in the futibatinib phase I trial (NCT02052778) and sequenced using a targeted, 73-gene panel. A separate retrospective analysis was conducted to evaluate possible evolution of genomic aberrations in CCA patients with FGFR2 fusion/rearrangement who had additional tumor and/or plasma next-generation sequencing (NGS) following FGFRi therapy. To assess the efficacy of futibatinib in cells with FGFR2 fusions in vitro, a FGFR2-BICC1 fusion H69 cholangiocyte cell line was developed. MAPK pathway alterations (BRAF_V600E or KRAS_G12D) were introduced to determine the impact of these co-alterations on FGFRi sensitivity, and combinations were tested to determine if efficacy could be enhanced. Cell viability assays, colony formation assays, and western blots were utilized to determine the effects of these agents in engineered cells. Results: A total of 58 plasma samples were collected from 17 patients with FGFR pathway alterations who were enrolled in the futibatinib phase I trial, including 13 (76.5%) of which had CCA. One patient with a FGFR2-CTNNA3 fusion who had NRAS G12D and BRAF A694T at baseline which were undetected during treatment had a dramatic increase in their variant allele frequency (VAF) upon progression (4.2%-0.0%-100% and 2.8%-0.0%-50.9%, respectively). Further, additional MAPK alterations were detected at time of progression, including BRAF V600E, NRAS Q61K, NRAS G12C, NRAS G13D and KRAS G12K mutations. In our separate retrospective series, of 17 patients who underwent repeat tumor and/or plasma NGS following treatment with one or more FGFRi, 10 (58.8%) had newly detectable alterations in MAPK pathway genes, 10 (58.8%) had new FGFR2 alterations, and 7 (41.2%) developed new alterations in both FGFR2 and MAPK pathway genes. In vitro studies demonstrated that in isogenic H69 biliary cell lines, introduction of FGFR2-BICC1 robustly sensitized to FGFRi when compared to a parental cell line, which was blunted by the introduction of secondary KRAS_G12D or BRAF_V600E mutations. Conclusions: Convergent genomic evolution in the MAPK pathway may be a potential mechanism of acquired resistance to FGFRi therapy. Work is ongoing to determine if targeting co-alterations may enhance the efficacy of FGFRi in FGFR2-fusion driven malignancies. Citation Format: TImothy P. DiPeri, Ming Zhao, Tyler Moss, Michael Kahle, Payal Rauli, Sunyoung S. Lee, Abdel Halim, Hiroshi Hirai, Volker Wacheck, Karim Benhadji, Jordi Rodon, Milind Javle, Funda Meric-Bernstam. Convergent MAPK pathway alterations mediate acquired resistance to FGFR inhibitors in cholangiocarcinoma with FGFR fusions/rearrangements [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 2618.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. CT020-CT020
    Abstract: Background: The phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway is aberrantly activated in malignant mesothelioma. LY3023414 (LY) is a potent oral ATP- competitive inhibitor that selectively and potently inhibits class I PI3K isoforms, mTOR, and DNA-PK. The recommended phase 2 dose (RP2D) of LY monotherapy was previously established to be 200 mg twice daily (BID). Here we report safety and clinical activity of LY monotherapy in advanced mesothelioma patients (pts) as part of a multi-cohort Phase 1 study (NCT01655225). Methods: In this cohort, 42 pts with malignant pleural or peritoneal mesothelioma with measurable disease as per RECIST v1.1, ECOG PS 0-1 and refractory or ineligible to standard therapies were enrolled to receive LY 200 mg BID. Primary objective was to explore single agent activity of LY. Other objectives included assessment of pharmacokinetics (PK), antitumor activity, and biomarker analysis. Results: 41 pts received treatment. Common treatment-related adverse events (TRAEs) included fatigue (44%), nausea (37%), vomiting (34%), decreased appetite (29%), diarrhea (22%) and maculo-papular rash (15%). Grade(G) 3/4 TRAEs reported were G3 fatigue (10%), G3 maculo-papular rash (7%), G3 pruritus (5%), G3 constipation (2%), G3 hyperglycemia (2%), G3 hyponatraemia (2%), G3 dyspnoea exertional (2%), G3 hypoxia (2%) and G4 hyperglycemia (2%). Serious adverse events related to study treatment were reported in 7(17%) pts. There were no deaths related to study treatment. Median duration of treatment was 12 weeks (range 3-48). In the 41 pts evaluable for tumor response, 2 pts (5%) had durable partial response (PR) and 17 pts (42%) had stable disease (SD) according to RECIST for an ORR of 5% and DCR of 46%. Overall, 29% of pts had a decrease in tumor lesions from baseline. The median progression-free survival for this cohort is 2.8 months (90% CI 2.5, 3.9). Alterations in the BAP1 gene, a potent tumor suppressor implicated in PI3K signaling pathway and in the pathogenesis of malignant mesothelioma, was identified in one pt with PR and in 4 of 17 pts (24%) with SD. Further biomarker analysis is ongoing. Conclusions: LY monotherapy at the RP2D of 200 mg BID demonstrated an acceptable and manageable safety profile. Single agent LY activity is limited in pts with advanced mesothelioma and more work is needed to identify the characteristics of benefitting patients. Citation Format: Marjorie G. Zauderer, David M. Hyman, Evan W. Alley, Johanna C. Bendell, Silvia Novello, Todd M. Bauer, Axel-Rainer Hanauske, Anna M. Szpurka, Suhyun Kang, Anindya Chatterjee, Volker Wacheck, Patrick Brueck, Anna M. Varghese. Safety and tolerability of the dual PI3K/mTOR inhibitor LY3023414 in treatment of patients with advanced mesothelioma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr CT020.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 8
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2019
    In:  Journal of Oncology Practice Vol. 15, No. 11 ( 2019-11), p. e925-e933
    In: Journal of Oncology Practice, American Society of Clinical Oncology (ASCO), Vol. 15, No. 11 ( 2019-11), p. e925-e933
    Abstract: Olaratumab is a human monoclonal immunoglobulin G1 antibody against platelet-derived growth factor receptor-α. We report the nature and frequency of infusion-related reactions (IRRs) with olaratumab in clinical trials and postmarketing reports. METHODS: Data from patients exposed to olaratumab across nine clinical trials were reviewed for IRRs. Blood samples were also analyzed for pre-existing immunoglobulin E anti–galactose-α-1,3-galactose (anti–α-Gal) antibodies. RESULTS: In the clinical trials, IRRs were identified in 70 of 485 patients (14.4%). The most frequent symptoms included flushing, fever or chills, and dyspnea. For 68 of 70 patients (97.1%), the first IRR occurred during the first two cycles of treatment. Grade 3 or worse IRRs were reported in 11 patients (2.3%), all during the first infusion and usually within 15 minutes of the start of the infusion. One IRR-related fatality (0.2%) occurred in a nonpremedicated patient with grade 3 or worse cardiac comorbidities. There was an association between grade 3 or worse IRRs and pre-existing anti–α-Gal antibodies, with a trend toward higher IRR rates in US geographies known to have a higher prevalence of anti–α-Gal antibodies. IRRs in postmarketing reports were consistent in nature and severity with those in the clinical trials. CONCLUSION: Premedication with corticosteroids and antihistamines should occur in all patients before olaratumab infusion, as indicated in labels in the United States and the European Union. Patients receiving olaratumab should be monitored for IRRs in a setting where resuscitation equipment is available for the treatment of IRRs.
    Type of Medium: Online Resource
    ISSN: 1554-7477 , 1935-469X
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2019
    detail.hit.zdb_id: 3005549-0
    detail.hit.zdb_id: 2236338-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 528-528
    Abstract: Background: MET ex14 skipping, present in ~3% of lung cancer, is a strong oncogenic driver which is further evidenced by case reports of patients response to MET TKI treatment. ~15% of tumors in patients that harbor MET ex14 skipping also have MET amplification (amp). Merestinib is a type II MET kinase inhibitor1. Emibetuzumab, a bivalent MET Ab, internalizes MET receptor. Each single agent and the combination were evaluated in the Hs746t gastric cancer line bearing MET ex14 skipping and MET amp. Methods: Each agent was evaluated in vitro for inhibition of Hs746t cell proliferation and pMET levels. In vivo study in Hs746t-derived xenograft mouse model (n=7 mice/ arm, 28 day dosing) initiated when tumors were 150-350mm3: merestinib at 6 mg/kg (suboptimal dose - insufficient target coverage for 24 hrs) or 12 mg/kg (optimal dose) qd orally, emibetuzumab at 10 mg/kg qw by IP. Results: Merestinib inhibited Hs746t cell proliferation with IC50=34 nM and totally eliminated pMET at 65-100 nM. Emibetuzumab slightly inhibited Hs746t cell proliferation (IC50 & gt;100 nM), reduced 10-20% cell surface MET, and no effect on pMET expression (at 130-650 nM). In the Hs746t xenograft model, merestinib (12 mg/kg) treatment resulted in 91.8% tumor regression after 21 day dosing, while 6 mg/kg merestinib provided transient tumor regression followed by re-growth while on treatment with T/C=18.3% after 21 day dosing. No tumor re-growth was observed in 6/7 mice in the 12 mg/kg merestinib cohort during the 5 weeks post-treatment. Emibetuzumab treatment provided transient tumor regression (37.7%) after 3 doses, but tumors re-grew while on treatment. Combination of 6 mg/kg merestinib and 10 mg/kg emibetuzumab resulted in 85% tumor regression for the duration of the 28 day dosing period and the treatment was well tolerated. Tumors in animals re-grew upon termination of this combination treatment. Conclusion: Merestinib (12 mg/kg) treatment resulted in durable and complete response in 6/7 mice bearing Hs746t tumors with MET ex14 skipping and MET amp. When used singly, merestinib (6 mg/kg) or emibetuzumab (10 mg/kg) resulted in only transient tumor regression in this model, while the combination resulted in substantial tumor regression while on treatment. This combination treatment was however, not as durable as was observed with single agent 12 mg/kg merestinib. Single agents and the combinations were well tolerated. As a type II MET inhibitor, merestinib may provide a therapeutic option to treatment naïve patients or those who have progressed on type I MET inhibitor treatment, whose tumors have MET exon 14 skipping and/or MET amplification 2,3. Data in this study support a clinical evaluation of merestinib in patients with MET exon 14 skipping (NCT02920996). 1 - Yan et al. Invest New Drugs 2013;31:833-844 2 - Ou et al. J Thorac Oncol. 2016; PMID:27666659 3 - Heist et al. J Thorac Oncol. 2016;11:1242-1245 Citation Format: Sau-Chi Betty Yan, Suzane L. Um, Victoria L. Peek, Jennifer R. Stephens, Wei Zeng, Bruce W. Konicek, Ling Liu, Volker Wacheck, Richard A. Walgren. Evaluation of single agent merestinib (LY2801653) or emibetuzumab (LY2875358) and the combination in a xenograft tumor model bearing MET exon 14 skipping [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 528. doi:10.1158/1538-7445.AM2017-528
    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
    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