Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (21)
  • 1
    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 ...
  • 2
    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 ...
  • 3
    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 ...
  • 4
    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 ...
  • 5
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. CT090-CT090
    Abstract: Background: Inhibition of the vascular endothelial growth factor receptor-2 (VEGFR-2) blocks angiogenesis and attenuates tumor growth, but cancers may evade this effect through activation of MET, the hepatocyte growth factor (HGF) receptor. Preclinical data suggest that co-targeting VEGFR and MET/HGF leads to greater tumor shrinkage when compared to a VEGFR blockade alone. Here we report the safety and clinical activity observed during dose escalation and tumor-specific expansion cohorts for the combination of the anti-VEGFR-2 antibody ramucirumab (R) and emibetuzumab (E), a bivalent monoclonal anti-MET antibody that inhibits ligand-dependent and ligand-independent MET signaling (NCT02082210). Methods: Patients (pts) with advanced solid tumors, measurable disease, and a fresh pretreatment tumor biopsy were enrolled in a Phase 1b/2 study receiving either 750 or 2000 mg E in combination with 8 mg/kg R, both intravenously administered every two weeks (Q2W). Following dose escalation, pts were assigned to tumor-specific expansion cohorts including gastric/gastroesophageal junction adenocarcinoma (G/GEJ), renal cell cancer (RCC), and non-small cell lung cancer (NSCLC). The primary objective was to determine the dose range of E that can be safely given in combination with R. Additional objectives included evaluation of anti-tumor activity by RECIST v1.1, pharmacokinetic (PK) parameters, immunogenicity, and biomarkers for MET/HGF and VEGF pathways. Results: As of Sept 2016, 6 pts were treated in the dose escalation and 45 pts in the cohort expansions (n=17 G/GEJ, n=15 RCC, n=13 NSCLC) with a median of 2 prior systemic therapies (range 1-7). No dose-limiting toxicities were observed in the dose escalation, and 750 mg E and 8 mg/kg R were recommended for the tumor expansion in line with the recommended phase 2 dose in other ongoing E phase 2 studies. Common possibly related AEs of all grades included fatigue (33%), nausea (20%), and dyspnea (16%). Grade 3 or higher AEs observed in more than 1 patient were fatigue (n=3) and pulmonary embolism (n=3). Disease control rates in the cohort expansions for G/GEJ, RCC, and NSCLC pts were 53%, 47%, and 85%, respectively. Unconfirmed partial responses were observed in an ovarian and a gastric pt. The median progression-free survival was: 2.73 months (95% CI: 1.35, 4.60) in the G/GEJ cohort, 4.17 months (95% CI: 1.18, 7.39) in the RCC cohort, and 6.57 months (95% CI: 2.76, 10.61) in the NSCLC cohort. Pretreatment tumoral MET expression and other biomarker analyses are ongoing and will be presented at the meeting. Conclusions: Treatment with E at a dose of 750 or 2000 mg can be safely administered with 8 mg/kg R Q2W with minimal toxicity and shows a potential to stabilize disease in G/GEJ, RCC, and NSCLC pts. Citation Format: Johanna Bendell, Charles Fuchs, Martin Voss, Todd M. Bauer, Toni K. Choueiri, Alexander Drilon, Katharine Thorn, Sameera Wijayawardana, Brian Moser, Arantxa Uruñuela, Volker Wacheck, James J. Harding. A phase 1b/2 study of ramucirumab in combination with emibetuzumab in patients with advanced solid tumors [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 CT090. doi:10.1158/1538-7445.AM2017-CT090
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2004
    In:  Clinical Cancer Research Vol. 10, No. 12 ( 2004-06-15), p. 4185-4191
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 10, No. 12 ( 2004-06-15), p. 4185-4191
    Abstract: Purpose: Little is known about the role that Mcl-1, an antiapoptotic Bcl-2 family member, plays in solid tumor biology and susceptibility to anticancer therapy. We observed that the Mcl-1 protein is widely expressed in human sarcoma cell lines of different histological origin (n = 7). Because the expression of antiapoptotic Bcl-2 family proteins can significantly contribute to the chemoresistance of human malignancies, we used an antisense strategy to address this issue in sarcoma. Experimental Design: SCID mice (n = 6/group) received s.c. injections of SW872 liposarcoma cells. After development of palpable tumors, mice were treated by s.c.-implanted miniosmotic pumps prefilled with saline or antisense or universal control oligonucleotides (20 mg/kg/day for 2 weeks). On days 2, 6, and 10, mice were treated with low-dose cyclophosphamide (35 mg/kg i.p) or saline control. During the experiments, tumor weight was assessed twice weekly by caliper measurements. On day 14, animals were sacrificed. Tumors were weighed and fixed in formalin for immunohistochemistry and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling analysis. Results: Mcl-1 antisense oligonucleotides specifically reduced Mcl-1 protein expression but produced no reduction in tumor weight compared with saline-treated control animals. Cyclophosphamide monotreatment caused only modest tumor weight reduction compared with saline control. However, use of Mcl-1 antisense oligonucleotides combined with cyclophosphamide clearly enhanced tumor cell apoptosis and significantly reduced tumor weight by more than two-thirds compared with respective control treatments. Conclusion: A combination of Mcl-1 antisense oligonucleotides with low-dose cyclophosphamide provides a synergistic antitumor effect and might qualify as a promising strategy to overcome chemoresistance in human sarcoma.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    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 ...
  • 8
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 15, No. 10 ( 2016-10-01), p. 2344-2356
    Abstract: The PI3K/AKT/mTOR pathway is among the most frequently altered pathways in cancer cell growth and survival. LY3023414 is a complex fused imidazoquinolinone with high solubility across a wide pH range designed to inhibit class I PI3K isoforms and mTOR kinase. Here, we describe the in vitro and in vivo activity of LY3023414. LY3023414 was highly soluble at pH 2–7. In biochemical testing against approximately 266 kinases, LY3023414 potently and selectively inhibited class I PI3K isoforms, mTORC1/2, and DNA-PK at low nanomolar concentrations. In vitro, inhibition of PI3K/AKT/mTOR signaling by LY3023414 caused G1 cell-cycle arrest and resulted in broad antiproliferative activity in cancer cell panel screens. In vivo, LY3023414 demonstrated high bioavailability and dose-dependent dephosphorylation of PI3K/AKT/mTOR pathway downstream substrates such as AKT, S6K, S6RP, and 4E-BP1 for 4 to 6 hours, reflecting the drug's half-life of 2 hours. Of note, equivalent total daily doses of LY3023414 given either once daily or twice daily inhibited tumor growth to similar extents in multiple xenograft models, indicating that intermittent target inhibition is sufficient for antitumor activity. In combination with standard-of-care drugs, LY3023414 demonstrated additive antitumor activity. The novel, orally bioavailable PI3K/mTOR inhibitor LY3023414 is highly soluble and exhibits potent in vivo efficacy via intermittent target inhibition. It is currently being evaluated in phase I and II trials for the treatment of human malignancies. Mol Cancer Ther; 15(10); 2344–56. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 519-519
    Abstract: Background: MET amplification (amp) is a resistance mechanism to EGFR TKI treatment. Emibetuzumab, a bivalent MET antibody (Ab) blocks HGF binding to MET and internalizes the receptor. Combination of emibetuzumab with EGFR TKIs (erlotinib, AZD9291, CO1686) or EGFR Ab (necitumumab, cetuximab) was evaluated in 3 ER xenograft models. Methods: Model 1: ER cell line HCC827ERL with high focal MET amp, high pMET, EGFR ex19 del (no T790M) was created from parental HCC827 NSCLC (EGFR ex19 del, EGFR amp, no MET amp) by increasing concentration of erlotinib in vitro over 7 months. Model 2: ER cell line HCC827-A8 was derived from HCC827 parental xenograft tumor serially passed in vivo with long term treatment of gefitinib and erlotinib. HCC827-A8 cells express high focal MET amp, high pMET/AXL (Western blot) while retaining EGFR ex19 del (no T790M). Model 3: LU0858 was an ER patient-derived NSCLC xenograft tumor, with focal MET amp, EGFR L858R (no T790M). MET amp and EGFRmt was determined by FISH and LNA-PCR sequencing respectively. Compound dosing: emibetuzumab 20 mg/kg qw; necitumumab 4 mg/kg or 20 mg/kg biw; cetuximab 4 mg/kg biw; erlotinib 25 mg/kg qd; 5 mg/kg AZD9291 qd; 30 mg/kg CO1686 bid. Results: EGFR inhibitors, but not emibetuzumab showed significant single agent anti-tumor effect in xenograft tumors derived from non-MET amp HCC827 parental cells. In MET amp ER models, single agent emibetuzumab resulted in tumor growth inhibition in Model 1 (T/C= 51.7%-61.0%, p & lt;0.05) and 3 (T/C=2.8%, p & lt;0.05)] but no tumor regression, and no anti-tumor effect in Model 2. Where evaluated, EGFR inhibitors showed no anti-tumor effect in the 3 ER models as monotherapy, except necitumumab (20 mg/kg) in Model 1 (T/C = 36.2%, p & lt;0.05). However, combination of emibetuzumab with AZD9291, CO1686, necitumumab (20 mg/kg), or erlotinib resulted in 80.4%, 58.2%, 44.4%, 69.1% tumor regression respectively (p & lt;0.001) in Model 1, while emibetuzumab + cetuximab (4 mg/kg) resulted in tumor stasis (T/C=0.2%, p & lt;0.05). In Model 2, emibetuzumab + AZD9291 resulted in tumor stasis (T/C = 12.9%, p & lt;0.05). In Model 3, emibetuzumab + necitumumab (20 mg/kg) resulted in 80.1% tumor regression (p & lt;0.001). Conclusion: The three erlotinib resistant models with MET amp and retaining sensitizing EGFRmt (ex19 del or L858R), and no acquired T790M were found resistant to other EGFR inhibitors (Abs and TKIs). Emibetuzumab in combination with either EGFR TKI or Ab showed anti-tumor activity in MET amp ER xenograft models including tumor regression in 2 out of 3 models. The combination of emibetuzumab with erlotinib is being evaluated in NSCLC patients with EGFR activating mutation (NCT01897480). Citation Format: Suzane L. Um, Victoria L. Peek, Jennifer R. Stephens, Jessica A. Baker, Holly K. Cannon, Joel D. Cook, Isabella H. Wulur, Roger Agyei, Sudhakar Chintharlapalli, Robert J. Evans, William J. Feaver, Lysiane Huber, Linda N. Lee, Ling Liu, Liandong Ma, Ruslan Novosiadly, Volker Wacheck, Sau-Chi Betty Yan. Antitumor activity of MET antibody emibetuzumab (LY2875358) in combination with EGFR inhibitors in erlotinib resistant (ER) xenograft mouse models [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 519. doi:10.1158/1538-7445.AM2017-519
    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 ...
  • 10
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 16 ( 2011-08-15), p. 5322-5332
    Abstract: Purpose: In this study, we tested the antitumor activity of the dual phosphoinositide 3-kinase (PI3K)/mTOR inhibitor BEZ235 against gastric cancer in vitro and in vivo. Experimental Design: Gastric cancer cell lines (N87, MKN45, and MKN28) were incubated with BEZ235 and assessed for cell viability, cell cycle, and PI3K/mTOR target inhibition. In vivo, athymic nude mice were inoculated with N87, MKN28, or MKN45 cells and treated daily with BEZ235. 3′-Deoxy-3′-[18F]fluorothymidine ([18F] FLT) uptake was measured via small animal positron emission tomography (PET). Results: In vitro, BEZ235 dose dependently decreased the cell viability of gastric cancer cell lines. The antiproliferative activity of BEZ235 was linked to a G1 cell-cycle arrest. In vivo, BEZ235 treatment resulted in PI3K/mTOR target inhibition as shown by dephosphorylation of AKT and S6 protein in all xenograft models. However, BEZ235 treatment only inhibited tumor growth of N87 xenografts, whereas no antitumor effect was observed in the MKN28 and MKN45 xenograft models. Sensitivity to BEZ235 in vivo correlated with downregulation of the proliferation marker thymidine kinase 1. Accordingly, [18F]FLT uptake was only significantly reduced in the BEZ235-sensitive N87 xenograft model as measured by PET. Conclusion: In conclusion, in vivo sensitivity of gastric cancer xenografts to BEZ235 did not correlate with in vitro antiproliferative activity or in vivo PI3K/mTOR target inhibition by BEZ235. In contrast, [18F]FLT uptake was linked to BEZ235 in vivo sensitivity. Noninvasive [18F] FLT PET imaging might qualify as a novel marker for optimizing future clinical testing of dual PI3K/mTOR inhibitors. Clin Cancer Res; 17(16); 5322–32. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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