Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 845-845
    Abstract: IGF-1 is a well-known growth factor for human myeloma cells. Thus, therapeutic strategies targeting IGF-1R have been proposed for the treatment of multiple myeloma. In this study, we investigated the effect of a humanized antagonistic monoclonal antibody to the IGF-1R (AVE1642). We showed that AVE1642 selectively inhibits IGF-1R but not insulin signaling in human myeloma cell lines (HMCL). Since we had previously shown the functional relevance of CD45 expression in the growth of myeloma cells, and the association of CD45 negative (CD45neg) status with a less favorable clinical outcome, both CD45 positive (CD45pos) and CD45neg HMCL were studied. We found that AVE1642 strongly inhibited the growth of CD45neg HMCL, leading to a G1 growth arrest, whereas it had almost no effect on the growth of CD45pos HMCL. Indeed, by cell counting over four days in standard culture conditions, AVE1642 induced a significant growth inhibition of the CD45neg LP-1 HMCL of 90% whereas it had no effect on the growth of the CD45pos XG-1 HMCL. Furthermore, AVE1642 binding induced a significant reduction of IGF-1R expression. In western blot, a decrease of 60% the IGF-1R expression of the LP-1 CD45neg HMCL was observed. This decrease was also confirmed by flow cytometry analysis. Finally, a stable CD45 silencing XG-1 cell line was generated and became sensitive to AVE1642. By cell counting over four days in standard culture conditions, AVE1642 inhibited the growth of the shCD45neg XG-1 HMCL of 56% whereas it had almost no effect on the growth of the shLuci-XG-1 HMCL. Thus, for the first time, we provided direct evidence that the expression of CD45 renders myeloma cells resistant to IGF1R inhibition. Furthermore, AVE1642 in combination with Bortezomib strongly increased the apoptosis induced by Bortezomib alone on CD45negHMCL. The CD45neg LP-1 HMCL was considered as a modestly sensitive cell line since 10nM of Bortezomib, which is in the range of therapeutic concentrations, induced 40±16% of apoptosis. Bortezomib in combination with AVE1642 was found to induce 80±1% of apoptosis in the CD45neg LP-1 HMCL. Apoptosis induction by AVE1642/Bortezomib combination was associated with an important induction of Noxa, a BH3-only pro-apoptotic protein that was increased by the addition of AVE1642. We also found a strong increase of caspase-3, -8 activation under this combination compared to Bortezomib alone. Taken together, these results support that therapy directed against IGF-1R associated with Bortezomib, could be beneficial in treating CD45neg patients, a population known for poor clinical outcome.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Molecular and Cellular Biology, Informa UK Limited, Vol. 23, No. 13 ( 2003-07-01), p. 4586-4597
    Type of Medium: Online Resource
    ISSN: 1098-5549
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2003
    detail.hit.zdb_id: 1474919-1
    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. 72, No. 8_Supplement ( 2012-04-15), p. 1786-1786
    Abstract: The receptor tyrosine kinase MET is a prime target in clinical oncology because of its aberrant activation in a broad spectrum of malignancies. Here, we tested the anti tumor activity of a novel MET inhibitor, EMD1214063, on cancer cells overexpressing MET and in eight transfected NIH3T3 cell lines expressing different activating mutants of the MET kinase. Our results demonstrate a dose-dependent decrease in MET autophosphorylation in both MET overexpressing cell lines (IC50 of 20nM and 30nM for GTL-16 and H1993, respectively) and in six of the eight cell lines transfected with MET activating mutants (IC50 5-18nM). Blockade of MET by EMD1214063 resulted in reduced downstream activation of AKT, ERK and PLCγ. In contrast, lack of MET inhibition by EMD1214063 in the resistant Y1248H and L1213V MET mutants was associated with high levels of AKT, ERK and PLCγ activation. In all sensitive mutants, EMD1214063 attenuated MET-dependent cellular proliferation and significantly altered cell cycle distribution profiles (increase in G1 and concomitant decrease of S phase). Furthermore, EMD1214063 strongly affected MET-driven biological functions, such as cellular morphology, motility and anchorage-independent growth. To assess the in vivo efficacy of EMD1214063, we established tumor transplantation models exploiting NIH3T3 cells expressing the H1112L or L1213V MET mutants, respectively sensitive and resistant, to in vitro treatment with EMD1214063. Animals bearing H1112L or L1213V tumors were randomized for the treatment with EMD1214063 (50mg/kg/day) or vehicle only. Remarkably, 5 days of treatment with EMD1214063 induced complete regression of sensitive H1112L tumors, while tumor growth remained unaffected in mice injected with resistant L1213V tumor cells and in vehicle-treated mice. Taken together, these results underscore the concordance between in vitro and in vivo effects of EMD1214063 and strongly support its efficacy in inhibiting MET-driven tumor cell growth. 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 1786. doi:1538-7445.AM2012-1786
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 925-925
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 925-925
    Abstract: The mesenchymal-epithelial transition factor (c-Met) receptor, also known as hepatocyte growth factor receptor (HGFR), controls morphogenesis, a process physiologically required for embryonic development and tissue repair. Aberrant c-Met activation is associated with human malignancies, including hepatocellular carcinomas (HCC). The aim of this study was to evaluate the effect of a novel, highly selective c-Met inhibitor, MSC2156119J (EMD 1214063), in hepatocellular cancer models. MHCC97H is a human HCC cell line co-expressing c-Met and HGF, and exhibiting high metastatic potential. The effect of MSC2156119J on MHCC97H was evaluated in a Balb/c nu/nu murine xenograft model in a subcutaneous and orthotopic setting. Tumor-bearing mice were orally treated with 100 mg/kg MSC2156119J (5 days on/2 days off). In the subcutaneous setting, MSC2156119J resulted in tumor regression in 10/10 mice, achieving tumor eradication in 9/10 mice. In the orthotopic model, MHCC97H tumor fragments proliferate in the liver and invariably metastasize to the lung (100%). MSC2156119J treatment started 7 days after tumor fragment implantation. After 5 weeks of treatment, mice were necropsied and a series of parameters were assessed. Primary tumor size and weight, and circulating Alpha-Feto-Protein levels were significantly lower in mice treated with MSC2156119J compared to controls (p & lt;0.001). Notably, treatment with MSC2156119J resulted in a reduced number of mice with visible lung metastasis (6/9), and correlated with a significantly lower number of metastatic foci in the lungs compared to control mice (p & lt;0.01). To better predict the clinical efficacy of MSC2156119J, 9 patient-derived primary HCC explants models were used. The explants were subdivided into 3 groups, based on their c-Met levels and HGF expression. Each group comprised 3 tumors expressing high, intermediate, and low levels of c-Met and HGF, respectively. Mice were treated with MSC2156119J, sorafenib, or a combination of both. MSC2156119J (100 mg/kg/5 out of 7 days) strongly inhibited tumor growth in 4/9 models (TGD range of 370% to 41%). Analysis of intratumoral c-Met, phospho c-Met and HGF levels indicated that explants with high levels of c-Met and HGF were more sensitive to MSC2156119J than low-expressing models. In 3/4 responsive models, MSC2156119J exhibited a better anti-tumor activity than sorafenib, while sorafenib was more efficacious in a model characterized by intermediate c-Met/HGF expression. MSC2156119J was not efficacious in models exhibiting low or no signs of c-Met signalling. When used in combination, MSC2156119J did not enhance the activity of sorafenib. MSC2156119J monotherapy was well tolerated while sorafenib alone and in combination with MSC2156119J induced significant body weight loss. Overall, these data indicate that MSC2156119J may be a valuable therapeutic option for liver cancers with high levels of c-Met expression/activation. Citation Format: Friedhelm Bladt, Andree Blaukat, Dieter Dorsch, Manja Friese-Hamim, Michael Meyring, Oliver Schadt. The c-Met inhibitor MSC2156119J effectively inhibits growth of liver cancer models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 925. doi:10.1158/1538-7445.AM2013-925
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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. 71, No. 8_Supplement ( 2011-04-15), p. 2786-2786
    Abstract: The involvement of the mesenchymal endothelial transition factor (c-Met) in the primary event of oncogenic transformation and the secondary ability to mediate metastatic spread has been convincingly demonstrated in preclinical and early clinical settings. The clinical benefits of c-Met kinase inhibitors with various modes of actions and selectivity profiles are currently under investigation hoping that inhibitors of c-Met might emerge as valuable cancer therapeutics in the future. During an HTS run 3-(diethylamino)propyl N-[3-[[5-(3,4-dimethoxyphenyl)-2-oxo-6H-1,3,4-thiadiazin-3-yl]methyl] phenyl]carbamate was identified as an attractive lead structure with an interesting overall profile (clogD (7.4): 2.5, S (pH 7.4): & gt;100 µg/ml, IC50 (cMet enzyme): 30 nM, IC50 (cMet A549): 800 nM) providing a valid starting point. The co-crystal structure of 1 revealed the binding mode and the essential structural features. The initial HTS hit was bound in a DFG-in conformation interacting with the main chain nitrogen atom of Met1160 within the hinge region and with the main chain nitrogen of Asp1222. After subsequent optimization of potency, efficacy, PK properties and the safety profile of thiadiazinone 1, EMD1204831 was identified as development compound. EMD1204831 is currently being investigated in a phase 1 clinical trial. The pyridazinone EMD1204831 inhibits enzymatic and cellular c-Met kinase activity with IC50 values of 12 nM and 15 nM, respectively. EMD1204831 displayed an exquisite selectivity when tested in vitro against a panel of more than 400 potential off-targets, including kinases, GPCRs, ion channels, transporters and various enzymes. EMD 1204831 demonstrated excellent anti-tumor activity in vivo in a variety of xenograft models, including U87-MG glioblastoma cells (autocrine HGF expression), TPR-Met-transformed mouse fibroblasts (oncogenic Met fusion protein) or Hs746T gastric cancer cells (c-Met gene amplification and HGF-independent activation). Depending on the particular model, complete regressions were observed with doses as low as 6 mg/kg/d administered per os. PK/PD analysis revealed efficient, dose- and time-dependent inhibition of c-Met phosphorylation, reduction of IL-8 and cyclin D1 expression as well as an induction of the cell cycle inhibitor p27. The overall profile of EMD1204831 including first time structural disclosure, some structure activity relationships, in vitro potency, selectivity profile and in-vivo data will be presented. 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 2786. doi:10.1158/1538-7445.AM2011-2786
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 15, No. 17 ( 2009-09-01), p. 5445-5456
    Abstract: Purpose: The insulin-like growth factor (IGF) signaling axis is frequently dysregulated in hepatocellular carcinoma (HCC). Therefore, we investigated whether the specific targeting of the IGF type 1 receptor (IGF-1R) might represent a new therapeutic approach for this tumor. Experimental Design: Total and phosphorylated levels of IGF-1R were measured in 21 paired samples of human HCCs and adjacent nontumoral livers using ELISA. The antineoplastic potency of a novel anti–IGF-1R antibody, AVE1642, was examined in five human hepatoma cell lines. Results: Overexpression of IGF-1R was detected in 33% of HCCs and increased activation of IGF-1R was observed in 52% of tumors. AVE1642 alone had moderate inhibitory effects on cell viability. However, its combination with gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, induced supra-additive effects in all cell lines that were associated with cell cycle blockage and inhibition of AKT phosphorylation. The combination of AVE1642 with rapamycin also induced a synergistic reduction of viability and of AKT phosphorylation. Of marked interest, AVE1642 alone up-regulated the phosphorylated and total levels of HER3, the main partner of EGFR, and AVE1642-induced phosphorylation of HER3 was prevented by gefitinib. Moreover, the down-regulation of HER3 expression with siRNA reduced AKT phosphorylation and increased cell sensitivity to AVE1642. Conclusions: These findings indicate that hepatoma cells overcome IGF-1R inhibition through HER3 activation in an EGFR-dependent mechanism, and that HER3 represents a critical mediator in acquired resistance to anti-IGF-1R therapy. These results provide a strong rational for targeting simultaneously EGFR and IGF-1R in clinical trials for HCC]. (Clin Cancer Res 2009;15(17):5445–56)
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4706-4706
    Abstract: Background: In HCC, acquired resistance to receptor tyrosine kinase receptors has been associated with an increased expression and activation of the c-MET pathway. In addition, initial data suggest that carbonic anhydrase 9 (CA9), a hypoxia marker, is associated with HCC development. The aim of this study was to correlate c-MET and CA9 expression levels by immunochemistry with clinicopathologic characteristics and disease-free survival (DFS) in patients with HCC. Materials and Methods: One-hundred HCC resection specimens were evaluated by immunohistochemistry. Staining results were qualitatively and quantitatively assessed and correlated with clinicopathologic parameters. We elaborated our own dedicated macro, using ImageJ software, for automated evaluation. c-MET high and CA9 high expression were defined as moderate to strong staining. Univariate analyses were performed using Fisher's exact or chi square test, and multivariate analyses using Cox regression model. Median DFS were calculated by Kaplan-Meier method. Results: Patients were mainly males (84%). Tumors were classified as BCLC A1 (92%), uninodular (53%), low-AFP-expressing (82%), moderately differentiated (63%), and with pathologic vascular invasion (71%). c-MET high expression was observed in 51% of tumors and was higher in patients with viral hepatitis-associated HCC (p=0.02) and in patients with AFP & gt;400UI/L (p=0.03). CA9 high expression was observed in 41% of tumors and was correlated with viral hepatitis (p=0.002), pathological vascular invasion (p=0.007), and poor differentiation (p=0.007). c-MET and CA9 expression levels significantly correlated with each other (p=0.008). Median DFS was shorter in the c-MET high (12.9 months, vs & gt;80 months in c-MET low patients; p=0.018) and CA9 high (10.2 months, vs 34.4 months in CA9 low patients; p=0.02) populations. Comparison of c-MET and CA9 status discriminated 3 groups with distinct prognosis. The shortest median DFS was 10.7 months in the c-MET high / CA9 high group, whereas median DFS was not reached in the c-MET low / CA9 low group (p=0.003). The group comprising c-MET high / CA9 low and c-MET low/ CA9 high expressing patients was defined as an intermediate risk populations (DFS=19 months). In a multivariate analysis, smaller tumor size (p=0.025), uninodular morphology (p=0.004), and CA9-low levels (p=0.007) were independently associated with a prolonged DFS. In addition, c-MET high expression level was significantly associated with shorter DFS when CA9 was excluded from the model (p=0.024). Data on c-MET amplification will be added. Conclusion: c-MET expression is a useful prognostic marker in HCC that could be used for patient stratification. Patients with HCC overexpressing c-MET and CA9 represent a subgroup with a particularly poor prognosis who might benefit from therapy with c-MET inhibitors. Citation Format: Annemilai Tijeras-Raballand, Miguel Albuquerque, Cindy Neuzillet, Nathalie Colnot, Friedhelm Bladt, Christian Ihling, Manfred Klevesath, Hongxia Zheng, Eric Raymond, Armand de Gramont, Sandrine Faivre, Valérie Paradis. Biomarker selection defines a subgroup of hepatocellular carcinoma (HCC) patients with poor prognosis who are candidates for MET inhibition strategy. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4706. doi:10.1158/1538-7445.AM2014-4706
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2724-2724
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2724-2724
    Abstract: Introduction: MSC2156119J, a highly selective, potent, reversible, ATP-competitive c-Met inhibitor currently under clinical testing, efficiently inhibits c-Met phosphorylation and downstream signaling in vivo and induces regression of established tumors in xenograft models in preclinical studies (Bladt et al. Clin Cancer Res. 2013;19:2941-51). One initial event following c-Met activation is Y1234/35 phosphorylation in the activation loop of the kinase domain. This results in kinase activation and triggers phosphorylation of tyrosine residues in the c-Met C-terminal tail (eg, Y1349), creating multifunctional docking sites for intracellular adapters. The development of an assay allowing the measurement of these phospho-c-Met epitopes in tumor biopsies will be crucial for establishing an optimal biologic dose for MSC2156119J. Methods: We describe here the development of a highly sensitive Luminex assay that can reliably and reproducibly measure the phosphorylation state of Y1234/35 and Y1349 residues of the c-Met receptor. The high sensitivity of the assay was confirmed by measuring the phospho-c-Met levels in biopsies from patients before and during MSC2156119J treatment. Results: We first assessed the stability of phosphorylated Y1234/35 and Y1349 epitopes in preclinical tumor samples. Analyses revealed that the total c-Met protein is rather stable over time, while the phospho-c-Met epitopes Y1234/35 and Y1349 are relatively unstable; their detection requires rapid processing of tumor samples derived from preclinical tumors or from patients. In tumor samples derived from human xenografts treated with MSC2156119J, Y1349 phosphorylation inhibition varied in different tumor models and did not show strong dose dependence. In contrast, phosphorylation of Y1234/35 residues was effectively inhibited by MSC2156119J in all tumor models tested. Based on these findings, phosphorylation of c-Met Y1234/35 was also assessed in tumor biopsies from patients treated with MSC2156119J in the first-in-man trial (NCT01014936; Falchook et al. J Clin Oncol. 2013;31(suppl):2506). When compared to pretreatment tumor biopsies, on-treatment samples showed effective, dose-dependent inhibition of & gt;90% of c-Met Y1234/35 phosphorylation. Conclusions: We successfully developed an assay capable of detecting phosphorylation of the c-Met Y1234/35 epitope, which is crucial for c-Met activation. Using this assay on tumor samples from xenograft models and on patient-derived tumor biopsies, we demonstrated that MSC2156119J inhibits phosphorylation of c-Met Y1234/35 in a dose-dependent fashion. Therefore, phosphorylation of c-Met Y1234/35 can be used as a pharmacodynamic biomarker of c-Met inhibition and will be an important and valuable element for the selection of the optimal biologic dose of MSC2156119J. Citation Format: Friedhelm Bladt, Frank Jaehrling, Manja Friese-Hamim, Gerald S. Falchook, Hesham M. Amin, Manfred B. Klevesath, Andree Blaukat. Development of a sensitive assay for measuring pharmacodynamic modulation of c-Met in biopsies. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2724. doi:10.1158/1538-7445.AM2014-2724
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 9
    In: Molecular Oncology, Wiley, Vol. 9, No. 7 ( 2015-08), p. 1434-1446
    Abstract: RAS oncogenic variants counteract MET receptor tyrosine kinase inhibition. MET‐driven tumors harboring mutated RAS display resistance to MET inhibition. MAPK‐activating RAS mutations differentially interfere with MET‐mediated biological effects of MET inhibition. The effects of RAS mutants can be reversed when MET inhibition is combined with MEK inhibitors.
    Type of Medium: Online Resource
    ISSN: 1574-7891 , 1878-0261
    Language: English
    Publisher: Wiley
    Publication Date: 2015
    detail.hit.zdb_id: 2322586-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. CT236-CT236
    Abstract: Introduction: Overexpression of the c-Met oncoprotein can enhance invasive tumor behavior and is correlated with poor prognosis. MSC2156119J, a highly selective, ATP-competitive, reversible c-Met inhibitor, suppresses tumor growth and induces regression of HGF-dependent and -independent tumors in preclinical models. Methods: This dose-escalation phase I trial (3+3 design) included patients with advanced solid tumors (NCT01014936). The primary endpoint was to assess the maximum tolerated dose (MTD). Secondary endpoints included safety, antitumor activity, pharmacokinetics (PK), and pharmacodynamics (Pd). Patients were treated once a day with oral MSC2156119J in 21-day cycles: days 1-14 followed by 7 days rest (regimen [R] 1), 3 times per week (R2), or continuously day 1-21 (R3). An optimized formulation was introduced in Aug 2011. Results: Up to Sep 30, 2013, data were available for 125 patients (R1=42; R2=45; R3=38). Doses were escalated from 30-230 mg/day in R1 and 30-115 mg/day in R2 with the initial formulation, and from 30-400 mg/day in R1, 60-315 mg/day in R2, and 300-1400 mg/day in R3 with the optimized formulation. Bioavailability was higher with the optimized formulation; AUC and Cmax increased with dose. Dose-limiting toxicities (DLTs) were reported in 6 patients: asymptomatic G4 lipase and G3 amylase increase (R1; 115 mg/day), asymptomatic G3 lipase increase (R2; 60 and 100 mg/day optimized formulation), G3 nausea and vomiting (R2; 130 mg/day optimized formulation), G3 ALT elevation (R3; 1000 mg/day optimized formulation), and G3 fatigue (R3; 1400 mg/day optimized formulation). Additional ≥G3 drug-related adverse events (AEs) included G3 peripheral edema (1 patient; R3; 300 mg/day optimized formulation), and G3 AST elevation (1 patient; R3; 1000 mg/day optimized formulation). Most frequent G2 drug-related AEs (R1-3) included fatigue (n=8), peripheral edema (n=3), vomiting (n=3), nausea (n=2), asymptomatic lipase elevation (n=2), and neutropenia (n=2). Most patients (80%) had no drug-related AE & gt;G1. In 19/21 evaluable patients, paired tumor biopsies (pre- and on-therapy) revealed phospho-c-Met inhibition. One patient (esophageal adenocarcinoma) achieved confirmed partial response (PR); 2 patients (nasopharyngeal and colorectal carcinoma) had unconfirmed PRs. Eighteen patients had stable disease ≥4 mo, including 1 patient with stable disease & gt;32 mo. Based on preclinical PK/Pd models and clinical Pd data, a dose of 500 mg was considered biologically active and sufficient to achieve target inhibition of ≥95%. Conclusions: MSC2156119J showed antitumor activity and was well tolerated. An MTD has not yet been reached at the doses explored. 500 mg once/day was defined as the recommended phase II dose. Additional Pd and biomarker data will be presented, including c-Met IHC from baseline and on-treatment biopsies, and baseline Met FISH. Citation Format: Gerald S. Falchook, David S. Hong, Hesham M. Amin, Siqing Fu, Sarina Anne Piha-Paul, Filip Janku, J. Gabrielle Granda, HongXia Zheng, Manfred B. Klevesath, Karola Köhler, Friedhelm Bladt, Andreas Johne, Razelle Kurzrock. MSC2156119J (EMD 1214063), an oral selective c-Met inhibitor, in patients with advanced solid tumors: results of the first-in-human phase I trial. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr CT236. doi:10.1158/1538-7445.AM2014-CT236
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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