Skip to main content
Erschienen in: BMC Cancer 1/2016

Open Access 01.12.2016 | Research article

Anti-c-Met monoclonal antibody ABT-700 breaks oncogene addiction in tumors with MET amplification

verfasst von: Jieyi Wang, Liliane Goetsch, Lora Tucker, Qian Zhang, Alexandra Gonzalez, Kedar S. Vaidya, Anatol Oleksijew, Erwin Boghaert, Minghao Song, Irina Sokolova, Ekaterina Pestova, Mark Anderson, William N. Pappano, Peter Ansell, Anahita Bhathena, Louie Naumovski, Nathalie Corvaia, Edward B. Reilly

Erschienen in: BMC Cancer | Ausgabe 1/2016

Abstract

Background

c-Met is the receptor tyrosine kinase for hepatocyte growth factor (HGF) encoded by the MET proto-oncogene. Aberrant activation of c-Met resulting from MET amplification and c-Met overexpression is associated with poor clinical outcome in multiple malignancies underscoring the importance of c-Met signaling in cancer progression. Several c-Met inhibitors have advanced to the clinic; however, the development of inhibitory c-Met-directed therapeutic antibodies has been hampered by inherent agonistic activity.

Method

We generated and tested a bivalent anti-c-Met monoclonal antibody ABT-700 in vitro for binding potency and antagonistic activity and in vivo for antitumor efficacy in human tumor xenografts. Human cancer cell lines and gastric cancer tissue microarrays were examined for MET amplification by fluorescence in situ hybridization (FISH).

Results

ABT-700 exhibits a distinctive ability to block both HGF-independent constitutive c-Met signaling and HGF-dependent activation of c-Met. Cancer cells addicted to the constitutively activated c-Met signaling driven by MET amplification undergo apoptosis upon exposure to ABT-700. ABT-700 induces tumor regression and tumor growth delay in preclinical tumor models of gastric and lung cancers harboring amplified MET. ABT-700 in combination with chemotherapeutics also shows additive antitumor effect. Amplification of MET in human cancer tissues can be identified by FISH.

Conclusions

The preclinical attributes of ABT-700 in blocking c-Met signaling, inducing apoptosis and suppressing tumor growth in cancers with amplified MET provide rationale for examining its potential clinical utility for the treatment of cancers harboring MET amplification.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12885-016-2138-z) contains supplementary material, which is available to authorized users.

Competing interests

JW, LT, QZ, KV, AO, EB, MA, WP, PA, AB, LN and ER are employees of AbbVie. LG, AG and NC are employees of Pierre Fabre. MS, IS and EP are employees of Abbott. The design, study conduct, and financial support for this research were provided by AbbVie, Pierre Fabre & Abbott. AbbVie, Pierre Fabre and Abbott participated in the interpretation of data, review, and approval of the manuscript.

Authors’ contributions

Conception and design: JW, LG, ER. Development of methodology: JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, MA, WP, PA. Acquisition of data (provided animals acquired and managed patients, provided facilities, etc.): LT, QZ, AG, KSV, AO, MS, IS, MA, WP, PA. Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, EP, MA, WP, PA, AB, LN, NC and ER. Writing, review, and/or revision of the manuscript: JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, EP, MA, WP, PA, AB, LN, NC and ER. Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, EP, MA, WP, PA. Study supervision: JW, LG, EB, IS, EP, PA, AB, LN, NC and ER. All authors have read and approved the final version of the manuscript.
Abkürzungen
ABT-700
humanized monoclonal antibody against c-Met
5D5
mouse hybridoma antibody against c-Met
MetMAb
one-armed antibody derived from 5D5
FISH
fluorescence in situ hybridization

Background

Amplification of the MET gene, with consequent c-Met receptor tyrosine kinase (RTK) overexpression and constitutive kinase activation, is an oncogenic driver in multiple malignancies [14]. Unlike other oncogene RTKs including the ERBB family members which have been clinically targeted with therapeutic antibodies, the development of inhibitory c-Met-directed therapeutic antibodies has been challenging [3, 57]. Binding of c-Met by HGF or overexpression of c-Met on cell surface independent of ligand induces dimerization and activation of the receptor tyrosine kinase [2, 8]. Previously reported bivalent antibodies generated against c-Met often mimic HGF, promoting productive dimerization and activation of c-Met [9, 10]. The engineered monovalent antibody, MetMAb (onartuzumab), avoids this agonistic activity [11] but the monovalent nature of MetMAb may limit the scope of its activity to HGF-dependent c-Met signaling, similar to the HGF-binding antibodies [6].
ABT-700 is a bivalent humanized IgG1 that displays distinctive properties compared to other c-Met-targeting antibodies. ABT-700 binds cellular c-Met and disrupts its productive dimerization and activation induced by HGF or by the high density of c-Met on the cell surface independent of ligand. We hypothesize that ABT-700 might be effective in treating cancers harboring amplified MET and focused preclinical studies to assess its antitumor activity in models driven by MET amplification. These findings provide scientific rationale for the clinical activity observed in patients with MET amplified tumors following treatment with ABT-700.

Methods

Antibodies, reagents and cell culture

ABT-700, an anti-human c-Met antibody derived from the mAb 224G11 [12] was produced in a stable CHO line. Fab and F(ab)’2 of mAb224G11 (ABT-700) were generated by digestion with papain or pepsin as described in the literature [13]. Control human IgG was purchased from Sigma (I4506). 5D5 mouse anti-human c-Met antibody, the parental bivalent antibody from which the single armed antibody onartuzumab was derived, was purified from hybridoma supernatant (ATCC #HB11895). The anti-c-Met antibody, LY2875358, was expressed in and purified from HEK293 cells using amino acid sequences derived from published patent application US201012936. The c-Met tyrosine kinase inhibitor, PF-4217903, was purchased from Selleck (Catalog No.S1094). Recombinant human c-Met extracellular domain with a histidine tag (rh-c-Met ECD-6His) was expressed in and purified from HEK293 cells. HGF was purchased from R&D (rhHGF, #294-HGN/CF). The tumor cell lines A549 (ATCC #CCL-185), EBC1 (JCRB #0820), Hs746T (ATCC #HTB-135), and OE33 (Sigma #96070808) were maintained in DMEM (Gibco-Invitrogen cat. No. 11995) supplemented with 10 % fetal bovine serum (FBS) (HyClone SH30070.03). IM95 (JCRB #1075) were also maintained in DMEM, 10 % FBS with 10 mg/L insulin. SNU5 (ATCC #CRL-5973), NCI-H441 (ATCC #HTB-174), NCI-H1993 (ATCC #CRL-5909), MKN45 (JCRB 0245), SNU620 (KCLB #00620), and SNU638 (KCLB #00638) were cultured in RPMI-1640 (Gibco-Invitrogen, cat. No. 11875) supplemented with 10% FBS. MCF7 cells (ATCC HTB-22) were infected with control lentivirus or lentivirus containing human c-Met cDNA in pLVX-IRES-puro vector (Clontech). Stable clones overexpressing human c-Met protein indicated by Western Blot and FACS were isolated. These cells were grown in DMEM (Gibco-Invitrogen cat. No. 11995) supplemented with 10 % fetal bovine serum (FBS) (HyClone SH30070.03) and 2 μg/mL puromycin (Sigma). All cell lines were expanded in culture upon receipt and cryopreserved to provide cells at similar stage passages for all subsequent experiments. For cell lines not authenticated in the 6 months before use, c-Met expression was confirmed by FACS analysis. Information of additional cell lines is summarized in Additional file 1: Table S1.

Binding ELISA

96-well plates (Costar #3369) were coated with 100 μL/well of mouse anti-His antibody (Invitrogen #37-2900) at 1 μg/mL in PBS pH7.4 at 4 °C overnight, and then blocked using Superblock (Pierce, #37535) for one hour at room temperature. Plates were washed 4 times with PBST and then incubated with 100 μL of recombinant human c-Met extracellular domain (rh-c-Met ECD-6His) at 2 μg/mL in 10 % Superblock in PBST for 1 h at room temperature. Plates were washed 4 times with PBST and then incubated with ABT-700 or control human IgG in serial dilutions in 10 % Superblock in triplicate wells at room temperature for 1 h. Plates were washed 4 times with PBST and then incubated with 100 μL of 1:15,000 goat anti-human IgG-HRP (Thermo-scientific Pierce, Cat#31412) at room temperature for 1 h. Plates were washed 4 times in PBST and 100 μL of TMB (Pierce, #34028) was added to each well and incubated at room temperature until color developed (approximately 10 min). Reactions were stopped by addition of 2N sulfuric acid (Mallinckrodt chemicals, Cat#H381-05) and optical density (OD) was read at 450 nm.

FACS analysis

For cellular c-Met binding studies, cells were harvested from flasks when approximately 80 % confluent using Cell Dissociation Buffer (Invitrogen #13151-014 or #13150-016). Cell viability was checked by trypan blue staining to ensure >90 % live cells. Cells were washed once in PBS/1 % FBS (FACS buffer) then resuspended at 1.5-2.5 × 106 cells/mL in FACS buffer. Cells were added to a round bottom 96-well plate (BD Falcon #3910) at 100 μL/well. Ten μL of a 10x concentration of ABT-700 or controls in duplicate wells was added and plates were incubated at 4 °C for four hours. Wells were washed twice with FACS buffer then resuspended in 50 μL of 1:500 anti-human IgG Ab (AlexaFluor 488, Invitrogen #11013) diluted in FACS buffer. Plates were incubated at 4°C for one hour then washed twice with FACS buffer. Cells were then resuspended in 100 μL of PBS/1 % formaldehyde and analyzed on a Becton Dickinson LSRII flow cytometer. FACS binding studies were performed for each cell line in at least two independent experiments.
For Annexin V apoptosis detection, tumor cells were plated at 300,000 cells/ well in 12-well dishes in 2 ml serum-free media (RPMI, 0.1 % BSA). Cells were incubated overnight at 37 °C, 5 % CO2. Cells were treated with control hIgG (Sigma I4506) and ABT-700 at 10 μg/ml for 24 h. Cells were transferred from 12-well plate into 1.5 ml microcentrifuge tubes, pelleted, and washed with cold PBS. Cells were resuspended in 0.1 ml 1X Binding Buffer provided in kit (BD Pharmingen kit cat# 556547). 5 μl of FITC Annexin V and 5 μl propidium iodide (PI) were added and cells were incubated in the dark for 15 min. 400 μl of 1X Binding Buffer was added and cells were analyzed on a Becton Dickinson LSRII flow cytometer within one hour.

Determination of cellular c-Met phosphorylation and total level

A549 cells were plated at 40,000 per well in 96-well plate in growth media. Twenty four hours later, cells were pretreated with antibodies in duplicate wells for one hour at 37 °C, and then stimulated with HGF for 10 min at 37 °C. 1 nM (~100 ng/mL) HGF was used to stimulate c-Met as described in the literature [11]. For SNU5 cells that have constitutively phosphorylated c-Met, the cells were plated at 20,000 per well in 96-well V-bottom plate in serum free medium. Twenty four hours later, cells were treated with antibodies in duplicate wells for six hours at 37 °C. Media were then removed and cells were lysed with 100 (for A549) or 150 (for SNU5) μL/well of Cell Lysis Buffer (Cell Signaling Technology #9803) supplemented with protease inhibitor tablet (Roche #11714900). ELISA capture plates were generated by pre-coating wells with 100 μL of an anti-c-Met antibody (R&D systems, # MAB3581) at 2 μg/mL) at 4 °C overnight, followed by blocking with 200 μL/well PBS/1 % BSA treatment for one hour at room temperature, and washed three times in PBST. Cell lysates were added to capture plates and incubated at 4°C overnight. Plates were washed 3 times in PBST, and incubated with anti-phospho-tyrosine 4G10-HRP conjugate (Millipore #16-105; 1:1000 diluted in PBST + 1 %BSA) for 2 h at room temperature. To determine total c-Met, secondary anti-c-Met HRP conjugate was used. Plates were washed 3 times in PBST and 100 μL of TMB was added to each well and incubated at room temperature until color developed. Reactions were stopped by addition of 100 μL/well 2N sulfuric acid, and the OD was read at 450 nm. These studies were performed for each cell line in at least two independent experiments.

Western blot analysis

Cells were plated at 300,000 per well in 12-well tissue culture plates and were incubated overnight in growth media. Cells were incubated with ABT-700 or control for the time points as indicated at 37 °C. Cells were then lysed with 100 μL/well of 2X LDS NuPAGE sample buffer (Invitrogen NP0007) with reducing reagent (Invitrogen NP0009). Cell lysates were resolved by SDS-PAGE using 4-12 % Bis-Tris NuPAGE gels (Invitrogen NP0322) and transferred to PVDF membranes (Millipore Immobilon-FL # IPFL07810). Blots were blocked with Odyssey Blocking Buffer (LI-COR # 927-40000) for one hour at room temperature, washed three times with PBST, and then incubated overnight with appropriate primary antibodies at 4 °C. Following overnight incubation with primary antibodies, blots were washed three times with PBST for ten minutes, and then incubated with either AlexaFluor680 goat anti-rabbit IgG (Invitrogen A21109, 1:10000) or goat anti-mouse IRDye 800CW (Odyssey # 926–32210, 1:5000) for one hour at room temperature. Blots were then washed three times with PBST, and visualized by scanning using an LI-COR Instrument Odyssey (Model # 9120). Primary antibodies included mouse anti-c-Met (Cell Signaling # 3148 or # 3127), rabbit anti-phospho-Y1234 c-Met (Cell Signaling Technology, # 3077), phosphor-PLCr (Cell Signaling #2821) , phosphor-Erk (Invitrogen #44680G), phosphor-Bad (Cell Signaling #9291), total bad (Cell Signaling #9292), Bim (Cell Signaling #2819), Bcl-xL (BD #51-6646GR), cytochrome C (BD Pharmingen Cat 556433), cleaved PARP (Epitomic #1074-1), and mouse anti-actin (Sigma, # A5441). At least two independent experiments were carried out for each cell line.

Cytochrome C release assay

SNU5 cells (4×106) were treated with control hIgG or ABT-700 for 24 h. Cells were washed in PBS, pelleted and resuspended in 50–100 μL Digitonin Lysis buffer (75 mM NaCl, 8 mM Na2HPO4, 1 mM NaH2PO4, 1 mM EDTA, 350 μg/mL digitonin, and 250 mM sucrose) by pipetting up and down, and incubated for 30 s. Cells were pelleted at high speed in microcentrifuge for 1 min, supernatant (cytosolic fraction) was collected and 2x NuPAGE sample buffer was added for Western analysis. Pellets (organelle-containing membrane fraction) were washed in cold PBS 3 times, and lysed by sonication in 1x NuPAGE sample buffer for Western analysis. The study was performed in two independent experiments.

Proliferation assay

Tumor cells were plated in 96-well plate (Falcon 35–3075) in 180 μL growth media at 3000–5,000 cells/well. The cells were incubated overnight at 37 °C with 5 % CO2. On Day 2, dilutions of testing articles were added to the cell plate (20 μL/well) in triplicate wells. Untreated control wells (for 0 % control) and wells treated with 10 μM staurosporin (for 100 % kill) were included in each plate. The plates were incubated for 3–5 days at 37 °C with 5 % CO2. To quantify live cells, media was removed and 1x Cell Titer Aqueous One Solution (Promega, G3581) diluted in Opti-mem media (Invitrogen # 31985–070) was added to plates and the plates were then incubated for one hour at 37 °C. The OD at 490 nm was read on a M5 Spectramax plate reader (Molecular Probes). Percent inhibition was calculated based on 100 % kill and untreated control wells using the following formula: 100x (0 % control - treated)/(0 % control - 100 % kill). For cells grown in suspension such as SNU5, cells were plated in 96-well plate in 180 μL medium at 10,000 cells/well and incubated overnight at 37 °C with 5 % CO2. The same protocol as above was used for treatment and data processing except live cells were detected by adding forty μL of Cell Titer Aqueous One Solution to each well and the plates were then incubated for one hour at 37 °C. The experiments were repeated at least twice for each cell line.

Anti-tumor efficacy studies in vivo

Studies with gastric (SNU5 and SNU620), lung (EBC1, NCI-H441) and glioblastoma (U87MG) were carried out at Pierre Fabre. Animal Ethical Committee was registered under the CEA-CIPF-108 number. All experiments conformed to the United Kingdom Co-ordinating Committee on Cancer Research (UKCCCR) Guidelines for the Welfare of Animals in Experimental Neoplasia UKCCR for animal care and use. SCID mice were from Charles River Laboratories (L’Arbresle, France). Athymic Nude Mice were acquired from Harlan (Gannat, France). All animals were housed on a 12 h light/dark cycle, in sterilized filter-topped cages, in a temperature 22 +/−2 °C and in humidity (30 to 70 %) controlled room. Mice were maintained in sterile conditions with food and water provided ad libitum and manipulated according to French and European guidelines. Animals were examined before the initiation of experiments to ensure that they were healthy and acclimated to the laboratory environment.
Cells (5–10 × 106) were implanted s.c. into the right flank region of mice. Tumor bearing mice were size matched and randomized into study groups (n = 5 or 6 as shown in figure legends). Each experiment consisted of an ABT-700 dose evaluation injected i.p. compared to controls. Evaluation of the anti-tumor activity was determined by measuring tumor volume twice a week using the formula: π/6 × length × width × height.
For the ectopic EBC1 lung metastasis and survival model, mice were injected s.c. with 7x106 EBC1 cells on D0, and after 5 days, when tumor volume reached 60 mm3 to 80 mm3, mice were size matched and randomized into groups (n = 7) for treatment with ABT-700 or control administered by i.p. injections every 21 days. From D5 to D21, tumor volume was monitored twice a week with an electronic caliper. On day 21, subcutaneous primary tumors were resected from mice anesthetized with a Ketamine/Xylazine mixture (70/30) injected intra-muscularly. ABT-700 antitumor activity was monitored by following animal mortality.
Additional animal groups were introduced for the gastric xenograft model SNU5 in order to evaluate pharmacodynamic markers by immunohistochemistry as described in detail in Additional file 2: Supplementary methods.
Experiments with the gastric Hs746T s.c. xenograft model were conducted at AbbVie in compliance with AbbVie’s Institutional Animal Care and Use Committee and the National Institutes of Health Guide for Care and Use of Laboratory Animals guidelines in a facility accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care. SCID mice were obtained from Charles River (Wilmington, MA). Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments.
Hs746T cells (2 × 106) were inoculated in the flanks of male SCID mice and tumor-bearing animals were size matched and randomly assigned to cohorts to a mean tumor volume of approximately 225 mm3 per group (N = 10) 15 days post inoculation of cells. Dosing for all agents was initiated on day 16. ABT-700 (10 mg/kg) was administered twice a week i.p. while docetaxel (7.5 mg/kg) was administered i.v. as a single dose. A human IgG control antibody was used as a negative control agent. Tumor dimensions were determined twice weekly and volume determined with the formula (L x W2)/2.

Fluorescence in situ hybridization (FISH) analysis

MET gene copy numbers in cell lines, tumor xenografts and human tumor tissue specimens was detected with a probe mix (Vysis LSI MET SpectrumOrange/Vysis CEP 7 SpectrumGreen, Abbott Molecular) using protocols described in detail in Additional file 2: Supplementary methods.

Statistical analysis

Results are expressed as the mean ± SEM. All data were analyzed with GraphPad Prism V6.05 (GraphPad Software, Inc., San Diego, CA). The difference in tumor growth between different groups was analyzed by two-way ANOVA Turkey’s multiple comparison tests. The survival data were analyzed using Log-rank (Mantel-Cox) test. A P value <0.05 was considered significant.

Results

Characterization of ABT-700 binding properties

ABT-700 is a humanized bivalent IgG1/kappa monoclonal antibody derived from the m224G11 mouse hybridoma [12]. ELISA and FACS-based assays were used to characterize the binding properties of ABT-700. In an ELISA format, ABT-700 binds recombinant human c-Met with an apparent affinity of 0.22 nM (Table 1). Similar binding to cynomolgus monkey derived c-Met was also observed although there was no detectable binding to mouse derived c-Met (not shown). These binding properties are similar to those observed for the parental mAb m224G11 [12].
Table 1
Binding affinity of ABT-700
 
ABT-700 (nM)
Biding to c-Met protein by ELISAa
0.22
Binding to cellular c-Met by FACSb
 
 A549
0.23
 IM95
0.24
 SNU5
0.55
 EBC1
0.24
 MCF7
No binding
 MCF7-human c-Met
1.3
aEC50 values were derived from an ELISA in which c-Met ECD was captured on the plate via a histidine tag. Value shown is averages of 6 experiments
bEC50 values were derived from FACS analysis of ABT-700 on various cell lines. Values are representative of at least two experiments
FACS analysis was used to compare the binding of ABT-700 to human tumor cell lines that express c-Met or harbor MET amplification. ABT-700 shows monophasic and saturable binding to cell lines expressing endogenous c-Met including A549, IM95, SNU5 and EBC1 with EC50 values of 0.2 – 1.3 nM (Table 1). There was no ABT-700 binding to the c-Met negative MCF7 breast cancer cell line, however following transfection to introduce endogenous c-Met expression into this cell line, significant ABT-700 binding was observed (Fig. 1a and Table 1). These results indicate specific and high affinity binding of ABT-700 to human c-Met.

ABT-700 antagonizes c-Met signaling in both HGF-dependent and -independent settings

To determine the effect of ABT-700 on cellular c-Met phosphorylation induced by exogenous ligand HGF (1 nM) and/or by over-expressed c-Met in the absence of added HGF, we used ELISA-based assays to quantify phospho-c-Met in cell lysates of A549 and SNU5. ABT-700 and parental mAb m224G11 blocked the HGF-induced c-Met phosphorylation of A549 tumor cell line with an IC50 of 1.0 nM (Fig. 1b). 5D5, the parent murine monoclonal antibody of MetMAb [11], was also effective in blocking HGF-induced c-Met phosphorylation (Fig. 1b) although in the absence of HGF it can induce c-Met phosphorylation and agonistic activity (see section below). In contrast to the HGF-dependent A549 cell line, SNU5 gastric cancer cells express high levels of constitutively phosphorylated c-Met. ABT-700 treatment, but not 5D5 treatment (data not shown), caused more than 80% reduction of phosphorylated c-Met in SNU5 cells with an IC50 of 0.4 nM (Fig. 1c). ABT-700 also caused a decrease of total cellular c-Met by ~40 % in this cell line (Fig. 1d). Given that the decrease of c-Met phosphorylation was greater than that of c-Met protein, inhibition of phosphorylation and down-regulation of receptor may be caused through distinct mechanisms by ABT-700. The antagonistic activity of ABT-700 required the bivalent nature of the antibody because the Fab fragment did not inhibit c-Met phosphorylation or induce c-Met down-regulation (Fig. 1c and d).
Activation of c-Met tyrosine kinase induces phosphorylation of receptor adaptor proteins such as Gab1, triggering Akt and Erk signaling cascades [2, 3, 14]. Treatment with the agonistic c-Met antibody 5D5 mimics activity of the native ligand HGF that induces elevating levels of phosphorylated Gab1, Akt and Erk in both the HGF ligand-dependent U87MG and the MET constitutively activated Hs746T cells after treatment (Fig. 1e). In contrast, ABT-700 and its parental mouse hybridoma antibody m224G11 antagonize cellular c-Met signaling without causing agonist activity as evidenced by a decrease in phosphorylated signaling molecules including Gab1, Akt and Erk in both cell lines (Fig. 1e).
ABT-700 activity differs from the recently reported anti-c-Met antibody LY2875358 [15] in tumor cells where c-Met signaling can be driven by both receptor expression and HGF stimulation. SNU620 gastric cancer cells have high basal level of phosphorylated c-Met as shown in Fig. 1f, and their growth in vitro is stimulated by the agonistic anti-c-Met antibodies 5D5 (Fig. 1f). Similar effects on cell proliferation were also observed after treatment with HGF (data not shown). ABT-700 showed more robust inhibition of c-Met signaling and proliferation in SNU620 cells than LY2875358 although the latter was similarly effective (not shown) in other cellular models examined for ABT-700 as shown in Fig. 1. This difference was seen also in efficacy trials in vivo where ABT-700 but not LY2875358 inhibited the SNU620 tumor growth (described in sections below).
Taken together, these data suggest that ABT-700 is a differentiated anti-c-Met antibody with strong antagonistic effect on both HGF-independent constitutive c-Met signaling and HGF-dependent activation of c-Met.

ABT-700 induces apoptosis in cell lines harboring MET amplification

To identify tumor lines with aberrant c-Met signaling suitable for further examination of the effect of ABT-700, we evaluated a panel of 35 human cancer cell lines for MET amplification, c-Met protein expression and their dependence on c-Met activity for growth in vitro. Most of the cell lines evaluated were derived from gastric cancer because of the high incidence of MET amplification associated with that cancer type [16, 17]. As summarized in Table 2 and Fig. 2a, we identified eight tumor cell lines exhibiting MET gene amplification as determined by fluorescent in situ hybridization (FISH) with an average ratio of MET and CEP 7 (a centromere control) copy numbers of 2 or greater (Table 2 and Additional file 3: Figure S1). All cell lines harboring MET amplification except for NCI-H1573 showed dependence on c-Met signaling for growth in vitro (Table 2). NCI-H1573 cells do not have high level of c-Met protein despite possessing low level amplification of the MET gene (Table 2). On the other hand, there are cell lines that do not have MET amplification but show c-Met signaling dependence likely due to the presence of autocrine HGF (e.g. IM95) and/or c-Met overexpression (e.g. SNU638 and NCI-H820).
Table 2
Summary of MET status and sensitivity to ABT-700 in a panel of 35 human cancer lines
Cell line
Type
Source
FISH
WB
Proliferation
 
   
MET (mean/cell)
CEP7 (mean/cell)
Ratio MET/CEP7
Total c-Meta
Phospho-(Y1234) c-Meta
Max Inhibition by (%)
        
PF-4217903b
ABT-700c
SNU620
gastric
KCLB
41.6
4.4
10.6
6.7
56
95
80
H1993
lung
ATCC
32
11.9
2.8
5.4
829
50
30
Hs746T
gastric
ATCC
22.2
3.9
6.6
6
377
55
40
OE33
esophageal
Sigma
21.4
6.9
3.1
6
430
60
70
SNU5
gastric
ATCC
20.2
7.9
2.6
12
910
95
90
EBC1
lung
JCRB
15.6
3.1
5.2
6
374
85
30
MKN45
gastric
JCRB
12.6
3.4
4
10
297
96
50
H1573
lung
ATCC
11.5
4.3
2.7
1.8
5
0
0
H2342
lung
ATCC
8.6
5.2
1.7
0.7
1.3
0
0
H820
lung
ATCC
8.3
5
1.7
2.9
5.7
20
0
NUGC-2
gastric
JCRB
7.8
8.2
1
0.5
0.1
0
0
FU97
gastric
JCRB
7.6
7.5
1
0.1
0.1
0
0
NUGC-4
gastric
JCRB
6
5.1
1.3
1
15
50
0
SNU-16
gastric
ATCC
5.1
5.7
0.9
0.5
2.5
0
0
KATOIII
gastric
ATCC
4.6
5
0.9
0.9
16
0
0
SNU-216
gastric
KCLB
4.5
4.7
1
0.8
0.5
0
0
MKN-1
gastric
JCRB
4.4
3.8
1.2
0.2
0.1
0
0
SNU-484
gastric
KCLB
4.2
4
1
0.1
0.1
0
0
SNU-668
gastric
KCLB
3.6
3.6
1
1.9
0.5
0
0
SNU-1
gastric
ATCC
3.3
3.2
1
NA
NA
0
0
RERF-GC-1B
gastric
JCRB
3.1
4.2
0.8
0.6
0.6
0
0
OCUM-1
gastric
JCRB
3
2.9
1
0.4
0.3
0
0
SCH
gastric
JCRB
3
3.5
0.9
0.3
0
0
0
NCC-StC-K140
gastric
JCRB
2.8
9
0.3
0.5
0.3
30
0
SNU-719
gastric
KCLB
2.7
3.5
0.8
0.4
0.1
0
0
IM95
gastric
JCRB
2.5
2.5
1.1
0.2
0.5
90
60
MKN74
gastric
JCRB
2.4
2.6
1
0.1
0.1
0
0
SNU-601
gastric
KCLB
2.4
3
0.8
0.4
2.6
0
0
U87MG
glioblastoma
ATCC
2.1
2.1
1
0.5
3
30
30
23132/87
gastric
DSMZ
2
1.9
1.2
0.5
0.2
0
0
AGS
gastric
ATCC
2
2.2
1
0.2
0.1
0
0
NUGC-3
gastric
JCRB
2
3.2
0.7
0.5
1.1
0
0
SNU638
gastric
KCLB
2
2
1
3.6
28
80
50
TAKIGAW
gastric
JCRB
2
5.1
0.4
0.4
0.1
0
0
NCI-N87
gastric
ATCC
1
2
0.5
NA
NA
0
0
A549
lung
ATCC
NA
NA
NA
1
1
0
0
aNormalized to actin and relative A549 which was assigned 1
bat 1 μM concentration that is known to fully inhibit cellular c-Met with minimal off-target effect [21, 28]
cat 10 μg/mL (~67 nM) that saturates c-Met binding
The SNU5 gastric cancer cells harboring MET amplification and constitutively activated c-Met signaling was selected for further analysis to compare ABT-700 and antibody 5D5. The SNU5 cells contained an average of 20 copies of MET with a MET/CEP 7 ratio of 2.6 (Fig. 2b and Table 2) and high levels of total and phosphorylated c-Met protein (Fig. 2c). The IM95 gastric cancer cell line was included for comparative analysis since it exhibits autocrine HGF signaling [18] and dependence on c-Met signaling in the absence of MET amplification (Fig. 2a and Additional file 3: Figure S1). ABT-700, but not 5D5, inhibited the proliferation of the MET amplified SNU5 cells, whereas both ABT-700 and 5D5 inhibited the growth of the HGF-driven IM95 cells (Fig. 2d). These results demonstrate that only ABT-700 blocked both amplification-driven (HGF-independent) and HGF-dependent c-Met activation. The antagonistic activity of ABT-700 against the constitutively activated c-Met requires bivalent binding because the Fab fragment of ABT-700 does not inhibit proliferation (Fig. 2e), consistent with the effects on c-Met phosphorylation observed in these cells (Fig. 1c).
Given that ABT-700 inhibited SNU5 cell growth by more than 90%, we further examined its effect on c-Met signaling pathways and induction of apoptosis in these cells. SNU5 cells possess constitutively activated c-Met as indicated by the high levels of phosphorylated c-Met (Fig. 2c and f). ABT-700 treatment down-regulated c-Met, and abrogated the level of phosphorylated c-Met and downstream signaling molecules including PLCγ, and Erk (Fig. 2f). ABT-700 treatment also activated the intrinsic apoptosis machinery resulting from the imbalance of pro- and anti-apoptotic proteins such as Bad and Bcl-xL. Phosphorylation of Bad leads to its ubiquitylation and degradation [19]. ABT-700 induced a decrease of phosphorylated Bad and the consequent stabilization of this pro-apoptotic protein, which was accompanied by elevation of the pro-apoptotic Bim and a decrease of the anti-apoptotic Bcl-xL (Fig. 2f). Other events consistent with the activation of the intrinsic apoptosis pathway were detected in the ABT-700 treated cells, including the release of cytochrome C into the cell cytoplasm and cleavage of the DNA repair enzyme, poly ADP ribose polymerase (PARP) (Fig. 2f). The increase in Annexin V staining of SNU5 cells following ABT-700 treatment further confirmed the occurrence of apoptosis (Fig. 2g). Furthermore, induction of apoptosis by ABT-700 was seen in other cell lines with amplified MET including SNU620 gastric cancer and EBC1 NSCLC but not in Hs746T and MKN45 gastric cancer cells (data not shown). Collectively, these results demonstrate that ABT-700 can inhibit the proliferation and consequently induce apoptotic cell death in cancer cells addicted to c-Met signaling driven by MET amplification.

ABT-700 inhibits tumor growth in models driven by MET amplification

Next we examined the effects of ABT-700 on tumor growth of subcutaneously implanted human tumor xenografts harboring amplified MET. ABT-700 inhibited the growth of established SNU5 tumors harboring amplified MET at doses ranging from 2.5 to 40 mg/kg, with long-term tumor regressions including complete responses observed at doses of ≥ 10 mg/kg (Fig. 3a). ABT-700 decreased both total and phosphorylated c-Met and the downstream signaling proteins, phosphorylated Akt and Erk, as measured by immunohistochemistry from treated tumor tissue collected on day 7 and/or 21 after a single dose at 10mg/kg (Fig. 3b). Cell proliferation in tumor tissue, indicated by Ki67 staining, was suppressed at both time points (Fig. 3b). These pharmacodynamic responses correlated with activity of inhibition of tumor growth; ABT-700 at doses below 5 mg/kg was not effective in inhibiting the tumor growth or the signaling proteins (Additional file 4: Figure S2). ABT-700 also inhibited the growth of four additional human tumor xenografts harboring MET amplification including EBC1 NSCLC and SNU620, Hs746T and MKN45 gastric cancer (Fig. 3c and 3d; and ref. [12]). Furthermore, ABT-700 was effective in prolonging survival in an EBC1 metastasis model (Fig. 3e). These results indicate that ABT-700 effectively antagonizes constitutively activated c-Met and downstream signaling leading to inhibition of tumor growth in preclinical tumor models with MET amplification.
We also investigated whether ABT-700 in combination with established therapies would provide additional therapeutic benefit in preclinical animal models. In the MET-amplified Hs746T gastric cancer model, the combination of ABT-700 with docetaxel, an anti-mitotic microtubule inhibitor clinically used for gastric cancer, was more efficacious than either agent given as monotherapy (Fig. 4a). Although ABT-700 or docetaxel monotherapy at near maximal tolerated dose inhibited tumor growth in this model, the longer duration of response observed following combination therapy suggests that this combination may be a useful clinical strategy for delaying tumor relapse. A similar outcome was observed in NSCLC models where combination of ABT-700 and standard of care chemotherapeutics such as the nucleoside analog gemcitabine or the anti-mitotic drug vinorelbine (Navelbine) at near maximal tolerated doses. ABT-700 alone effectively suppressed the EBC1 tumor growth but was not able to induce complete responses (Figs. 3c and 4b). Single agent gemcitabine was also only partially effective (Fig. 4b). Combination of these two agents produced tumor regressions and eradications (Fig. 4b). In the NCI-H441 NSCLC xenograft model that is known to have overexpression of c-Met with gene amplification [20] (Fig. 4c), combination of the ABT-700 parental mAb m224G11 with Navelbine also resulted in more complete and durable control of tumor growth. U87MG is an aggressive glioma that possesses autocrine HGF c-Met signaling [21]. mAb m224G11 suppressed U87MG tumor growth demonstrating single agent activity against this HGF-dependent tumor (Fig. 4d). The current SOC therapy for GBM is radiation treatment in combination with the DNA alkylating agent, temozolomide (TMZ), although novel treaments that extend patient survival are urgently needed [22]. Addition of TMZ at near maximal tolerated dose to m224G11 therapy in the U87MG glioma xenograft model resulted in enhanced anti-tumor effects compared to either agent alone (Fig. 4d). Because neither the NCI-H441 nor the U87MG tumor models is MET amplified, these results suggest that ABT-700 in combination with existing therapies may broaden177 the clinical indications beyond those tumors with MET amplification to those tumors with c-Met overexpression, or autocrine HGF stimulation.

MET amplification in gastric cancers and potential clinical utility of ABT-700

MET gene amplification ranging from frequencies of 1-20% has been documented in a variety of human cancers, including liver metastases from colon carcinoma [23], non-small cell lung carcinomas with acquired resistance to EGFR inhibitors [24] and gastric cancers [16, 17]. A MET FISH assay was developed for detecting MET gene amplification in human biopsied cancer tissues utilizing a probe that spans 456 k-bases encompassing the entire MET gene located on the chromosome locus 7q31.2. This MET FISH assay was applied to a tissue microarray of 140 human gastric cancer samples obtained from Asian patients. As anti-HER2 antibody is indicated for the treatment of HER2 positive gastric cancer, we also performed HER2 FISH analysis. MET amplification as defined by MET/CEP7 ratio ≥2 was seen in 10% of 134 evaluable patient samples, and 9 of the 13 MET amplified samples were also positive for HER2 amplification (Fig. 5). This data suggest that patients with HER2 positive gastric cancers should not be excluded from treatment with c-Met inhibitors although further studies are required to confirm the co-existence of amplification of MET and HER2 in this disease setting. Together with earlier studies [16, 17], the data suggest that MET amplification may represent a critical genetic aberration in gastric cancers and this patient population could benefit from treatment with ABT-700.

Discussion

MET amplification is associated with poor prognosis in a variety of cancers highlighting the significant unmet medical need of this patient population. We describe preclinical results that justify the clinical investigation of ABT-700 in cancer types with addiction to the MET oncogene. In a phase 1 clinical trial in patients with advanced solid tumors [25], ABT-700 was well tolerated and the monotherapy at the recommended dose of 15 mg/kg demonstrated anti-tumor activity in patients with MET amplified solid tumors [26]. By RECIST, 3 of 5 patients with tumors harboring MET amplification, as determined by FISH, had a partial response (1 each ovarian, gastric and esophageal). Among these 3 patients, the duration of response was 19, 23, and 24 weeks, respectively [26]. This study has been expanded to enroll additional patients with MET amplification and to better define predictive biomarkers, safety and clinical benefit.
The tumor cell lines evaluated in our study herein that harbor MET gene amplification, as defined by FISH analysis, and overexpress c-Met protein, are sensitive to ABT-700. Cancer cells addicted to constitutively activated c-Met undergo apoptosis upon exposure to ABT-700 monotherapy leading to tumor regression in preclinical animal models. Our data emphasizes the need for a patient selection strategy that harbor MET amplification as they most likely would benefit from ABT-700 treatment. Ultimately, ABT-700 may have broader clinical utility as the prevalence of MET amplified tumors increases with disease progression, recurrence and/or treatment regimens [14]. Thus, acquiring fresh tumor biopsies for FISH analysis may be needed to identify patients with MET amplification. Recently somatic splice site alterations at MET exon 14 (METex14) that result in exon skipping and MET activation were described in human cancers [27]. Although we have not included METex14 analysis in our studies, it will be important to establish the clinical relevance of these genetic aberrations.
In addition to inhibiting tumors with MET gene amplification, ABT-700 also inhibits the subcutaneous xenograft growth of human tumor cell lines that have c-Met protein overexpression or autocrine HGF. Patients with tumors driven by HGF-dependent c-Met activation may benefit from combination of ABT-700 with chemotherapy. In this context, the demonstration of more robust and sustainable anti-tumor activity in animal models following the combination of ABT-700 with different standard of care cytoreductive chemotherapies provides the basis for exploration of effective combination therapy in the clinic.
ABT-700 is among the first reported bivalent anti-c-Met antibodies that lack agonistic activity [12, 15]. By inhibiting both ligand-dependent and c-Met overexpression-induced signaling in broad types of cancer cells, ABT-700 differentiates from other therapeutic agents targeting the HGF/c-Met axis [6, 7, 11] including the recently disclosed antagonistic IgG4 c-Met targeting antibody LY2875358 [15]. ABT-700 binds a unique epitope on c-Met outside the Sema domain which 5D5 recognizes [11, 12] and antagonizes c-Met signaling in a variety of cancer cells including SNU620 gastric cancers where LY2875358 demonstrated no in vivo antitumor activity. Since ABT-700 contains a humanized IgG1 heavy chain, antibody effector functions may also contribute to its antitumor activity. Results from in vitro ADCC functional assays indicate that ABT-700 can mediate the lysis of cellular targets by human natural killer cells (data not shown).

Conclusions

ABT-700 shows preclinical activities in blocking both HGF-dependent and HGF-independent c-Met signaling, inducing apoptosis and suppressing tumor growth in cancers with amplified MET. Collectively, the attributes of ABT-700 coupled with early signs of activity in a phase 1 clinical trial support investigation of this antibody, as monotherapy and in combination, in malignancies characterized by aberrant activation of c-Met signaling including cancers harboring MET amplification.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

JW, LT, QZ, KV, AO, EB, MA, WP, PA, AB, LN and ER are employees of AbbVie. LG, AG and NC are employees of Pierre Fabre. MS, IS and EP are employees of Abbott. The design, study conduct, and financial support for this research were provided by AbbVie, Pierre Fabre & Abbott. AbbVie, Pierre Fabre and Abbott participated in the interpretation of data, review, and approval of the manuscript.

Authors’ contributions

Conception and design: JW, LG, ER. Development of methodology: JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, MA, WP, PA. Acquisition of data (provided animals acquired and managed patients, provided facilities, etc.): LT, QZ, AG, KSV, AO, MS, IS, MA, WP, PA. Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, EP, MA, WP, PA, AB, LN, NC and ER. Writing, review, and/or revision of the manuscript: JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, EP, MA, WP, PA, AB, LN, NC and ER. Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): JW, LG, LT, QZ, AG, KSV, AO, EB, MS, IS, EP, MA, WP, PA. Study supervision: JW, LG, EB, IS, EP, PA, AB, LN, NC and ER. All authors have read and approved the final version of the manuscript.
Literatur
1.
Zurück zum Zitat Yu S, Yu Y, Zhao N, Cui J, Li W, Liu T. C-Met as a prognostic marker in gastric cancer: a systematic review and meta-analysis. PLoS One. 2013;8(11):e79137.PubMedCentralCrossRefPubMed Yu S, Yu Y, Zhao N, Cui J, Li W, Liu T. C-Met as a prognostic marker in gastric cancer: a systematic review and meta-analysis. PLoS One. 2013;8(11):e79137.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Trusolino L, Bertotti A, Comoglio PM. MET signalling: principles and functions in development, organ regeneration and cancer. Nat Rev. 2010;11(12):834–48.CrossRef Trusolino L, Bertotti A, Comoglio PM. MET signalling: principles and functions in development, organ regeneration and cancer. Nat Rev. 2010;11(12):834–48.CrossRef
3.
Zurück zum Zitat Goetsch L, Caussanel V, Corvaia N. Biological significance and targeting of c-Met tyrosine kinase receptor in cancer. Front Biosci (Landmark Ed). 2013;18:454–73.CrossRef Goetsch L, Caussanel V, Corvaia N. Biological significance and targeting of c-Met tyrosine kinase receptor in cancer. Front Biosci (Landmark Ed). 2013;18:454–73.CrossRef
4.
Zurück zum Zitat Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A. 2006;103(7):2316–21.PubMedCentralCrossRefPubMed Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A. 2006;103(7):2316–21.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Knudsen BS, Vande Woude G. Showering c-MET-dependent cancers with drugs. Curr Opin Genet Dev. 2008;18(1):87–96.CrossRefPubMed Knudsen BS, Vande Woude G. Showering c-MET-dependent cancers with drugs. Curr Opin Genet Dev. 2008;18(1):87–96.CrossRefPubMed
6.
Zurück zum Zitat Patnaik A, Weiss GJ, Papadopoulos KP, Hofmeister CC, Tibes R, Tolcher A, et al. Phase I ficlatuzumab monotherapy or with erlotinib for refractory advanced solid tumours and multiple myeloma. Br J Cancer. 2014;111(2):272–80.PubMedCentralCrossRefPubMed Patnaik A, Weiss GJ, Papadopoulos KP, Hofmeister CC, Tibes R, Tolcher A, et al. Phase I ficlatuzumab monotherapy or with erlotinib for refractory advanced solid tumours and multiple myeloma. Br J Cancer. 2014;111(2):272–80.PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Sheridan C. Genentech to salvage anti-MET antibody with subgroup analysis. Nat Biotechnol. 2014;32(5):399–400.CrossRefPubMed Sheridan C. Genentech to salvage anti-MET antibody with subgroup analysis. Nat Biotechnol. 2014;32(5):399–400.CrossRefPubMed
8.
Zurück zum Zitat Tolbert WD, Daugherty-Holtrop J, Gherardi E, Vande Woude G, Xu HE. Structural basis for agonism and antagonism of hepatocyte growth factor. Proc Natl Acad Sci U S A. 2010;107(30):13264–9.PubMedCentralCrossRefPubMed Tolbert WD, Daugherty-Holtrop J, Gherardi E, Vande Woude G, Xu HE. Structural basis for agonism and antagonism of hepatocyte growth factor. Proc Natl Acad Sci U S A. 2010;107(30):13264–9.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Pietronave S, Forte G, Locarno D, Merlin S, Zamperone A, Nicotra G, et al. Agonist monoclonal antibodies against HGF receptor protect cardiac muscle cells from apoptosis. Am J Physiol Heart Circ Physiol. 2010;298(4):H1155–1165.CrossRefPubMed Pietronave S, Forte G, Locarno D, Merlin S, Zamperone A, Nicotra G, et al. Agonist monoclonal antibodies against HGF receptor protect cardiac muscle cells from apoptosis. Am J Physiol Heart Circ Physiol. 2010;298(4):H1155–1165.CrossRefPubMed
10.
Zurück zum Zitat Prat M, Crepaldi T, Pennacchietti S, Bussolino F, Comoglio PM. Agonistic monoclonal antibodies against the Met receptor dissect the biological responses to HGF. J Cell Sci. 1998;111(Pt 2):237–47.PubMed Prat M, Crepaldi T, Pennacchietti S, Bussolino F, Comoglio PM. Agonistic monoclonal antibodies against the Met receptor dissect the biological responses to HGF. J Cell Sci. 1998;111(Pt 2):237–47.PubMed
11.
Zurück zum Zitat Jin H, Yang R, Zheng Z, Romero M, Ross J, Bou-Reslan H, et al. MetMAb, the one-armed 5D5 anti-c-Met antibody, inhibits orthotopic pancreatic tumor growth and improves survival. Cancer Res. 2008;68(11):4360–8.CrossRefPubMed Jin H, Yang R, Zheng Z, Romero M, Ross J, Bou-Reslan H, et al. MetMAb, the one-armed 5D5 anti-c-Met antibody, inhibits orthotopic pancreatic tumor growth and improves survival. Cancer Res. 2008;68(11):4360–8.CrossRefPubMed
12.
Zurück zum Zitat Corvaïa N, Gonzalez A, Boute N, Beau-Larvor C, Fabre-Lafay S, Géronimi F, Broussas Lepecquet AM, Robert A, Wurch T, Haeuw JF, Bailly C, Goetsch L. First bivalent fully antagonist anti-c-Met antibody targeting the c-Met receptor: I) in vitro mechanism of action. Cancer Res 2009, 69. In: Proc Am Assoc Cancer Res; 2009 Apr 2018–2022; Denver, CO. Philadelphia (PA): AACR; 2009. Abstract nr 2835. Corvaïa N, Gonzalez A, Boute N, Beau-Larvor C, Fabre-Lafay S, Géronimi F, Broussas Lepecquet AM, Robert A, Wurch T, Haeuw JF, Bailly C, Goetsch L. First bivalent fully antagonist anti-c-Met antibody targeting the c-Met receptor: I) in vitro mechanism of action. Cancer Res 2009, 69. In: Proc Am Assoc Cancer Res; 2009 Apr 2018–2022; Denver, CO. Philadelphia (PA): AACR; 2009. Abstract nr 2835.
13.
Zurück zum Zitat Porter RR. Lecture for the Nobel Prize for physiology or medicine 1972: Structural studies of immunoglobulins. Scand J Immunol. 1991;34(4):381–9.CrossRefPubMed Porter RR. Lecture for the Nobel Prize for physiology or medicine 1972: Structural studies of immunoglobulins. Scand J Immunol. 1991;34(4):381–9.CrossRefPubMed
14.
Zurück zum Zitat Sheth PR, Hays JL, Elferink LA, Watowich SJ. Biochemical basis for the functional switch that regulates hepatocyte growth factor receptor tyrosine kinase activation. Biochemistry. 2008;47(13):4028–38.PubMedCentralCrossRefPubMed Sheth PR, Hays JL, Elferink LA, Watowich SJ. Biochemical basis for the functional switch that regulates hepatocyte growth factor receptor tyrosine kinase activation. Biochemistry. 2008;47(13):4028–38.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Liu L, Zeng W, Wortinger MA, Yan SB, Cornwell P, Peek VL, Stephens JR, Tetreault JW, Xia J, Manro JR et al.: LY2875358, a Neutralizing and Internalizing Anti-MET Bivalent Antibody, Inhibits HGF-Dependent and HGF-Independent MET Activation and Tumor Growth. Clin Cancer Res. 2014;20(23):6059-70. doi: 10.1158/1078-0432.CCR-14-0543. Epub 2014 Sep 17. Liu L, Zeng W, Wortinger MA, Yan SB, Cornwell P, Peek VL, Stephens JR, Tetreault JW, Xia J, Manro JR et al.: LY2875358, a Neutralizing and Internalizing Anti-MET Bivalent Antibody, Inhibits HGF-Dependent and HGF-Independent MET Activation and Tumor Growth. Clin Cancer Res. 2014;20(23):6059-70. doi: 10.​1158/​1078-0432.​CCR-14-0543. Epub 2014 Sep 17.
16.
Zurück zum Zitat Nakajima M, Sawada H, Yamada Y, Watanabe A, Tatsumi M, Yamashita J, et al. The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer. 1999;85(9):1894–902.CrossRefPubMed Nakajima M, Sawada H, Yamada Y, Watanabe A, Tatsumi M, Yamashita J, et al. The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer. 1999;85(9):1894–902.CrossRefPubMed
17.
Zurück zum Zitat Kuniyasu H, Yasui W, Kitadai Y, Yokozaki H, Ito H, Tahara E. Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem Biophys Res Commun. 1992;189(1):227–32.CrossRefPubMed Kuniyasu H, Yasui W, Kitadai Y, Yokozaki H, Ito H, Tahara E. Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem Biophys Res Commun. 1992;189(1):227–32.CrossRefPubMed
18.
Zurück zum Zitat Iwai M, Matsuda M, Iwai Y. Cloning of a cancer cell-producing hepatocyte growth factor, vascular endothelial growth factor, and interleukin-8 from gastric cancer cells. In Vitro Cell Dev Biol Anim. 2003;39(7):288–90.CrossRefPubMed Iwai M, Matsuda M, Iwai Y. Cloning of a cancer cell-producing hepatocyte growth factor, vascular endothelial growth factor, and interleukin-8 from gastric cancer cells. In Vitro Cell Dev Biol Anim. 2003;39(7):288–90.CrossRefPubMed
19.
Zurück zum Zitat Fueller J, Becker M, Sienerth AR, Fischer A, Hotz C, Galmiche A. C-RAF activation promotes BAD poly-ubiquitylation and turn-over by the proteasome. Biochem Biophys Res Commun. 2008;370(4):552–6.CrossRefPubMed Fueller J, Becker M, Sienerth AR, Fischer A, Hotz C, Galmiche A. C-RAF activation promotes BAD poly-ubiquitylation and turn-over by the proteasome. Biochem Biophys Res Commun. 2008;370(4):552–6.CrossRefPubMed
20.
Zurück zum Zitat Ma PC, Schaefer E, Christensen JG, Salgia R. A selective small molecule c-MET Inhibitor, PHA665752, cooperates with rapamycin. Clin Cancer Res. 2005;11(6):2312–9.CrossRefPubMed Ma PC, Schaefer E, Christensen JG, Salgia R. A selective small molecule c-MET Inhibitor, PHA665752, cooperates with rapamycin. Clin Cancer Res. 2005;11(6):2312–9.CrossRefPubMed
21.
Zurück zum Zitat Zou HY, Li Q, Lee JH, Arango ME, Burgess K, Qiu M, et al. Sensitivity of selected human tumor models to PF-04217903, a novel selective c-Met kinase inhibitor. Mol Cancer Ther. 2012;11(4):1036–47.CrossRefPubMed Zou HY, Li Q, Lee JH, Arango ME, Burgess K, Qiu M, et al. Sensitivity of selected human tumor models to PF-04217903, a novel selective c-Met kinase inhibitor. Mol Cancer Ther. 2012;11(4):1036–47.CrossRefPubMed
22.
Zurück zum Zitat Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.CrossRefPubMed
23.
Zurück zum Zitat Zeng ZS, Weiser MR, Kuntz E, Chen CT, Khan SA, Forslund A, et al. c-Met gene amplification is associated with advanced stage colorectal cancer and liver metastases. Cancer Lett. 2008;265(2):258–69.PubMedCentralCrossRefPubMed Zeng ZS, Weiser MR, Kuntz E, Chen CT, Khan SA, Forslund A, et al. c-Met gene amplification is associated with advanced stage colorectal cancer and liver metastases. Cancer Lett. 2008;265(2):258–69.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316(5827):1039–43.CrossRefPubMed Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316(5827):1039–43.CrossRefPubMed
26.
Zurück zum Zitat Strickler JH, Patricia LR, Chia-Jui Y, Chia-Chi L, Yoon-Koo K, Patrick K, et al. Phase 1, open-label, dose escalation,and expansion study of ABT-700, an anti-c-met antibody, in patients (pts) with advanced solid tumors. J Clin Oncol: 2014. 2014;2014:abstr 2507. Strickler JH, Patricia LR, Chia-Jui Y, Chia-Chi L, Yoon-Koo K, Patrick K, et al. Phase 1, open-label, dose escalation,and expansion study of ABT-700, an anti-c-met antibody, in patients (pts) with advanced solid tumors. J Clin Oncol: 2014. 2014;2014:abstr 2507.
27.
Zurück zum Zitat Frampton GM, Ali SM, Rosenzweig M, Chmielecki J, Lu X, Bauer TM, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015. [Epub ahead of print]. Frampton GM, Ali SM, Rosenzweig M, Chmielecki J, Lu X, Bauer TM, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015. [Epub ahead of print].
28.
Zurück zum Zitat Timofeevski SL, McTigue MA, Ryan K, Cui J, Zou HY, Zhu JX, et al. Enzymatic characterization of c-Met receptor tyrosine kinase oncogenic mutants and kinetic studies with aminopyridine and triazolopyrazine inhibitors. Biochemistry. 2009;48(23):5339–49.CrossRefPubMed Timofeevski SL, McTigue MA, Ryan K, Cui J, Zou HY, Zhu JX, et al. Enzymatic characterization of c-Met receptor tyrosine kinase oncogenic mutants and kinetic studies with aminopyridine and triazolopyrazine inhibitors. Biochemistry. 2009;48(23):5339–49.CrossRefPubMed
Metadaten
Titel
Anti-c-Met monoclonal antibody ABT-700 breaks oncogene addiction in tumors with MET amplification
verfasst von
Jieyi Wang
Liliane Goetsch
Lora Tucker
Qian Zhang
Alexandra Gonzalez
Kedar S. Vaidya
Anatol Oleksijew
Erwin Boghaert
Minghao Song
Irina Sokolova
Ekaterina Pestova
Mark Anderson
William N. Pappano
Peter Ansell
Anahita Bhathena
Louie Naumovski
Nathalie Corvaia
Edward B. Reilly
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2016
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2138-z

Weitere Artikel der Ausgabe 1/2016

BMC Cancer 1/2016 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.