<<

Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Molecular Cancer Preclinical Development Therapeutics

Antitumor Action of the MET Tyrosine Inhibitor (PF-02341066) in Gastric Cancer Positive for MET Amplification

Wataru Okamoto1, Isamu Okamoto1, Tokuzo Arao2, Kiyoko Kuwata1, Erina Hatashita1, Haruka Yamaguchi1, Kazuko Sakai2, Kazuyoshi Yanagihara3, Kazuto Nishio2, and Kazuhiko Nakagawa1

Abstract Therapeutic strategies that target the MET hold promise for gastric cancer, but the mechanism underlying the antitumor activity of such strategies remains unclear. We examined the antitumor action of the MET tyrosine kinase inhibitor crizotinib (PF-02341066) in gastric cancer cells positive or negative for MET amplification. Inhibition of MET signaling by crizotinib or RNA interference–mediated MET depletion resulted in induction of accompanied by inhibition of AKT and extracellular signal–regulated kinase phosphorylation in gastric cancer cells with MET amplification but not in those without it, suggesting that MET signaling is essential for the survival of MET amplification–positive cells. Crizotinib upregulated the expression of BIM, a proapoptotic member of the Bcl-2 family, as well as downregulated that of survivin, X-linked inhibitor of apoptosis protein (XIAP), and c-IAP1, members of the inhibitor of apoptosis protein family, in cells with MET amplification. Forced depletion of BIM inhibited crizotinib-induced apoptosis, suggesting that upregulation of BIM contributes to the proapoptotic effect of crizotinib. Crizotinib also exhibited a marked antitumor effect in gastric cancer xenografts positive for MET amplification, whereas it had little effect on those negative for this genetic change. Crizotinib thus shows a marked antitumor action both in vitro and in vivo specifically in gastric cancer cells positive for MET amplification. Mol Cancer Ther; 11(7); 1557–64. 2012 AACR.

Introduction (FDA) for the treatment of patients with Gastric cancer is the second most frequent cause of positive for fusion of the echinoderm microtubule-asso- cancer deaths worldwide (1). has a bene- ciated protein–like 4 (EML4) and anaplastic ficial effect on survival in individuals with advanced- kinase (ALK) genes. This agent also inhibits MET in stage gastric cancer, but even so overall survival is usually addition to oncogenic fusion variants of the tyrosine still only about 1 year (1, 2). Substantial advances in the kinase ALK (9, 10), and it may thus be an attractive development of molecularly targeted therapies for gastric therapeutic option for individuals with gastric cancer cancer have been achieved in recent years (3). Amplifica- positive for MET amplification. We have therefore now tion of the proto- MET is a frequent molecular investigated the effects of crizotinib on cell survival and abnormality in gastric cancer (4–6), and a MET-tyrosine signal transduction in gastric cancer cells with MET kinase inhibitor (TKI) has been shown to induce apoptosis amplification. We further examined the molecular mech- in gastric cancer cells with MET amplification (7, 8). anism underlying the antitumor action of MET inhibition. Crizotinib (PF-02341066; Fig. 1A) was recently approved by the U.S. Food and Drug Administration Materials and Methods Cell culture and reagents Human gastric cancer cell lines positive (SNU5, Authors' Affiliations: Departments of 1Medical Oncology and 2Genome Hs746T, MKN45, HSC58, 58As1, 58As9) or negative Biology, Kinki University Faculty of Medicine, Osaka-Sayama, Osaka; and (SNU1, N87, AGS, MKN1, MKN7, NUGC3, AZ521, 3Laboratory of Health Sciences, Department of Life Sciences, Yasuda Women's University Faculty of Pharmacy, Asaminami, Hiroshima, Japan MKN28, HSC39, SNU216) for MET amplification were obtained as previously described (8, 11). All cells were Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). cultured under a humidified atmosphere of 5% CO2 at 37C in RPMI-1640 medium (Sigma) supplemented with Corresponding Author: Isamu Okamoto, Department of Medical Oncol- ogy, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka- 10% FBS and were passaged for 3 months before renewal Sayama, Osaka 589-8511, Japan. Phone: 81-72-366-0221; Fax: 81-72- from frozen early-passage stocks obtained from the 360-5000; E-mail: [email protected] respective sources. Cells were regularly screened for doi: 10.1158/1535-7163.MCT-11-0934 mycoplasma with the use of a MycoAlert Mycoplasma 2012 American Association for Cancer Research. detection (Lonza). Crizotinib was kindly provided by

www.aacrjournals.org 1557

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Okamoto et al.

Figure 1. Effects of crizotinib on MET, AKT, ERK, and STAT3 phosphorylation, as well as on apoptosis in human gastric cancer cells. A, structure of crizotinib. B, the indicated cell lines were incubated in the absence (Cont) or presence of crizotinib (PF; 200 nmol/L) for 48 hours, after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to phosphorylated (p) or total forms of MET, AKT, ERK, or STAT3, to caspase-3, to PARP, or to b-actin (loading control). The cleaved (cl) forms of caspase-3 and PARP are indicated. C, cells were incubated as in B, after which the number of apoptotic cells was determined by staining with Annexin V followed by flow cytometry. Data are means SD from 3 independent experiments. , P < 0.05 versus the corresponding control value.

Pfizer Global Research & Development, and BEZ235 and Systems; and those to b-actin were from Sigma. All anti- AZD6244 were from ShangHai Biochempartner. bodies were used at a 1:1,000 dilution, with the exception of those to b-actin (1:200). The membrane was then Immunoblot analysis washed with PBS containing 0.05% Tween-20 before incu- Cells were washed twice with ice-cold PBS and bation for 1 hour at room temperature with horseradish then lysed with 1 Cell Lysis Buffer (Cell Signaling peroxidase–conjugated secondary antibodies (GE Health- Technology) containing 20 mmol/L Tris-HCl (pH 7.5), care). Immune complexes were finally detected with ECL 150 mmol/L NaCl, 1 mmol/L EDTA (disodium salt), Western Blotting Detection Reagents (GE Healthcare). 1 mmol/L EGTA, 1% Triton X-100, 2.5 mmol/L sodium pyrophosphate, 1 mmol/L b-glycerophosphate, 1 mmol/L Gene silencing m Na3VO4, leupeptin (1 g/mL), and 1 mmol/L phenyl- Cells were plated at 50% to 60% confluence in 6-well methylsulfonyl fluoride. The protein concentration of plates or 25-cm2 flasks and then incubated for 24 hours cell lysates was determined with a BCA Protein Assay Kit before transient transfection for the indicated times with (Thermo Fisher Scientific), and equal amounts of protein siRNAs mixed with the Lipofectamine reagent (Invitro- were subjected to SDS-PAGE on a 7.5% or 12% gel (Bio- gen). The siRNAs specific for MET (MET-1, 50-ACAA- Rad). The separated proteins were transferred to a nitro- GAUCGUCAACAAAAA-30;MET-2,50-CUACAGAAAU- cellulose membrane, which was then incubated with GGUUUCAAA-30), BIM (BIM-1, 50-GGAGGGUAUUU- Blocking One solution (Nacalai Tesque) for 20 minutes at UUGAAUAA-30; BIM-2, 50-AGGAGGGUAUUUUUGA- room temperature before incubation overnight at 4C AUA-30), or AKT (AKT-1, 50-CCAGGUAUUUUGAU- with primary antibodies. Rabbit polyclonal antibodies to GAGGA-30;AKT-2,50-CAACCGCCAUCCAGACUGU- phosphorylated human MET (pY1234/pY1235), total 30) mRNAs, as well as corresponding scrambled (control) AKT, phosphorylated AKT, phosphorylated extracellular siRNAs were obtained from Nippon EGT. The data pre- signal–regulated kinase (ERK), total STAT3, phosphory- sented for the effects of MET, BIM, or AKT depletion were lated STAT3 (phospho-Tyr705), PARP, caspase-3, BIM, obtained with the corresponding siRNA-1, but similar Bcl-2, and X-linked inhibitor of apoptosis protein (XIAP) results were obtained with each siRNA-2. were obtained from Cell Signaling Technology; those to total ERK were from Santa Cruz Biotechnology; those Annexin V–binding assay to total MET were from Zymed/Invitrogen; those to sur- The binding of Annexin V to cells was measured with vivin were from Novus; those to c-IAP1 were from R&D the use of an Annexin-V-FLUOS Staining Kit (Roche).

1558 Mol Cancer Ther; 11(7) July 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Antitumor Action of Crizotinib in Gastric Cancer

Cells were harvested by exposure to trypsin-EDTA, Table 1. IC values of crizotinib for inhibition of washed with PBS, and centrifuged at 200 g for 5 minutes. 50 the growth of gastric cancer cells in vitro The cell pellets were resuspended in 100 mL of Annexin-V- FLUOS labeling solution, incubated for 10 to 15 minutes at MET 15 to 25 C, and then analyzed for fluorescence with a flow Crizotinib Crizotinib response Cell line IC , mmol/L amplification cytometer (FACSCalibur) and Cell Quest software (Bec- 50 ton Dickinson). Sensitive MKN45 0.04 þ HSC58 0.05 þ Cell growth inhibition assay 58As1 0.06 þ Cells were transferred to 96-well flat-bottomed plates 58As9 0.17 þ and cultured for 24 hours before exposure to various SNU5 0.03 þ concentrations of crizotinib for 72 hours. Tetra Color One Hs746T 0.05 þ (5 mmol/L tetrazolium monosodium salt and 0.2 mmol/L Resistant MKN1 8.57 1-methoxy-5-methyl phenazinium methylsulfate; Seika- AZ521 1.96 gaku Kogyo) was then added to each well, and the cells SNU216 4.19 were incubated for 3 hours at 37C before measurement of N87 6.10 absorbance at 490 nm with a Multiskan Spectrum instru- MKN7 >10 ment (Thermo Labsystems). Absorbance values were SNU1 0.80 expressed as a percentage of that for untreated cells, and HSC39 2.75 the concentration of crizotinib resulting in 50% growth AGS 0.90 inhibition (IC50) was calculated. MKN28 3.73 NUGC3 2.65 in vivo Growth inhibition assay NOTE: Data are means of triplicates from experiments that All animal studies were conducted in accordance with were repeated a total of 3 times with similar results. the Recommendations for Handling of Laboratory Ani- mals for Biomedical Research compiled by the Committee on Safety and Ethical Handling Regulations for Labora- tory Animal Experiments, Kinki University (Osaka, negative for MET amplification. Six cell lines with MET Japan). The ethical procedures followed conformed to the amplification (MKN45, HSC58, 58As1, 58As9, SNU5, guidelines of the United Kingdom Co-ordinating Com- Hs746T) were sensitive to crizotinib, with IC50 values of mittee on Cancer Research (12). Tumors cells (5 106) less than 200 nmol/L (Table 1). In contrast, crizotinib did were injected subcutaneously into the axilla of 5- to 6- not substantially inhibit the proliferation of gastric cancer week-old female athymic nude mice (BALB/c nu/nu; cells without such gene amplification (Table 1). Tran- CLEA Japan). Treatment was initiated when tumors in scripts of the EML4–ALK were not detected each group of 6 mice achieved an average volume of 300 to in the crizotinib-sensitive gastric cancer cell lines by 900 mm3. Treatment groups consisted of vehicle control reverse transcription PCR analysis (data not shown). and crizotinib (25 or 50 mg/kg). Crizotinib was adminis- These data suggested that crizotinib has a marked anti- tered by oral gavage daily for 4 weeks, with control proliferative effect specifically in gastric cancer cells with animals receiving a 0.5% (w/v) aqueous solution of MET amplification. hydroxypropylmethylcellulose as vehicle. Tumor volume was determined from caliper measurements of tumor Effects of crizotinib on downstream signaling of MET L W and on apoptosis in gastric cancer cells with or length ( ) and width ( ) according to the formula MET LW2/2. Both tumor size and body weight were measured without amplification twice per week. We next examined the effects of crizotinib on phos- phorylation of ERK, AKT, and STAT3 in gastric cancer cell lines. Crizotinib markedly inhibited the phosphorylation Statistical analysis of ERK, AKT, and STAT3, as well as that of MET in cells Quantitative data are presented as means SD or SEM with MET amplification (Fig. 1B; Supplementary Fig. S1). from 3 independent experiments or for 6 animals per In contrast, crizotinib had little effect on the phosphory- group, unless indicated otherwise, and were analyzed lation of ERK, AKT, or STAT3 in gastric cancer cells with the Student 2-tailed t test. A P value of <0.05 was without amplification of MET (Fig. 1B). Determination of considered statistically significant. cell-cycle distribution in SNU5, HSC58, 58As1, and 58As9 cells, all of which manifest MET amplification, revealed Results that treatment with crizotinib for 72 hours increased the Crizotinib inhibits the proliferation of gastric cancer size of the cell population in sub-G1 phase, indicative of cells with MET amplification the induction of apoptosis, as well as reduced that of the We first examined the effect of the MET-TKI crizotinib cell population in S-phase (Supplementary Fig. S2). We on the proliferation of gastric cancer cells positive or further investigated the effect of crizotinib on apoptosis.

www.aacrjournals.org Mol Cancer Ther; 11(7) July 2012 1559

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Okamoto et al.

Immunoblot analysis showed that crizotinib triggered in SNU5 and 58As9 cells, both of which manifest MET the generation of the cleaved forms of caspase-3 and amplification (Fig. 2B). Similartotheeffectsofcrizoti- PARP in cells with MET amplification but not in those nib, depletion of MET by RNA interference (RNAi) also without it (Fig. 1B). Consistent with these results, an resulted in upregulation of BIM and downregulation of Annexin V–binding assay revealed that crizotinib members of the IAP family, whereas it had little effect induced a substantial level of apoptosis in MET ampli- on the expression of Bcl-2 in such cells (Fig. 2B). These fication–positive cells but was largely without effect in data thus suggested that the upregulation of BIM and cell lines without MET amplification (Fig. 1C). These the downregulation of members of the IAP family, data thus suggested that crizotinib inhibits the phos- including survivin, XIAP, and c-IAP1, are related to the phorylation of ERK, AKT, and STAT3, resulting in induction of apoptosis by the MET inhibitor in gastric induction of apoptosis, in gastric cancer cells with MET cancer cells with MET amplification. amplification, whereas such effects were not observed in cells without MET amplification. Inhibition of the MEK-ERK or PI3K-AKT pathways results in BIM upregulation and survivin Effects of crizotinib on the expression of apoptosis- downregulation, respectively, in MET amplification– related proteins in MET amplification–positive positive cells gastric cancer cells To identify the signaling pathways responsible for Given that crizotinib induced apoptosis in MET ampli- upregulation or downregulation of apoptosis-related pro- fication–positive gastric cancer cells, we examined the teins by crizotinib, we examined the effects of specific effects of this drug on the expression of apoptosis-related inhibitors of the mitogen-activated protein (MAP)/ERK proteins in such cells. Crizotinib upregulated the expres- kinase (MEK) and of phosphoinositide 3-kinase (PI3K) in sion of BIM, a proapoptotic member of the Bcl-2 family of MET amplification–positive cell lines. The MEK inhibitor proteins, whereas it had little effect on the expression of AZD6244 induced BIM expression without affecting the other Bcl-2 family members including Bcl-2 (Fig. 2A). abundance of the other proteins examined (Fig. 3A), Furthermore, crizotinib downregulated the expression of suggesting that expression of BIM is regulated by the members of the IAP family including survivin, XIAP, and MEK-ERK pathway. On the other hand, the PI3K inhibitor c-IAP1 in cells with MET amplification (Fig. 2A). BEZ235 reduced the abundance of survivin without To verify that the effects of crizotinib on the expres- affecting that of the other IAP family proteins including sion of apoptosis-related proteins in MET amplifica- c-IAP1 and XIAP (Fig. 3A). We also found that depletion tion–positive gastric cancer cells are indeed mediated of AKT by RNAi resulted in downregulation of survivin by MET inhibition rather than by nonspecific inhibition but not of XIAP or c-IAP1 in the MET amplification– of other , we transfected gastric cancer cells with positive SNU5 and 58As9 cell lines (Fig. 3B). Together, siRNAs specific for MET mRNA. Transfection with these data suggested that crizotinib regulates BIM and METsiRNAsresultedinamarkeddecreaseinthe survivin expression through inhibition of the MEK-ERK abundance of MET, which was accompanied by gener- and PI3K-AKT signaling pathways, respectively, in MET ation of the cleaved forms of both caspase-3 and PARP, amplification–positive gastric cancer cells.

Figure 2. Effects of MET inhibition or depletion on the expression of Bcl-2 family and IAP family proteins in human gastric cancer cells. The indicated cell lines were incubated with or without crizotinib (200 nmol/L) for 24 hours (A) or were transfected with nonspecific (Cont) or MET siRNAs for 48 hours (B), after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to the indicated proteins. The position of the band corresponding to BIMEL is indicated.

1560 Mol Cancer Ther; 11(7) July 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Antitumor Action of Crizotinib in Gastric Cancer

Figure 3. Effects of inhibition of the MEK-ERK or PI3K-AKT signaling pathways on the expression of Bcl-2 family and IAP family proteins in MET amplification–positive gastric cancer cells. The indicated cell lines were incubated in the absence (Cont, 0.1% dimethyl sulfoxide) or presence of AZD6244 (0.2 mmol/L) or BEZ235 (0.2 mmol/L) for 24 hours (A) or were transfected with nonspecific (Cont) or AKT siRNAs for 48 hours (B), after which cell lysates were prepared and subjected to immunoblot analysis with antibodies to the indicated proteins.

Role of BIM induction in crizotinib-induced ciated with inhibition of crizotinib-induced apoptosis, as apoptosis in gastric cancer cells with MET revealed by a reduced extent of caspase-3 and PARP amplification cleavage (Fig. 4A). The Annexin V–binding assay also Induction of the proapoptotic BH3-only protein BIM revealed that such transfection resulted in inhibition of has been found to be important for TKI-induced apoptosis crizotinib-induced apoptosis (Fig. 4B). These data thus in EGF receptor (EGFR) gene mutation–positive lung suggested that the induction of apoptosis by crizotinib in cancer and HER2 amplification–positive gastric cancer cells with MET amplification is mediated, at (13–17). To investigate whether the upregulation of BIM least in part, by upregulation of BIM. is related to the induction of apoptosis by crizotinib, we transfected MET amplification–positive gastric cancer Effect of crizotinib on the growth of gastric cancer cells with siRNAs specific for BIM mRNAs. Such trans- cells in vivo fection resulted in marked inhibition of the upregulation To determine whether the antitumor action of crizotinib of BIM by crizotinib (Fig. 4A). Immunoblot analysis observed in vitro might also be apparent in vivo,we showed that the attenuation of BIM induction was asso- injected 58As9 cells (positive for MET amplification),

Figure 4. Effect of BIM depletion on apoptosis induced by crizotinib in gastric cancer cells with MET amplification. A, the indicated cell lines were transfected with BIM or nonspecific siRNAs for 24 hours and then incubated for 48 hours with or without crizotinib (200 nmol/L). The cells were then lysed and subjected to immunoblot analysis with antibodies to the indicated proteins. B, cells treated as in (A) were assayed for apoptosis by staining with Annexin V followed by flow cytometry. Data are means SD from 3 independent experiments. , P < 0.05 for the indicated comparisons.

www.aacrjournals.org Mol Cancer Ther; 11(7) July 2012 1561

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Okamoto et al.

Figure 5. Effect of crizotinib on the growth of gastric cancer cells in vivo. Nude mice with tumor xenografts established by subcutaneous injection of 58As9, SNU5, AZ521, or MKN28 cells were treated daily for 4 weeks with vehicle (control) or crizotinib (25 or 50 mg/kg). Tumor volume was determined at the indicated times after the onset of treatment. Data are means SEM for 6 mice per group. P values are for the indicated comparisons at 28 days.

SNU5 cells (positive for MET amplification), AZ521 cells efficacy. The MET is considered (negative for MET amplification), or MKN28 cells (nega- one such potential target in cancer, and several MET-TKIs tive for MET amplification) into nude mice to elicit the are currently undergoing clinical trials in humans. Ampli- formation of solid tumors. After tumor formation, the fication of MET is often responsible for activation of MET mice were treated with vehicle (control) or with crizotinib signaling, with such amplification occurring frequently in at a daily dose of 25 or 50 mg/kg by oral gavage for 4 gastric cancer (4–6). We have now shown that crizotinib weeks. Crizotinib at either dose eradicated tumors in mice exerted a marked antitumor action in gastric cancer cells injected with 58As9 or SNU5 cells (Fig. 5; Supplementary with MET amplification. In such gastric cancer cells, Fig. S3). In contrast, tumors in mice injected with AZ521 or attenuation of MET function either by treatment with MKN28 cells were not affected by crizotinib treatment crizotinib or by MET-targeted RNAi resulted in inhibition even at the dose of 50 mg/kg/d (Fig. 5; Supplementary of AKT and ERK signaling as well as in the induction of Fig. S3). Treatment with crizotinib at either dose was well- apoptosis, indicating that tumor cells with MET amplifi- tolerated by the mice, with no signs of toxicity or weight cation are dependent on MET signaling for their growth loss during therapy (data not shown). Crizotinib thus and survival. Targeting of MET signaling by MET-TKIs is exhibited a marked antitumor effect in gastric cancer thus a potentially valuable therapeutic strategy for xenografts positive for MET amplification, whereas it had patients with gastric cancer with MET amplification. little effect on those negative for MET amplification, We also investigated the mediators of crizotinib- consistent with our results obtained in vitro. induced apoptosis in MET amplification–positive gastric cancer cells. We found that crizotinib induced upregula- tion of BIM, a key proapoptotic member of the Bcl-2 family Discussion of proteins that initiates apoptosis signaling by binding to Aberrant activation of receptor tyrosine kinase signal- and antagonizing the function of prosurvival Bcl-2 family ing pathways contributes to the development of various members (18). Furthermore, depletion of BIM by RNAi types of cancer. Small-molecule inhibitors that target these resulted in inhibition of crizotinib-induced apoptosis in activated kinases have been developed and have shown gastric cancer cells with MET amplification, indicating substantial efficacy in clinical trials. The identification of that upregulation of BIM contributes to the induction of patient subgroups that might actually benefit from treat- apoptosis by the MET-TKI in such cells. These findings are ment with such drugs would be expected to optimize their consistent with those of previous studies of lung cancer

1562 Mol Cancer Ther; 11(7) July 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Antitumor Action of Crizotinib in Gastric Cancer

cells with EGFR mutations and breast cancer cells with amplification–positive gastric cancer cells, the mecha- HER2 amplification (13–17). However, our observations nism by which the expression of such proteins is reg- revealed that depletion of BIM did not completely abolish ulated likely differs between MET and other receptor crizotinib-induced apoptosis, suggesting that another tyrosine kinases including EGFR and HER2. However, apoptotic regulator might contribute to MET-TKI– the signaling pathway responsible for downregulation induced apoptotic cell death. We also found that crizoti- of XIAP and c-IAP1 by MET inhibition remains nib induced downregulation of survivin, a member of the unknown. Further studies are thus required to clarify IAP family that protects cells against apoptosis by either the regulation of XIAP and c-IAP1 and the contribution directly or indirectly inhibiting the activation of effector of members of the IAP family to MET-TKI–induced caspases (19). We found that a PI3K inhibitor or RNAi- apoptosis in gastric cancer cells with MET amplification. mediated depletion of AKT reduced the abundance of In conclusion, our results have shown that crizotinib survivin, indicating that the expression of survivin is has pronounced effects on signal transduction and sur- regulated by PI3K-AKT signaling in MET amplifica- vival in gastric cancer cells with MET amplification. Cri- tion–positive gastric cancer cells. Previous studies have zotinib has recently been approved for treatment of ALK- shown that the expression of survivin is dependent on driven lung cancer by the FDA on the basis of safety and PI3K-AKT signaling that operates downstream of recep- effectiveness data. The IC50 values of crizotinib for inhi- tor tyrosine kinases and is essential for cell survival in bition of the growth of MET amplification–positive gastric EGFR mutation–positive non–small cell lung cancer cells cancer cell lines were lower than the mean trough con- as well as in breast cancer cells positive for HER2 ampli- centration of the drug achieved in the plasma of patients at fication (16, 17). These results suggest that downregula- steady state (292 ng/mL or 644 nmol/L; ref. 21). Indeed, tion of survivin via inhibition of the MET-PI3K-AKT crizotinib was recently found to exhibit antitumor activity pathway likely also contributes to the induction of apo- in 2 of 4 patients with MET amplification–positive gas- ptosis by crizotinib in MET amplification–positive gastric troesophageal cancer (22), supporting further study of the cancer cells. molecular mechanism underlying its antitumor action. In We have shown that crizotinib induced downregula- the present study, we showed that BIM and IAP family tion of XIAP and c-IAP1 in gastric cancer cells with MET members including survivin, XIAP, and c-IAP1 play a role amplification. We further showed that depletion of MET in crizotinib-induced apoptosis in association with inhi- by RNAi induced downregulation of XIAP and c-IAP1, bition of MET signaling in gastric cancer cells with MET indicating that these proteins are also regulated by MET amplification. Our observations provide a basis for the signaling in MET amplification–positive gastric cancer further development of MET- for cells. We investigated which signaling pathway is patients with gastric cancer with MET amplification. responsible for downregulation of XIAP and c-IAP1 resulting from inhibition of MET. Given that crizotinib Disclosure of Potential Conflicts of Interest inhibitedbothERKandAKTphosphorylationinMET No potential conflicts of interest were disclosed. amplification–positive cell lines, we examined the Authors' Contributions effects both of specific inhibitors of MEK and PI3K as Conception and design: W. Okamoto, I. Okamoto, K. Nakagawa well as of siRNAs specific for AKT mRNA in such cells. Development of methodology: W. Okamoto, T. Arao, K. Sakai, However, none of these agents induced downregulation K. Yanagihara Acquisition of data (provided animals, acquired and managed patients, of XIAP or c-IAP1 in gastric cancer cells with MET provided facilities, etc.): W. Okamoto, I. Okamoto, K. Kuwata, E. Hata- amplification. We previously showed that activation of shita, H. Yamaguchi, K. Sakai STAT3 is linked to MET signaling and that forced Analysis and interpretation of data (e.g., statistical analysis, biostatis- tics, computational analysis): W. Okamoto, I. Okamoto, K. Kuwata, expression of a constitutively active form of STAT3 E. Hatashita, H. Yamaguchi attenuated MET-TKI–induced apoptosis in MET-acti- Writing, review, and/or revision of the manuscript: W. Okamoto, I. Okamoto, K. Nishio, K. Nakagawa vated gastric cancer cells, suggesting that inhibition of Administrative, technical, or material support (i.e., reporting or orga- STAT3 activity contributes to MET-TKI–induced apo- nizing data, constructing databases): T. Arao, K. Kuwata, E. Hatashita, ptosis (20). To investigate whether the regulation of H. Yamaguchi, K. Sakai, K. Yanagihara Study supervision: I. Okamoto, K. Nakagawa XIAP or c-IAP1 expression is mediated by STAT3 sig- naling, we transfected SNU5 and 58As9 cells, both of Acknowledgments which manifest MET amplification, with an siRNA that The authors thank Pfizer for the provision of crizotinib (PF-02341066) targetsSTAT3mRNA.However,depletionofSTAT3by used in this study. RNAi had no substantial effect on expression of XIAP and c-IAP1 in such cells (data not shown). Previous Grant Support The authors received no grant support for this study. studies have shown that XIAP and c-IAP1 were not The costs of publication of this article were defrayed in part by the substantially affected by EGFR-TKIs in EGFR mutation– payment of page charges. This article must therefore be hereby marked positive non–small cell lung cancer cells or by HER2- advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. targeting agents in breast cancer cells positive for HER2 amplification (16, 17). Given that inhibition of MET Received November 16, 2011; revised March 30, 2012; accepted April 11, results in downregulation of XIAP and c-IAP1 in MET 2012; published OnlineFirst June 22, 2012.

www.aacrjournals.org Mol Cancer Ther; 11(7) July 2012 1563

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Okamoto et al.

References 1. Hartgrink HH, Jansen EP, van Grieken NC, van de Velde CJ. Gastric 12. United Kingdom Co-ordinating Committee on Cancer Research cancer. Lancet 2009;374:477–90. (UKCCCR). Guidelines for the welfare of animals in experimental 2. Wesolowski R, Lee C, Kim R. Is there a role for second-line neoplasia (second edition). Br J Cancer 1998;77:1–10. chemotherapy in advanced gastric cancer? Lancet Oncol 2009;10: 13. Costa DB, Halmos B, Kumar A, Schumer ST, Huberman MS, Boggon 903–12. TJ, et al. BIM mediates EGFR tyrosine kinase inhibitor-induced apo- 3. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, ptosis in lung cancers with oncogenic EGFR mutations. PLoS Med et al. in combination with chemotherapy versus chemo- 2007;4:1669–79; discussion 1680. therapy alone for treatment of HER2-positive advanced gastric or 14. Cragg MS, Kuroda J, Puthalakath H, Huang DC, Strasser A. Gefitinib- gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, induced killing of NSCLC cell lines expressing mutant EGFR requires randomised controlled trial. Lancet 2010;376:687–97. BIM and can be enhanced by BH3 mimetics. PLoS Med 2007;4:1681– 4. Kuniyasu H, Yasui W, Kitadai Y, Yokozaki H, Ito H, Tahara E. Frequent 89; discussion 1690. amplification of the c-met gene in scirrhous type stomach cancer. 15. Gong Y, Somwar R, Politi K, Balak M, Chmielecki J, Jiang X, et al. Biochem Biophys Res Commun 1992;189:227–32. Induction of BIM is essential for apoptosis triggered by EGFR kinase 5. Nessling M, Solinas-Toldo S, Wilgenbus KK, Borchard F, Lichter P. inhibitors in mutant EGFR-dependent lung adenocarcinomas. PLoS Mapping of chromosomal imbalances in gastric adenocarcinoma Med 2007;4:e294. revealed amplified protooncogenes MYCN, MET, WNT2, and ERBB2. 16. Okamoto K, Okamoto I, Okamoto W, Tanaka K, Takezawa K, Kuwata Genes Chromosomes Cancer 1998;23:307–16. K, et al. Role of survivin in EGFR inhibitor-induced apoptosis in non- 6. Sakakura C, Mori T, Sakabe T, Ariyama Y, Shinomiya T, Date K, et al. small cell lung cancers positive for EGFR mutations. Cancer Res Gains, losses, and amplifications of genomic materials in primary 2010;70:10402–10. gastric cancers analyzed by comparative genomic hybridization. 17. Tanizaki J, Okamoto I, Fumita S, Okamoto W, Nishio K, Nakagawa K. Genes Chromosomes Cancer 1999;24:299–305. Roles of BIM induction and survivin downregulation in - 7. Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald induced apoptosis in breast cancer cells with HER2 amplification. H, et al. Amplification of MET may identify a subset of cancers with Oncogene 2011;30:4097–106. extreme sensitivity to the selective tyrosine kinase inhibitor PHA- 18. Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, et al. 665752. Proc Natl Acad Sci U S A 2006;103:2316–21. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only 8. Okamoto W, Okamoto I, Yoshida T, Okamoto K, Takezawa K, Hata- ligands allows complementary apoptotic function. Mol Cell 2005;17: shita E, et al. Identification of c-Src as a potential therapeutic target for 393–403. gastric cancer and of MET activation as a cause of resistance to c-Src 19. Hengartner MO. The biochemistry of apoptosis. Nature 2000;407: inhibition. Mol Cancer Ther 2010;9:1188–97. 770–6. 9. Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, et al. An 20. Okamoto W, Okamoto I, Arao T, Yanagihara K, Nishio K, Nakagawa orally available small-molecule inhibitor of c-Met, PF-2341066, exhi- K. Differential roles of STAT3 depending on the mechanism of bits cytoreductive antitumor efficacy through antiproliferative and STAT3 activation in gastric cancer cells. Br J Cancer 2011;105: antiangiogenic mechanisms. Cancer Res 2007;67:4408–17. 407–12. 10. Tanizaki J, Okamoto I, Okamoto K, Takezawa K, Kuwata K, Yamaguchi 21. Bang YJ, Kwak EL, Shaw AT, Camidge DR, Iafrate AJ, Maki RG, et al. H, et al. MET tyrosine kinase inhibitor crizotinib (PF-02341066) shows Clinical activity of the oral ALK inhibitor PF-02341066 in ALK-positive differential antitumor effects in non-small cell lung cancer according to patients with non-small cell lung cancer (NSCLC). J Clin Oncol 28:18s, MET alterations. J Thorac Oncol 2011;6:1624–31. 2010 (suppl; abstr 3). 11. Yanagihara K, Takigahira M, Tanaka H, Komatsu T, Fukumoto H, 22. Lennerz JK, Kwak EL, Ackerman A, Michael M, Fox SB, Bergethon K, Koizumi F, et al. Development and biological analysis of peritoneal et al. MET amplification identifies a small and aggressive subgroup metastasis mouse models for human scirrhous stomach cancer. of esophagogastric adenocarcinoma with evidence of responsive- Cancer Sci 2005;96:323–32. ness to crizotinib. J Clin Oncol 2011;29:4803–10.

1564 Mol Cancer Ther; 11(7) July 2012 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst June 22, 2012; DOI: 10.1158/1535-7163.MCT-11-0934

Antitumor Action of the MET Tyrosine Kinase Inhibitor Crizotinib (PF-02341066) in Gastric Cancer Positive for MET Amplification

Wataru Okamoto, Isamu Okamoto, Tokuzo Arao, et al.

Mol Cancer Ther 2012;11:1557-1564. Published OnlineFirst June 22, 2012.

Updated version Access the most recent version of this article at: doi:10.1158/1535-7163.MCT-11-0934

Supplementary Access the most recent supplemental material at: Material http://mct.aacrjournals.org/content/suppl/2012/04/17/1535-7163.MCT-11-0934.DC1

Cited articles This article cites 22 articles, 5 of which you can access for free at: http://mct.aacrjournals.org/content/11/7/1557.full#ref-list-1

Citing articles This article has been cited by 7 HighWire-hosted articles. Access the articles at: http://mct.aacrjournals.org/content/11/7/1557.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://mct.aacrjournals.org/content/11/7/1557. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mct.aacrjournals.org on September 23, 2021. © 2012 American Association for Cancer Research.