Abstract
As one of the common malignant cancer types, gastric cancer (GC) is known for late‐stage diagnosis and poor prognosis. Overexpression of the receptor tyrosine kinase MET is associated with poor prognosis among patients with advanced stage GC. However, no MET inhibitor has been used for GC treatment. Like other tyrosine kinase inhibitors that fit the “occupancy‐driven” model, current MET inhibitors are prone to acquired resistance. The emerging proteolysis targeting chimera (PROTAC) strategy could overcome such limitations through direct degradation of the target proteins. In this study, we successfully transformed the MET‐targeted inhibitor crizotinib into a series of PROTACs, recruiting cereblon/cullin 4A E3 ubiquitin ligase to degrade the MET proteins. The optimized lead PROTAC (PRO‐6 E) effectively eliminated MET proteins in vitro and in vivo, inhibiting proliferation and motility of MET‐positive GC cells. In the MKN‐45 xenograft model, PRO‐6 E showed pronounced antitumor efficacy with a well‐tolerated dosage regimen. These results validated PRO‐6 E as the first oral PROTAC for MET‐dependent GC.
Keywords: gastric cancer, MET, proteolysis targeting chimera, receptor tyrosine kinase, ubiquitin‐mediated proteasome degradation
Proteolysis targeting chimera degrader PRO‐6 E specifically induced degradation of targeted proteins, and led to the suppression of tumor cell proliferation, motility, and invasiveness in MET‐positive gastric cancer models.

Abbreviations
- Co‐IP
co‐immunoprecipitation
- CRBN
cereblon
- CRO
crizotinib
- Dmax
maximum degradation concentration
- EMT
epithelial–mesenchymal transition
- GC
gastric cancer
- MS/MS
tandem mass spectrometry
- POI
protein of interest
- POM
pomalidomide
- PRO‐6 E‐Me
methylated PRO‐6 E
- PROTAC
proteolysis targeting chimera
- RTK
receptor tyrosine kinase
- SPPIER
separation of phases‐based protein interaction reporter
- UPS
ubiquitin–proteasome system
- VHL
von Hippel–Lindau
1. INTRODUCTION
Gastric cancer accounts for 5.6% of malignant cancers worldwide, and is known for low overall survival rate and poor prognosis. 1 Despite recent advancements in diagnosis and targeted therapies, there are no effective treatment options for GC because of high tumor heterogeneity, late diagnosis, and acquired resistance. 2 , 3 , 4 Approximately 37% of GC patients are partially amenable to RTK/RAS‐targeted therapy, because alterations in RTKs is a signature feature of GC. 5 , 6
Encoded by the proto‐oncogene MET, the RTK MET (also known as hepatocyte growth factor receptor, scatter factor receptor, and c‐Met) is among the most frequently aberrantly activated RTKs, 7 , 8 playing a major role in tumorigenesis and metastasis, especially among patients with advanced stage GC. 2 , 9 , 10 , 11 , 12 The amplification of the MET gene has been reported in approximately 4% of GC patients, 5 , 6 while high levels of MET mRNA have been detected in 20%–30% of GC patients. 12 Over 65% of GC biopsy samples showed high levels of MET protein, which facilitates metastatic progression. 13 , 14 Aberrant tumorigenic activation of MET has been considered a key driver and prognostic factor for GC. Preclinical studies strongly support MET as a potential target for MET‐dependent GC treatment. 12 However, MET‐targeting treatments showed limited activities in the past, and there are no approved GC drugs targeting MET. Therefore, a new therapeutic regimen is in urgent need. 15 , 16
Proteolysis targeting chimera has recently been successfully applied to degrade various pathogenic proteins, including kinases, transcription factors, and membrane‐bound receptors. 17 Proteolysis targeting chimera molecules consist of two active moieties, a targeting warhead for intracellular POI and an E3 ligase ligand, connected by the linker. 18 , 19 , 20 These ternary chemical complexes mediate the degradation of POI by recruiting E3 ligase to the specific target, followed by protein removal through the UPS. 21 Due to their “event‐driven” pharmacological mode, PROTACs afford the opportunities to reduce acquired drug resistance and toxicity 22 , 23 , 24 compared with the “occupancy‐driven” mode of conventional RTK inhibitors. 25 Through the degradation of specific POIs, PROTACs could yield sustained suppression of RTKs and related signaling pathways, even for the kinase‐independent functions (e.g., scaffolding). 26 , 27 , 28 , 29 The application of the PROTAC strategy for MET‐targeted degradation holds great promise for GC treatment.
In this study, we successfully designed and synthesized a series of MET‐targeted PROTACs, based on the MET inhibitor CRO. The lead compound PRO‐6 E promoted a rapid, efficient, and durable degradation of MET, following the formation of the ternary complex and the activation of UPS. In addition, PRO‐6 E significantly inhibited the proliferation, migration, and invasion of MET‐overexpressing GC cells. The therapeutic potential and mechanism(s) of action for PRO‐6 E were extensively examined, using in vitro and in vivo models for GC. Our findings afforded a new strategy for the treatment of MET‐dependent GC, further highlighting the potential of PRO‐6 E as the first oral PROTAC for MET‐dependent cancers.
2. MATERIALS AND METHODS
2.1. Antibodies
Antibodies for MET (# 8198, 1:1000), p‐MET (Tyr1234/1235, # 3077, 1:000), GAPDH (#51332, 1:1000), CRBN (#71810, 1:1000), ERK (#4695, 1:1000), p‐ERK (Thr202/Tyr204, #4370, 1:1000), E‐cadherin (#3195, 1:1000), N‐cadherin (#13116, 1:1000), vimentin (#5741, 1:1000), HRP‐conjugated anti‐rabbit IgG (#7074, 1:5000), HRP‐conjugated anti‐mouse IgG (#7076, 1:5000), and Alexa Fluor 488 conjugated‐anti‐rabbit IgG (#4340, 1:1000) were purchased from Cell Signaling Technology. The Abs for c‐Myc (Cat# ab32072, 1:1000), ubiquitin (ab134953, 1:500), IKZF1 (ab191394, 1:1000), IKZF3 (ab139408, 1:1000), and GSPT1 (ab234433, 1:1000) were obtained from Abcam.
2.2. Cell lines and cell culture
MKN‐45 cells with MET amplification and HEK293T cells were from the National Collection of Cell Bank of the Chinese Academy of Sciences. SNU‐638 and SNU‐1 cells were all purchased from the Zhong Qiao Xin Zhou Biotechnology Co., Ltd. All GC cell lines were cultured in RPMI‐1640 medium (#11875093; Gibco), and HEK293T cells in DMEM medium (#2120620). All mediums were supplemented with 10% FBS (#10099141C), 1% penicillin, and 1% streptomycin (SV30010; HyClone). All cells were maintained under 5% CO2 air at 37°C.
2.3. Cell viability assay
MKN45, SNU‐638, and SNU‐1 cells were seeded into 96‐well plates at the density of 5 × 10 4 cells/ml overnight and were exposed to drugs. At the end of treatment, CCK‐8 reagent (#MA0218; MeilunBio) was added and the cell viability was determined using the Cytation 5 Reader (Agilent BioTek).
2.4. Western blot analysis
Cells were lysed in ice‐cold RIPA buffer, supplemented with 1% phosphatase inhibitors (#MB12707; MeilunBio) and 1% cOmplete protease inhibitors (#11697498001; Sigma‐Aldrich). The concentration of proteins was determined by the BCA Assay Kit (#P0010S). Then, 10 μg proteins were separated on 7.5% SDS‐PAGE and transferred to PVDF membranes (#10600021; GE Healthcare). The membrane was then blocked and incubated with Abs. The signals were detected using the Tanon Imaging System.
2.5. Detection of ubiquitin MET protein
MKN‐45 cells (3 × 105 cells/well) were grown in 6‐well plates and exposed to PRO‐6 E (1 μM) for different time periods. Cells were lysed and the total proteins were separated by 7.5% SDS‐PAGE gel (EpiZyme). The membrane was then blocked and incubated with ubiquitin Ab. The signals were detected using the Tanon Imaging System. To verify whether the polyubiquitination process induced by PRO‐6 E (1 μM) was dependent on the ubiquitin proteasome, MKN‐45 cells were pretreated with MG132 (5 μM, 2 h) following 24 h of incubation with PRO‐6 E (1 μM). The signals were detected using the Tanon Imaging System.
2.6. Immunofluorescence
MKN‐45 cells were seeded into 35‐mm dishes (BS‐20‐GJM) and treated with PRO‐6 E for 48 h. The cells were fixed with 4% paraformaldehyde and blocked with 1% BSA. The anti‐MET primary Ab was added at 4°C overnight. Alexa Fluor 488‐conjugated goat anti‐rabbit IgG was added for 1 h and the cell nuclei were stained with DAPI (ab228549). Cells were photographed using either a confocal laser scanning microscope (Leica) or high content analysis system (PerkinElmer).
2.7. Co‐immunoprecipitation
MKN‐45 cells were seeded at the density of 4 × 105 cells per well and treated with PRO‐6 E (1 μM) for 48 h. Anti‐MET primary Ab (1:1000) was added and incubated at 4°C for 12 h. Protein A/G PLUS‐Agarose (#sc‐2003; Santa Cruz) (25 μl) was added to the mix and incubated at 4°C for 2 h. The samples were collected for western blot analysis. For the inverse‐phase Co‐IP experiment, we undertook Co‐IP assays immunoprecipitating with CRBN and blotting with MET.
2.8. Microscale thermophoresis assay
The labeled MET protein (#10692‐H08H; SinoBiological) was diluted with PBST (0.05% Tween‐20 in PBS), mixed with PRO‐6 E, CRO, or PRO‐6 E‐Me that have been subjected to serial dilution from a 100 μM stock solution, and measured using Monolith NT.115 (NanoTempe).
2.9. Proteomic analysis
Protein digestion and TMT labeling were carried out following the manufacturer's instruction. 30 The labeled peptides were desalinated and fractionated by high pH reversed‐phase liquid chromatography using the 1290 UPLC system (Agilent). The fractions were separated by nanoscale liquid chromatography and analyzed by on‐line electrospray MS/MS. Tandem mass spectra were extracted by Proteome Discoverer software version 2.4.1.15 (Thermo Fisher Scientific). All MS/MS samples were analyzed using SEQUEST, which was set up to search the Uniprot database. The percolator algorithm was used to control peptide level false discovery rates lower than 1%. The normalization of total peptide amount was used to correct experimental deviation.
2.10. SPPIER assay for ternary complex formation
HEK293T cells grown in 35‐mm plates (J04961; Jingan) was transfected with plasmids using Neofect DNA transfection reagent (#TF20121201) for 24 h. The cells were treated with PRO‐6 E or PRO‐6 E‐Me for 48 h. Fluorescence images were captured using a high content analysis system (PerkinElmer).
2.11. Cell cycle analysis
MKN‐45 cells seeded in 6‐well plates (4 × 105 cells/well) were treated with PRO‐6 E for 48 h. The cells were collected, fixed with 70% ethanol, and stained using a cell cycle detection kit (#KGA512). The cells were analyzed by flow cytometry (Beckman Coulter).
2.12. Wound healing assay
Cells (6 × 105 cells/mL) were seeded into 24‐well plates that were coated with collagen from rat tail (#40125ES10; Yeasen). When cell confluence reached approximately 90%, the wells were scratched with a sterile pipet tip and treated with PRO‐6 E as specified. The migration distance was analyzed using ImageJ.
2.13. Transwell invasion assay
MKN‐45 cells were seeded into Matrigel‐coated Transwell upper chambers (#3491; Corning). After the cells were attached, the conditioned media were replaced with serum‐free media, and the chambers were placed into 24‐well plates with 600 μl media containing 20% FBS. Noninvaded cells were removed with a cotton swab, and the invaded cells were fixed with 4% paraformaldehyde and stained with crystal violet. Images were captured and analyzed by Image‐Pro plus 6.0 software (MediaCybernetics).
2.14. Xenograft tumor models
Six‐week‐old female BALB/c nude mice were purchased from Shanghai Model Organisms Center. All studies followed the Institutional Animal Care and Use Committee guidelines of Shanghai Model Organisms Center. MKN‐45 cells (5 × 106 cells) were subcutaneously injected into the right flank of mice. When tumor volume reached approximately 50 mm3, animals were randomly divided into two groups (n = 5). PRO‐6 E was given at 30 mg/kg by oral gavage in 0.5% CMC‐Na (0.9% saline) daily for 15 days. Tumor volume was calculated using the formula (a × b × b)/2, with “a” representing length and “b” representing width. Finally, tumor tissues were collected for western blot, H&E, TUNEL, and Ki‐67 analysis.
2.15. Statistical analysis
Statistical analysis was carried out using a two‐tailed Student's t‐test or one‐way ANOVA followed by Tukey–Kramer multiple comparison test with GraphPad Prism 7. Data are presented as mean ± SD. Statistical significance: *p < 0.05, **p < 0.01, ***p < 0.001.
3. RESULTS
3.1. Design of PROTACs
To investigate the clinical significance of altered MET expression in GC, bioinformatic analysis was undertaken using the GEPIA2 database. 31 The mRNA level of MET in tumor tissues was significantly higher than that in normal tissues, and the level of MET expression correlated with poor prognosis in GC (Figure 1A,B). Immunohistochemical staining of biopsy specimens showed that the content of MET in GC tumor tissues was significantly higher than that in adjacent normal tissues (Figure 1C,D). Collectively, MET represents a potential target for GC.
FIGURE 1.

Design of proteolysis targeting chimeras (PROTACs). (A) MET expression in gastric cancer (GC) using GEPIA2 database. N, normal tissue; T, tumor tissue. (B) Overall survival of GC patients with different MET levels using GEPIA2 database. TPM, transcripts per million. (C) MET expression in GC patients. (D) Analysis of (C) (n = 3). (E, F) Cytotoxic activity of PROTACs for 72 h in MKN‐45 cells. CRBN, cereblon; VHL, von Hippel–Lindau. (G) Cytotoxic activity of GC cells after PRO‐6 E treatment for 48 h. Data are shown as mean ± SD (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001
To design PROTACs that mediate MET degradation, a competitive MET inhibitor, CRO, was selected as the targeting warhead because of its high affinity with MET. 32 In addition, the X‐ray crystal structure of the MET/CRO complex (PDB ID: 2WGJ) identified an available site (the nitrogen atom of piperidine moiety) for extending and connecting with linker. Two members of the E3 ubiquitin ligase complex, CRBN and VHL, have been successfully used to design various PROTACs. 33 , 34 , 35 , 36 , 37 , 38 , 39 Therefore, a set of linkers with different physicochemical properties were designed and connected to the nitrogen atom of CRO piperidine moiety for recruiting CRBN or pVHL (Figure 1E). In MKN‐45 cells, compounds coupling with CRBN showed stronger antiproliferative activities than the pVHL‐based molecules (Figure 1E). In addition, PRO‐6 E (containing flexible aliphatic chain) and 22‐153 (containing rigid quaternary ring) were superior than others. Given the lower molecular weight and bioactivities, the CRBN ligand was selected for constructing MET degraders.
A set of designed PROTACs was synthesized by changing the length of alkane chain in 22‐153, and it was found that 03‐191 had the most pronounced antitumor activity (Figure S1), indicating that the coexistence of a flexible aliphatic chain and rigid four‐member ring (with a total number of nine atoms) might be more conducive to bioactivity (Figure S1). When the position of the rigid ring was changed in 03‐191, the antiproliferative activity of 03‐191 was significantly reduced in 03‐114 (Figure S1), indicating that the position of the ring should be as far away from CRO as possible.
3.2. PRO‐6 E is a potent degrader of MET protein
Based on the cell viabilities in MKN‐45 cells, PRO‐6 E and 03‐191 (IC50 < 1 μM) were selected for further evaluation (Figures 1F and S2A,B). In the MKN‐45 cell‐based protein degradation study, MET protein was degraded with a Dmax of 81.9% when treated with PRO‐6 E (1 μM), in comparison to 03‐191 (Figure S2C,D). Considering the structural difference between PRO‐6 E and 03‐191 (Figure S2 E,F), the rigid ring structure in 03‐191 might have inhibited formation of the ternary complex, which led to poor MET degradation. In addition, two gastric cancer cell lines with high MET expression (SNU‐638) and low MET expression (SNU‐1) were used to test whether the antiproliferative activities of PRO‐6 E are correlated with the levels of MET in GC cells (Figure S2G). The results showed that MKN‐45 cells with the highest MET expression were indeed most sensitive to PRO‐6 E (Figure 1G). Therefore, PRO‐6 E was selected as the degrader for further investigation. The scheme of PRO‐6 E synthesis is shown in Figure 2A.
FIGURE 2.

PRO‐6 E was a potent MET degrader. (A) Synthesis of PRO‐6 E. CRBN, cereblon. (B) Molecular docking results of crizotinib (CRO, blue) or PRO‐6 E (purple) binding to MET (pink, PDB ID: 2WGJ). (C, D) Microscale thermophoresis results for the binding affinity between MET and PRO‐6 E (C) or CRO (D). (E) Cell viability detection when treated with PRO‐6 E or CRO for 48 h. Data are shown as mean ± SD (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001. n.s., no significant difference
Molecular docking was used to determine the binding mode between PRO‐6 E and MET, based on the X‐ray crystal structure of the MET/CRO complex (PDB ID: 2WGJ). The docking study identified two key hydrogen bonds between PRO‐6 E and Pro 1158/Met 1160 in the MET kinase domain (Figure 2B), consistent with the binding between CRO and MET (PDB ID: 2WGJ). The pyrazol ring of PRO‐6 E formed an extra H‐π conjugation with Ile 1084 in MET (Figure 2B), further enhancing the binding efficiency of PRO‐6 E (Kd ≈ 5.38 μM) when compared to CRO (Figure 2C,D). This observation suggests that the construction of PROTAC did not affect the affinity between CRO and MET (Kd ≈ 10.5 μM) (Figure 2D). In the CCK‐8 assay, PRO‐6 E and CRO showed similar antiproliferative activities against MKN‐45 cells for 48 h, with IC50 values of 0.127 μM and 0.106 μM, respectively (Figure 2E).
3.3. PRO‐6 E facilitates MET degradation
In MKN‐45 cells, PRO‐6 E induced MET degradation in a concentration‐dependent manner, with a half‐maximal degradation concentration of 0.203 μM and Dmax of 82.8% (Figure 3A,B). PRO‐6 E‐facilitated MET degradation is also time‐dependent, starting after 12 h of exposure (Figure 3C,D). Immunofluorescence‐based investigation revealed that PRO‐6 E promoted the internalization and degradation of MET protein (Figure 3G,H), consistent with previously reported. 17 To investigate the duration of PRO‐6 E's effect on MET degradation, PRO‐6 E was added to MKN‐45 cells, washed out, and the levels of MET protein monitored over a period of 72 h. The inhibition on MET or p‐MET protein accumulation lasted for at least 72 h after PRO‐6 E removal (Figure 3E,F). To exclude the possibility that “off‐target action” is responsible for PRO‐6 E‐imposed MET inhibition, the effect of PRO‐6 E on a number of newly identified CRBN substrates was examined. 40 , 41 , 42 While PRO‐6 E promoted MET degradation, it only weakly affected IKZF1, IKZF3, and GSPT1 protein levels (Figure S3A,B). Moreover, we used MKN‐45 cells with higher concentrations of PRO‐6 E (5 μM) and observed the hook effect (Figure S3C,D).
FIGURE 3.

PRO‐6 E was a MET degrader. (A) MET expression following PRO‐6 E or crizotinib treatment for 48 h. (B) Analysis of (A) (n = 3). (C) MET expression following PRO‐6 E (1 μM) treatment. (D) Analysis of (C) (n = 3). (E) MET and p‐MET expression. Following PRO‐6 E (1 μM, 48 h) treatment, the compound was removed, and the incubation continued for the specified amount of time. (F) Analysis of (E) (n = 3). (G) MET expression in PRO‐6 E treated (48 h) cells. (H) Internalization and degradation of MET following PRO‐6 E (1 μM) treatment for 48 h. ***p < 0.001. n.s., no significant difference
3.4. PRO‐6 E induces the formation of ternary complex to achieve ubiquitination degradation of MET
To further confirm the formation of a ternary complex induced by PRO‐6 E during MET degradation, we first investigated whether the effect of PRO‐6 E could be competitively antagonized by excessive CRBN ligand or targeting warhead CRO. It was found that MET depletion induced by PRO‐6 E was reversed by excess POM or CRO, whereas POM or CRO alone had no effect on MET expression (Figure 4A,B,D,E), indicating that the degradation effect of PRO‐6 E was dependent on the recruitment of CRBN and MET. Furthermore, in the presence of MG132, a protease inhibitor, the PRO‐6 E‐induced MET degradation was suppressed, indicating the proteasome‐dependent manner (Figure 4C,F). To determine PRO‐6 E's dependence on CRBN in the degradation of MET protein, we then knocked down CRBN using three CRBN siRNA (si‐CRBN) (Figure S4). Knockdown of CRBN significantly alleviated the inhibition effect of PRO‐6 E on MKN45 cells (Figure 4G), indicating the important role of CRBN in PRO‐6 E‐induced antitumor activity. In addition, the E1 inhibitor (MLN7243) or NEDD8 inhibitor (MLN4924) have also been used to further reinforce these claims. Pretreatment with MLN7243 or MLN4924 obviously alleviated the inhibition effect of PRO‐6 E (Figure 4H,I).
FIGURE 4.

PRO‐6 E induced the formation of ternary complexes. MET degradation was abolished when cotreatment with pomalidomide (POM; 15 μM) (A, D) or crizotinib (CRO; 1 μM) (B, E) with PRO‐6 E (1 μM) for 48 h. Pretreatment with MG132 (5 μM, 2 h) (C, F) abolished MET degradation induced by PRO‐6 E (1 μM, 48 h) (n = 3). Pretreatment with cereblon siRNA (si‐CRBN) (G), MLN4924 (0.2 μM, 2 h) (H), or MLN7243 (1 nM, 1 h) (I) abolished the cell viability inhibition induced by PRO‐6 E (0.3 μM, 48 h) (n = 3). (J) Formation of ternary complex following treatment with PRO‐6 E for 48 h. (K) Analysis of (J). (L, M) MKN‐45 cells were treated with PRO‐6 E (1 μM) for 12 h and the ternary complex was detected by co‐immunoprecipitation (IP). (N) Ubiquitination (Ub) of MET following treatment with PRO‐6 E (1 μM). (O) Expression of MET following treatment with PRO‐6 E (1 μM). (P) Ubiquitination of MET after pretreatment with MG132 (5 μM, 2 h) following PRO‐6 E (1 μM, 24 h) incubation. *p < 0.05, **p < 0.01, ***p < 0.001. n.s., no significant difference
Considering that the formation of a ternary complex plays a vital role in PROTAC‐induced degradation, we utilized a GFP phase transition‐based cellular assay named SPPIER, a simple signal pattern, to show in a straightforward and robust manner the protein–protein interaction induced by PRO‐6 E. 43 Based on the principle of SPPIER, we generated two plasmids by genetically fusing HO‐Tag3 to CRBN and HO‐Tag6 to MET. Coexpressing those two plasmids and adding PRO‐6 E in HEK293 cells, we observed the EGFP droplet‐shaped green fluorescence in a background of homogeneous fluorescence, which was weaker at lower concentration (1 μM) and stronger at higher concentration (5 μM) of PRO‐6 E (Figure 4J,K). To facilitate the degradation mechanism of PRO‐6 E, we synthesized PRO‐6 E‐Me. 34 , 44 The microscale thermophoresis assay showed that PRO‐6 E‐Me still had comparable high affinity with MET (Kd ≈ 16.2 μM) (Figure S5C), while the western blot assays showed that PRO‐6 E‐Me had no obvious effects on MET expression when compared to PRO‐6 E (Figure S5D,E,F). The CCK‐8 assays also showed that PRO‐6 E‐Me had a weak antiproliferation effect on tumor cells compared with PRO‐6 E (Figure S5A,B). In addition, the SPPIER technique was applied to visualize PRO‐6 E‐Me‐induced protein–protein interaction. As expected, the green fluorescence droplet formation was not observed in PRO‐6 E‐Me‐treated cells (Figure S5G,H). Taken together, PRO‐6 E formed the MET‐PRO‐6 E‐CRBN ternary complex in the process of MET degradation, which was highly dependent on the E3 ubiquitin ligase CRBN, and the antitumor effect of PRO‐6 E on GC cells is MET degradation‐dependent. We also examined the ability of PRO‐6 E to form a ternary complex with endogenic MET and CRBN in MKN‐45 cells. Immunoprecipitation with anti‐MET and immunoblotting revealed a robust PRO‐6 E‐induced ternary complex formation with endogenous MET protein (Figure 4L,M).
Western blot assays were further used to determine the ubiquitination degradation of MET induced by PRO‐6 E. When the MKN‐45 cells were treated with PRO‐6 E (1 μM) for 12 h, the formation of ubiquitin markers was significantly observed (Figure 4N,O). The absence of ubiquitinated labeled proteins at 24 h could be due to the rapid degradation of ubiquitinated proteins in the proteasome, as previously reported. 45 , 46 Therefore, MG132 was used to observe the ubiquitin proteins. In the presence of MG132, the content of ubiquitin proteins induced by PRO‐6 E increased significantly, supporting a proteasome‐dependent manner (Figure 4P). All of these results indicated that PRO‐6 E hijacked the UPS to facilitate MET degradation. 28
3.5. PRO‐6 E inhibits cell proliferation, migration, and invasion
TMT‐labeled quantitative proteomics analysis was used to determine the effect of PRO‐6 E on the biological function of MKN‐45 cells. Among the approximately 5500 quantified proteins, proteins related to the biological processes and molecular functions of intercellular adhesion and cell proliferation were significantly reduced by more than 2‐fold by PRO‐6 E (Figure 5A), indicating the potential of PRO‐6 E to effectively inhibit tumor cell proliferation and metastasis through MET degradation. Arrested cell cycle progression is an integrated result of impaired cell proliferation and metabolism. 47 We then examined the cell cycle by flow cytometry. As shown in Figure 5B, PRO‐6 E could cause a significant increase in the percentage of G2/M phase cells and decrease in the percentage of S phase cells.
FIGURE 5.

PRO‐6 E inhibited cell growth. (A) Multiple quantitative proteomics analysis of MKN‐45 cells following treatment with PRO‐6 E (1 μM) for 48 h. (B) Cell cycle analysis following treatment with PRO‐6 E for 48 h (n = 3). (C) Migration or invasion assays when cells were treated with PRO‐6 E. (D, E) Analyses of (C) (n = 3). (F, G) Protein expression (F) and analysis (G) when cells were treated with PRO‐6 E for 48 h (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001. n.s., no significant difference
Abnormal activation of MET is usually associated with the promotion of EMT, migration, and invasion effects. 48 , 49 , 50 In the wound healing assay and Transwell invasion assay, the migration and invasion ability of MKN‐45 cells were impaired in a dose‐dependent manner with PRO‐6 E treatment (Figure 5C,E). The detection of key proteins in the EMT process showed that PRO‐6 E could significantly increase the expression of epithelial marker E‐cadherin, accompanied by the decreased mesenchymal markers N‐cadherin and vimentin (Figure S6A,B), resulting in the obvious changes of cell morphology from a dispersed state to a tightly aggregated state (Figure S6C). In addition, PRO‐6 E induced cell cycle arrest in G2 phase of SNU‐638 cells (Figure S7A). We also found that the invasion ability and protein expression of SNU‐638 cells were also impaired with PRO‐6 E treatment (Figure S7B–D).
MET protein relies on its downstream multiple signaling pathways to play a carcinogenic role; of these, the ERK–MAPK pathway is the main mechanism responsible for tumor cell proliferation, cycle progression, and movement. Additionally, it had been proven that the activation of MET could lead to upregulated expression of c‐Myc and p‐ERK in MET‐addicted cells, promoting carcinoma proliferation and moveability. 47 , 51 , 52 The previously mentioned key regulatory proteins were then examined, and treatment with PRO‐6 E could effectively degrade MET protein and reduce the level of key downstream proteins, inducing c‐Myc and p‐ERK (Figure 5F,G).
Taken together, these data indicated that PRO‐6 E was an effective MET degrader with significantly inhibitory effects on c‐Myc and p‐ERK, thus suppressing the infiltration and metastasis of tumor cells.
3.6. PRO‐6 E induces efficient inhibition of tumor growth
To evaluate the antitumor effects and future clinical translation of PRO‐6 E in MKN‐45 cell‐derived xenograft tumor models, we treated the mice with PRO‐6 E at 30 mg/kg/day for 2 weeks with oral administration of CRO (Figure 6A). 53 , 54 , 55 PRO‐6 E could effectively inhibit the tumor growth (Figure 6C–E) without obvious toxicity (Figure 6B). Pathological and immunohistochemical analysis of tumor slices and major organ tissue slices proved that PRO‐6 E could induce tumor necrosis, decrease the ratio of Ki‐67‐positive cells and the expression of MET, and increase the ratio of TUNEL‐positive cells (Figure 6H,I), while no prominent organs damage could be observed (Figure 6J). It was also found that PRO‐6 E could significantly decrease the expression of MET and subsequently downstream proteins, such as c‐Myc and p‐ERK, achieving successful inhibition of tumor growth (Figure 6F,G).
FIGURE 6.

PEO‐6 E inhibited tumor growth in vivo. (A) Schematic diagram. (B, C) Bodyweight (B) and tumor volumes (C) changed in mice (n = 5). (D) Tumor weight (n = 5). (E) Tumor images. (F) Protein expression. (G) Analysis of (F) (n = 3). (H) Typical pictures of H&E, TUNEL, Ki‐67, and MET in tumor tissues. (I) Analysis of (H) (n = 3). (J) Pathological staining of normal tissues. ***p < 0.001
4. DISCUSSION
Dysregulation of MET is associated with tumor invasion, metastasis, recurrence, and poor survival. 56 MET is also one of the most important mechanisms of acquired resistance in multiple RTK targeted therapies, 57 , 58 , 59 , 60 and attracted extensive attention as a proven therapeutic target. A series of Abs and small molecule inhibitors targeting MET have been reported to have excellent antitumor effects in phase I/II clinical trials. However, the evidence of successful phase III clinical trials is still lacking. 15 , 61 , 62 , 63 The current strategy to block the phosphorylation of MET with Abs or inhibitors could not completely suppress the oncogenic function of MET due to its nonkinase functions, resulting in kinase recombination effects, reactivation, 64 , 65 , 66 and subsequent acquired resistance. 56 , 67 , 68 , 69 , 70 , 71 , 72 However, the strategy of using mRNA to interfere with the transcription of MET showed successful inhibiting effects on tumor cells, 73 , 74 , 75 but the poor drug forming properties of MET mRNA agents severely limited its clinical translation.
As a novel drug development strategy, PROTACs could comprehensively inhibit the pathogenic roles of targeted proteins through ubiquitination degradation. Recently, two leading PROTACs, ARV‐110 and ARV‐471, have completed phase II clinical trials with encouraging results. 21 Compared with conventional kinase inhibitors, the “event‐driven” mode and catalytic properties of PROTACs could help to resolve the therapeutic constraints imposed by target protein amplification and mutation. 26 , 37 , 76 Moreover, PROTACs could induce the complete suppression of targeted protein functions through effective degradation. 17 , 77 , 78 , 79 Although resistance to PROTACs has previously been shown to involve genomic alterations of E3 ligase complexes and upregulation of the drug efflux pump MDR1, 80 , 81 the enhanced therapeutic effects of PROTACs could reduce the risks of long‐term and high‐dose induced drug resistance and toxicity. 28 , 29 , 78
Foretinib‐based PROTAC has been developed as a c‐Met degrader in GC cell lines, highlighting the potential advantage of degradation. 17 However, foretinib lacked efficacy in unselected patients with metastatic GC. Additionally, foretinib has not been approved for clinical application, and no further clinical trial has started since 2014, 82 which raises concerns. In contrast, there is no antitumor effect reported in vivo in the previous research of foretinib‐based PROTACs. 17 Herein, we successfully developed a series of crizotinib‐based PROTACs targeting MET protein (Figure 7). Among them, PRO‐6 E showed effective and durable MET degradation, which inhibited downstream c‐Myc and p‐ERK protein expression, thus triggering a great suppression of tumor cell proliferation and motility without obvious cytotoxicity in vitro and in vivo. These findings provided important preliminary data for the further development of oral PROTACs targeting MET, thus paving the way for future clinical treatment of MET‐dependent cancer.
FIGURE 7.

Proteolysis targeting chimera (PROTAC) degrader PRO‐6 E induced degradation of targeted proteins, and led to the suppression of tumor cell proliferation, motility, and invasiveness in MET‐positive gastric cancer models. Ub, ubiquitin
AUTHOR CONTRIBUTIONS
JJC, JMJ, and WJG contributed to data curation, formal analysis, investigation, methodology, writing – original draft, and writing – review and editing. ZZ and HY contributed to data curation, methodology, and software. YDZ, DGN, QLX, and XMZ contributed to data curation and manuscript editing. QYS, YW, WDZ, and XL contributed to conceptualization, resources, supervision, funding acquisition, and writing – review and editing.
CONFLICT OF INTTEREST STATEMENT
None of the authors of this manuscript are current editors or editorial board members of Cancer Science. The authors declare no competing interests.
ETHICS STATEMENT
Approval of the research protocol by an institutional review board: N/A.
Informed consent: N/A.
Registry and registration no. of the study/trial: N/A.
Animal studies: All animal studies followed the Institutional Animal Care and Use Committee guidelines of Shanghai Model Organisms Center.
Supporting information
Figure S1 Chemical structures and cytotoxicity of proteolysis targeting chimeras designed based on 22‐153 and PRO‐6 E in MKN‐45 cells.
Figure S2 Antitumor efficacy of 03‐191 and PRO‐6 E.
Figure S3 Protein detection.
Figure S4 Protein detection of cereblon (CRBN).
Figure S5 Mechanism of action of PRO‐6 E.
Figure S6 Protein expression and cell morphology.
Figure S7 PRO‐6 E inhibited cell cycle, invasion, and protein expression.
Synthetic routes of PROTACs.
ACKNOWLEDGMENTS
This work was supported by the National Natural Science Foundation of China (Nos. 82173846 and 81903654), 2021 Shanghai Science and Technology Innovation Action Plan (No. 21S11902800), Shanghai Science and Technology Development Funds (No. 22QA1409100), Young Talent Lifting Project of China Association of Chinese Medicine (No. CACM‐(2021‐QNRC2‐A09)), Shanghai Rising‐Star Sailing Program (No. 22YF1445000), China Postdoctoral Science Foundation (No. 2021 M702215), Postdoctoral Innovative Talent Support Program (BX20220213), and Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (No. ZYYCXTD‐D‐202004).
Chen J‐J, Jin J‐M, Gu W‐J, et al. Crizotinib‐based proteolysis targeting chimera suppresses gastric cancer by promoting MET degradation. Cancer Sci. 2023;114:1958‐1971. doi: 10.1111/cas.15733
Jin‐Jiao Chen, Jin‐Mei Jin, and Wen‐Jie Gu contributed equally to this work.
Contributor Information
Qing‐Yan Sun, Email: sqy_2000@163.com.
Ye Wu, Email: wuye@shutcm.edu.cn.
Wei‐Dong Zhang, Email: wdzhangy@hotmail.com.
Xin Luan, Email: luanxin@shutcm.edu.cn.
REFERENCES
- 1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. [DOI] [PubMed] [Google Scholar]
- 2. Marano L, Chiari R, Fabozzi A, et al. c‐met targeting in advanced gastric cancer: an open challenge. Cancer Lett. 2015;365(1):30‐36. [DOI] [PubMed] [Google Scholar]
- 3. Van Cutsem E, Sagaert X, Topal B, et al. Gastric cancer. Lancet. 2016;388(10060):2654‐2664. [DOI] [PubMed] [Google Scholar]
- 4. Niclauss N, Gutgemann I, Dohmen J, et al. Novel biomarkers of gastric adenocarcinoma: current research and future perspectives. Cancer. 2021;13(22):5660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Deng N, Goh LK, Wang H, et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co‐occurrence among distinct therapeutic targets. Gut. 2012;61(5):673‐684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Cancer Genome Atlas Research Network . Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513(7517):202‐209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Christensen JG, Burrows J, Salgia R. c‐met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Lett. 2005;225(1):1‐26. [DOI] [PubMed] [Google Scholar]
- 8. Bottaro DP, Rubin JS, Faletto DL, et al. Identification of the hepatocyte growth factor receptor as the c‐met proto‐oncogene product. Science. 1991;251(4995):802‐804. [DOI] [PubMed] [Google Scholar]
- 9. Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell. 2003;3(4):347‐361. [DOI] [PubMed] [Google Scholar]
- 10. Peng Z, Zhu Y, Wang Q, et al. Prognostic significance of MET amplification and expression in gastric cancer: a systematic review with meta‐analysis. PLoS One. 2014;9(1):e84502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Catenacci DV, Ang A, Liao WL, et al. MET tyrosine kinase receptor expression and amplification as prognostic biomarkers of survival in gastroesophageal adenocarcinoma. Cancer. 2017;123(6):1061‐1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Bradley CA, Salto‐Tellez M, Laurent‐Puig P, et al. Targeting c‐MET in gastrointestinal tumours: rationale, opportunities and challenges. Nat Rev Clin Oncol. 2017;14(9):562‐576. [DOI] [PubMed] [Google Scholar]
- 13. Lee HE, Kim MA, Lee HS, et al. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer. 2012;107(2):325‐333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Sun Y, Tian MM, Zhou LX, You WC, Li JY. Value of c‐met for predicting progression of precancerous gastric lesions in rural chinese population. Chin J Cancer Res. 2012;24(1):18‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Tarazona N, Gambardella V, Huerta M, Roselló S, Cervantes A. Personalised treatment in gastric cancer: myth or reality? Curr Oncol Rep. 2016;18(7):41. [DOI] [PubMed] [Google Scholar]
- 16. Oue N, Sentani K, Sakamoto N, Uraoka N, Yasui W. Molecular carcinogenesis of gastric cancer: Lauren classification, mucin phenotype expression, and cancer stem cells. Int J Clin Oncol. 2019;24(7):771‐778. [DOI] [PubMed] [Google Scholar]
- 17. Burslem GM, Smith BE, Lai AC, et al. The advantages of targeted protein degradation over inhibition: an RTK case study. Cell Chem Biol. 2018;25(1):67‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Liu J, Ma J, Liu Y, et al. PROTACs: a novel strategy for cancer therapy. Semin Cancer Biol. 2020;67(Pt 2):171‐179. [DOI] [PubMed] [Google Scholar]
- 19. Jin J, Wu Y, Chen J, et al. The peptide PROTAC modality: a novel strategy for targeted protein ubiquitination. Theranostics. 2020;10(22):10141‐10153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Lin J, Jin J, Shen Y, et al. Emerging protein degradation strategies: expanding the scope to extracellular and membrane proteins. Theranostics. 2021;11(17):8337‐8349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Qi SM, Dong J, Xu ZY, Cheng XD, Zhang WD, Qin JJ. PROTAC: an effective targeted protein degradation strategy for cancer therapy. Front Pharmacol. 2021;12:692574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lai AC, Crews CM. Induced protein degradation: an emerging drug discovery paradigm. Nat Rev Drug Discov. 2017;16(2):101‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Wang Y, Jiang X, Feng F, Liu W, Sun H. Degradation of proteins by PROTACs and other strategies. Acta Pharm Sin B. 2020;10(2):207‐238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Jiang L, Wang Y, Li Q, et al. Design, synthesis, and biological evaluation of Bcr‐Abl PROTACs to overcome T315I mutation. Acta Pharm Sin B. 2021;11(5):1315‐1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Pratz KW, Cortes J, Roboz GJ, et al. A pharmacodynamic study of the FLT3 inhibitor KW‐2449 yields insight into the basis for clinical response. Blood. 2009;113(17):3938‐3946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Sun X, Rao Y. PROTACs as potential therapeutic agents for cancer drug resistance. Biochemistry. 2020;59(3):240‐249. [DOI] [PubMed] [Google Scholar]
- 27. Bondeson DP, Crews CM. Targeted protein degradation by small molecules. Annu Rev Pharmacol Toxicol. 2017;57:107‐123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Bondeson DP, Mares A, Smith IE, et al. Catalytic in vivo protein knockdown by small‐molecule PROTACs. Nat Chem Biol. 2015;11(8):611‐617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Salami J, Crews CM. Waste disposal‐an attractive strategy for cancer therapy. Science. 2017;355(6330):1163‐1167. [DOI] [PubMed] [Google Scholar]
- 30. Li J, Van Vranken JG, Pontano VL, et al. TMTpro reagents: a set of isobaric labeling mass tags enables simultaneous proteome‐wide measurements across 16 samples. Nat Methods. 2020;17(4):399‐404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98‐W102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Zou HY, Li Q, Lee JH, et al. An orally available small‐molecule inhibitor of c‐met, PF‐2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007;67(9):4408‐4417. [DOI] [PubMed] [Google Scholar]
- 33. Khan S, Zhang X, Lv D, et al. A selective BCL‐X(L) PROTAC degrader achieves safe and potent antitumor activity. Nat Med. 2019;25(12):1938‐1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Bai L, Zhou H, Xu R, et al. A potent and selective small‐molecule degrader of STAT3 achieves complete tumor regression in vivo. Cancer Cell. 2019;36(5):498‐511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Wang L, Shao X, Zhong T, et al. Discovery of a first‐in‐class CDK2 selective degrader for AML differentiation therapy. Nat Chem Biol. 2021;17(5):567‐575. [DOI] [PubMed] [Google Scholar]
- 36. Zorba A, Nguyen C, Xu Y, et al. Delineating the role of cooperativity in the design of potent PROTACs for BTK. Proc Natl Acad Sci U S A. 2018;115(31):E7285‐E7292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Posternak G, Tang X, Maisonneuve P, et al. Functional characterization of a PROTAC directed against BRAF mutant V600E. Nat Chem Biol. 2020;16(11):1170‐1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Paiva SL, Crews CM. Targeted protein degradation: elements of PROTAC design. Curr Opin Chem Biol. 2019;50:111‐119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Bekes M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022;21(3):181‐200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Kronke J, Udeshi ND, Narla A, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 2014;343(6168):301‐305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Lu G, Middleton RE, Sun H, et al. The myeloma drug lenalidomide promotes the cereblon‐dependent destruction of Ikaros proteins. Science. 2014;343(6168):305‐309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Matyskiela ME, Lu G, Ito T, et al. A novel cereblon modulator recruits GSPT1 to the CRL4(CRBN) ubiquitin ligase. Nature. 2016;535(7611):252‐257. [DOI] [PubMed] [Google Scholar]
- 43. Chung CI, Zhang Q, Shu X. Dynamic imaging of small molecule induced protein‐protein interactions in living cells with a fluorophore phase transition‐based approach. Anal Chem. 2018;90(24):14287‐14293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Lu J, Qian Y, Altieri M, et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem Biol. 2015;22(6):755‐763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Lu M, Liu T, Jiao Q, et al. Discovery of a Keap1‐dependent peptide PROTAC to knockdown tau by ubiquitination‐proteasome degradation pathway. Eur J Med Chem. 2018;146:251‐259. [DOI] [PubMed] [Google Scholar]
- 46. Li G, Lin SS, Yu ZL, et al. A PARP1 PROTAC as a novel strategy against PARP inhibitor resistance via promotion of ferroptosis in p53‐positive breast cancer. Biochem Pharmacol. 2022;206:115329. [DOI] [PubMed] [Google Scholar]
- 47. Shen A, Wang L, Huang M, et al. c‐Myc alterations confer therapeutic response and acquired resistance to c‐met inhibitors in MET‐addicted cancers. Cancer Res. 2015;75(21):4548‐4559. [DOI] [PubMed] [Google Scholar]
- 48. Wu X, Chen X, Zhou Q, et al. Hepatocyte growth factor activates tumor stromal fibroblasts to promote tumorigenesis in gastric cancer. Cancer Lett. 2013;335(1):128‐135. [DOI] [PubMed] [Google Scholar]
- 49. Taniguchi K, Yonemura Y, Nojima N, et al. The relation between the growth patterns of gastric carcinoma and the expression of hepatocyte growth factor receptor (c‐met), autocrine motility factor receptor, and urokinase‐type plasminogen activator receptor. Cancer. 1998;82(11):2112‐2122. [PubMed] [Google Scholar]
- 50. Toiyama Y, Yasuda H, Saigusa S, et al. Co‐expression of hepatocyte growth factor and c‐met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c‐met signaling in gastric cancer. Int J Cancer. 2012;130(12):2912‐2921. [DOI] [PubMed] [Google Scholar]
- 51. Knudsen BS, Vande WG. Showering c‐MET‐dependent cancers with drugs. Curr Opin Genet Dev. 2008;18(1):87‐96. [DOI] [PubMed] [Google Scholar]
- 52. Sohn SH, Sul HJ, Kim B, Kim BJ, Kim HS, Zang DY. Tepotinib inhibits the epithelial‐mesenchymal transition and tumor growth of gastric cancers by increasing GSK3β, E‐cadherin, and mucin 5AC and 6 levels. Int J Mol Sci. 2020;21(17):6027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Neklesa TK, Winkler JD, Crews CM. Targeted protein degradation by PROTACs. Pharmacol Ther. 2017;174:138‐144. [DOI] [PubMed] [Google Scholar]
- 54. Wang W, Zhou Q, Jiang T, et al. A novel small‐molecule PROTAC selectively promotes tau clearance to improve cognitive functions in Alzheimer‐like models. Theranostics. 2021;11(11):5279‐5295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Sahu A, Prabhash K, Noronha V, Joshi A, Desai S. Crizotinib: a comprehensive review. South Asian J Cancer. 2013;2(2):91‐97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. El DH, El SR, Abdel‐Rahman O. MET inhibitors for the treatment of gastric cancer: What's their potential? J Exp Pharmacol. 2020;12:349‐361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Bardelli A, Corso S, Bertotti A, et al. Amplification of the MET receptor drives resistance to anti‐EGFR therapies in colorectal cancer. Cancer Discov. 2013;3(6):658‐673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Chen CT, Kim H, Liska D, Gao S, Christensen JG, Weiser MR. MET activation mediates resistance to lapatinib inhibition of HER2‐amplified gastric cancer cells. Mol Cancer Ther. 2012;11(3):660‐669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Zhang Z, Wang J, Ji D, et al. Functional genetic approach identifies MET, HER3, IGF1R, INSR pathways as determinants of lapatinib unresponsiveness in HER2‐positive gastric cancer. Clin Cancer Res. 2014;20(17):4559‐4573. [DOI] [PubMed] [Google Scholar]
- 60. Yano S, Wang W, Li Q, et al. Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor‐activating mutations. Cancer Res. 2008;68(22):9479‐9487. [DOI] [PubMed] [Google Scholar]
- 61. Kawakami H, Okamoto I, Arao T, et al. MET amplification as a potential therapeutic target in gastric cancer. Oncotarget. 2013;4(1):9‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Iveson T, Donehower RC, Davidenko I, et al. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first‐line treatment for gastric or oesophagogastric junction adenocarcinoma: an open‐label, dose de‐escalation phase 1b study and a double‐blind, randomised phase 2 study. Lancet Oncol. 2014;15(9):1007‐1018. [DOI] [PubMed] [Google Scholar]
- 63. Catenacci D, Tebbutt NC, Davidenko I, et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first‐line therapy in advanced MET‐positive gastric or gastro‐oesophageal junction cancer (RILOMET‐1): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet Oncol. 2017;18(11):1467‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Hughes VS, Siemann DW. Have clinical trials properly assessed c‐met inhibitors? Trends Cancer. 2018;4(2):94‐97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Zhan H, Tu S, Zhang F, Shao A, Lin J. MicroRNAs and long non‐coding RNAs in c‐met‐regulated cancers. Front Cell Dev Biol. 2020;8:145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Murtuza A, Bulbul A, Shen JP, et al. Novel third‐generation EGFR tyrosine kinase inhibitors and strategies to overcome therapeutic resistance in lung cancer. Cancer Res. 2019;79(4):689‐698. [DOI] [PubMed] [Google Scholar]
- 67. Cepero V, Sierra JR, Corso S, et al. MET and KRAS gene amplification mediates acquired resistance to MET tyrosine kinase inhibitors. Cancer Res. 2010;70(19):7580‐7590. [DOI] [PubMed] [Google Scholar]
- 68. Frigault MM, Markovets A, Nuttall B, et al. Mechanisms of acquired resistance to Savolitinib, a selective MET inhibitor in MET‐amplified gastric cancer. JCO precis. Oncologia. 2020;4:PO.19.00386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kim HS, Chon HJ, Kim H, et al. MET in gastric cancer with liver metastasis: the relationship between MET amplification and met overexpression in primary stomach tumors and liver metastasis. J Surg Oncol. 2018;117(8):1679‐1686. [DOI] [PubMed] [Google Scholar]
- 70. Corso S, Ghiso E, Cepero V, et al. Activation of HER family members in gastric carcinoma cells mediates resistance to MET inhibition. Mol Cancer. 2010;9:121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Mcdermott U, Pusapati RV, Christensen JG, et al. Acquired resistance of non‐small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res. 2010;70(4):1625‐1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Qi J, Mctigue MA, Rogers A, et al. Multiple mutations and bypass mechanisms can contribute to development of acquired resistance to MET inhibitors. Cancer Res. 2011;71(3):1081‐1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Wang XL, Chen XM, Fang JP, Yang CQ. Lentivirus‐mediated RNA silencing of c‐met markedly suppresses peritoneal dissemination of gastric cancer in vitro and in vivo. Acta Pharmacol Sin. 2012;33(4):513‐522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Liu X, Sun R, Chen J, et al. Crosstalk mechanisms between HGF/c‐met axis and ncRNAs in malignancy. Front Cell Dev Biol. 2020;8:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Zheng Z, Yan D, Chen X, et al. MicroRNA‐206: effective inhibition of gastric cancer progression through the c‐met pathway. PLoS One. 2015;10(7):e128751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Toure M, Crews CM. Small‐molecule PROTACS: new approaches to protein degradation. Angew Chem Int Ed. 2016;55(6):1966‐1973. [DOI] [PubMed] [Google Scholar]
- 77. Kurimchak AM, Shelton C, Duncan KE, et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 2016;16(5):1273‐1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Mares A, Miah AH, Smith I, et al. Extended pharmacodynamic responses observed upon PROTAC‐mediated degradation of RIPK2. Commun Biol. 2020;3(1):140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Nalawansha DA, Crews CM. PROTACs: an emerging therapeutic modality in precision medicine. Cell Chem Biol. 2020;27(8):998‐1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Zhang L, Riley‐Gillis B, Vijay P, Shen Y. Acquired resistance to BET‐PROTACs (proteolysis‐targeting chimeras) caused by genomic alterations in core components of E3 ligase complexes. Mol Cancer Ther. 2019;18(7):1302‐1311. [DOI] [PubMed] [Google Scholar]
- 81. Kurimchak AM, Herrera‐Montavez C, Montserrat‐Sangra S, et al. The drug efflux pump MDR1 promotes intrinsic and acquired resistance to PROTACs in cancer cells. Sci Signal. 2022;15(749):eabn2707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Comoglio PM, Trusolino L, Boccaccio C. Known and novel roles of the MET oncogene in cancer: a coherent approach to targeted therapy. Nat Rev Cancer. 2018;18(6):341‐358. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure S1 Chemical structures and cytotoxicity of proteolysis targeting chimeras designed based on 22‐153 and PRO‐6 E in MKN‐45 cells.
Figure S2 Antitumor efficacy of 03‐191 and PRO‐6 E.
Figure S3 Protein detection.
Figure S4 Protein detection of cereblon (CRBN).
Figure S5 Mechanism of action of PRO‐6 E.
Figure S6 Protein expression and cell morphology.
Figure S7 PRO‐6 E inhibited cell cycle, invasion, and protein expression.
Synthetic routes of PROTACs.
