
The tropomyosin receptor kinase (Trk) family consists of three members, TrkA, TrkB, and TrkC, which serve as receptors for neurotrophins (Reichardt, 2006). The neurotrophic receptor kinase (NTRK) genes
Inhibition of TrkA kinase activity is a promising strategy for cancer therapy based on the deregulated activity of TrkA observed in several cancer types. Several TrkA inhibitors have been successfully developed as anticancer drugs. To date, the two Trk inhibitors, larotrectinib and entrectinib, were approved by the FDA in 2018 and 2019, respectively (Cocco
This study aimed to characterize the inhibitory effect of the benzoxazole compound KRC-108 on TrkA kinase and its anti-tumor activities. KRC-108 was previously reported to be an inhibitor of c-Met, Flt3, and ALK (Han
The design and synthesis of KRC-108 (3-(benzo[d]oxazol-2-yl)-5-(1-(piperidin-4-yl)-1H-pyrazol-4-yl)pyridin-2-amine) have been described previously (Cho
A time-resolved fluorescence-based HTRF KinEASE-TK kit (Cisbio, Codolet, France) was used to evaluate TrkA kinase activity. Recombinant proteins containing the TrkA kinase domain were purchased from Invitrogen (Thermo Fisher Scientific, Waltham, MA, USA). The reaction was performed in a 96-well plate with a kinase reaction mixture containing 0.1 μM TK-substrate biotin, 500 μM ATP, and 1 ng of TrkA kinase with a 3-fold serial dilution of the test compound in kinase reaction buffer (50 mM HEPES [pH 7.0], 5 mM MgCl2 1 mM DTT, 0.1 mM orthovanadate, 0.01% bovine serum albumin [BSA], and 0.02% NaN3). After addition of the detection reagents, the TR-FRET signal was measured with a Victor X5 multilabel reader (Perkin Elmer, Waltham, MA, USA) at 615 nm and 665 nm. An equal amount of 1% DMSO was added to each kinase reaction at every dose point. The dose-response curve was fitted by nonlinear regression, and the IC50 was calculated using Prism version 5.01 (GraphPad Software, San Diego, CA, USA).
The human colon cancer cell line KM12C was purchased from the Korean Cell Line Bank (Seoul, Korea). The cells were cultured in minimum essential medium (MEM; Thermo Fisher Scientific) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (Thermo Fisher Scientific) and maintained at 37°C in a humidified atmosphere with 5% CO2.
KM12C cells were plated in a 96-well plate (2,000 cells/well) and incubated with KRC-108 for 72 h at 37°C. Three-fold serial dilutions of KRC-108 were prepared in DMSO to create a 10-point curve at a starting concentration of 10 µM. Cells were treated with DMSO only as a negative control. Cell viability was evaluated using a tetrazolium-based assay with an EZ-Cytox Cell Viability Assay kit (No. EZ-12000; Daeil Lab Service Co. Ltd., Seoul, Korea), according to the manufacturer’s protocol. After incubation for 3 h, the absorbance was measured using a Victor X5 multilabel reader (Perkin Elmer). The 50% cell growth inhibition (GI50) values were calculated using nonlinear regression analysis in GraphPad Prism software 5.01 (GraphPad Software).
KM12C cells were plated in a 6-well plate at a density of 1.2×106 cells/well in 2 mL medium overnight. The monolayer of cells was wounded by scraping with a pipette tip and washed gently with phosphate-buffered saline (PBS) to remove the detached cells. The cells were subsequently treated with the indicated concentrations of KRC-108. Wounds were imaged at 0 and 24 h after the scratch under a light microscope (magnification, ×100; Axiovert 200, Carl Zeiss AG, Jena, Germany). The wound area that recovered over 24 h was measured using ImageJ software (National Institute of Health, Bethesda, MD, USA).
The cells were plated at a density of 2,000 cells/well and allowed to adhere overnight. A concentration range of 0.195-100 µM was used for 5-fluorouracil (5-FU) and KRC-108 was tested in a range of 15 nM to 7.5 μM. The 5-FU and KRC-108 were mixed in a 13.3:1 ratio as calculated by the GI50 ratio of 5-FU (2.90 µM) and KRC-108 (220 nM), and incubated with the cells for 72 h in a humidified chamber at 37°C. After incubation, the EZ-Cytox cell viability assay was performed as described previously. Possible combinations of the two drugs within these concentration ranges were analyzed for any additive, synergistic, or antagonistic effects. The data were analyzed for combination index (Chou and Talalay, 1984) using Compusyn software (version 1.0; ComboSyn Inc., Paramus, NJ, USA).
Cells were lysed in sodium dodecyl sulfate (SDS) lysis buffer, and protein concentrations were measured using the SMART BCA Protein Assay kit (iNtRON Biotechnology, Seongnam, Korea). Samples were resolved by SDS-polyacrylamide gel electrophoresis (10%-15% acrylamide) and transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Next, the membranes were blocked with 5% nonfat milk or 5% BSA and incubated overnight with specific primary antibodies. Antibodies against phospho-TrkA (Tyr490; #9141; 1:1000), cyclin D1 (#2922), phospho-Akt (Ser473; #9271; 1:1000), phospho-p44/42 MAPK (ERK1/2; #4370; 1:1000), phospho-PLCγ1 (Tyr783; #2821; 1:1000), PARP (#9542; 1:1000), and LC3A/B (D3U4C; #12741; 1:1000) were purchased from Cell Signaling Technology (Danvers, MA, USA). Anti-TrkA (sc-7268; 1:1000) and anti-ERK1/2 (sc-135900; 1:1000) were purchased from Santa Cruz Biotechnology (Dallas, TX, USA). β-actin (Sigma-Aldrich, St. Louis, MO, USA, #5441; 1:5000) was used as a loading control. On the following day, the membranes were washed with 1X TBST buffer and incubated in blocking buffer with goat anti-rabbit IgG (Jackson Laboratory, Bar Harbor, ME, USA, 111-035-003; 1:5000) or goat anti-mouse IgG (Jackson Laboratory; 115-035-003; 1:5000) secondary antibodies for 1 h at 22°C. Subsequently, the membranes were washed with 1X TBST buffer, and immunoblot signals were detected using the ECL Select western blotting detection reagent (Amersham ECL Select; GE Healthcare, London, UK).
Cells were plated in 100 mm dish (5×106 cells) and incubated overnight. After incubation, the cells were treated with different concentrations of KRC-108 (0, 0.01, 0.1, 1, or 10 μM) for 24 h. The cells were then detached, fixed, treated with RNase A (50 µg/mL), and stained with propidium iodide (PI; Sigma-Aldrich). Cell cycle distribution was analyzed by flow cytometry using a BD FACSVerseTM flow cytometer (BD, Franklin Lakes, NJ, USA). The percentage of cells in different phases of the cell cycle was determined using BD FACSuiteTM software (BD).
pAdCMV/V5-DEST-GFP-LC3B (Ad-GFP-LC3) is an adenovirus vector constructed to express the GFP-LC3 fusion protein and is used to monitor autophagy flux. The virus particles expressing GFP-LC3 proteins (Ad LC3-GFP) were kindly provided by Dr. Young-Sool Hah (Noh
Animal experiments were approved by the Institutional Animal Care and Use Committee of the Korea Institute of Toxicology and conducted according to the guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care International (approval number 2004-0003). A total of 1×107 KM12C cells were suspended in 200 μl of PBS/matrigel (1:1) and subcutaneously inoculated into the flanks of female BALB/c nu/nu (athymic nude) mice. When the tumor volume reached 100 mm3, the mice were randomly divided into three groups (n=8 each) and administered 40 mg/kg or 80 mg/kg KRC-108 in PBS or pure PBS by oral gavage. The drug or pure PBS was administered daily for 14 days. Tumor sizes were measured twice a week for 14 days, and tumor volumes were calculated using the following formula:
where
First, we tested the TrkA-inhibitory activity of KRC-108. The
KM12C colon cancer cells were subjected to a cell viability assay following treatment with KRC-108. KRC-108 exhibited potent growth inhibitory activity, with a GI50 of 220 nM (Fig. 2A). The effect of KRC-108 on cell growth in combination with 5-FU, a chemotherapeutic agent indicated for colon cancer patients, was investigated (Fig. 2B). First, the ratio of KRC-108 to 5-FU concentration for treatment was determined to be 1:13.3, based on the GI50 values of 5-FU (2.90 μM) and KRC-108 (220 nM). The combination index (CI) was calculated as 0.579 from the cell viability assay of the combination treatment, indicating a synergism between 5-FU and KRC-108 in the combined treatment. These combination treatment experiments showed that the addition of KRC-108 to a 5-FU-based regimen may be suitable for colon cancer therapy.
The effect of KRC-108 on cell migration was assessed using a wound-healing assay. Scratches were made to the cell monolayer, and cells were allowed to migrate into the wounds for 24 h in the absence and presence of KRC-108. In the absence of KRC-108, cells migrated and filled the gap in the scratches, whereas KRC-108 treatment suppressed cell migration in a dose-dependent manner (Fig. 2C). A total of 41.8% of the wound area was recovered following the migration of cells for 24 h without drug treatment. Only 31.2% and 17.2% of the wound area was recovered with treatments of 1 μM and 10 μM KRC-108, respectively.
Given the growth inhibitory effects of KRC-108, we next examined its effects on cell cycle progression. KM12C cells were treated with various concentrations of KRC-108 for 24 h and then subjected to cell cycle analysis. The cell population in the G1 phase increased from 66.6% (DMSO control) to 77.0% (1 μM KRC-108), indicating G1 phase arrest (Fig. 3). Consistent with this cell cycle arrest, the level of cyclin D1 decreased following treatment with KRC-108 concentrations >0.1 μM.
To examine the effect of KRC-108 on apoptosis as a means of suppressing cell growth, PARP cleavage was assessed (Fig. 3C). A slight increase in cleaved PARP was observed in cells following treatment with >1 μM KRC-108. These results indicate that both cell cycle arrest and apoptotic cell death contribute to KRC-108-mediated growth inhibition.
To evaluate the phenotypic changes induced by KRC-108, we analyzed the morphological changes in cells following KRC-108 treatment. As shown in Fig. 4A (right panel), increased vacuole formation was observed in KRC-108-treated cells. These appeared to be autophagic vacuoles and were further confirmed using the LC3 puncta assay and western blotting. LC3 protein, a marker of autophagy, was found to be induced following treatment with 10 μM KRC-108 (Fig. 4B). The formation of LC3 puncta by KRC-108 was also visualized using GFP-LC3 (Fig. 4A). These results suggest that KRC-108 causes an increase in the number of autophagosomes in cells.
Next, TrkA-regulated downstream signaling molecules were examined in the presence of KRC-108 (Fig. 5). Phosphorylated TrkA transduces signals and activates ERK1/2, Akt, and PLCγ via phosphorylation. Phosphorylation of the downstream signaling molecules was reduced following KRC-108 treatment. A slight reduction in phosphorylation level was observed following treatment with 0.1 μM KRC-108, whereas a sharp decrease in phosphorylation was observed in cells treated with 1 μM and 10 μM KRC-108. Phosphorylated PLCγ was completely undetectable in cells treated with 1 μM and 10 μM KRC-108. These results concerning the activation of downstream signaling molecules are consistent with KRC-108-mediated inhibition of TrkA activity, as shown in Fig. 1.
The
Upon completion of treatment, the tumor mass was dissected from
The Trk signaling pathway is a therapeutic target for pain management and cancer (Hirose
KRC-108 was previously reported as a multi-kinase inhibitor, mainly targeting c-Met, Flt3, and ALK (Han
Treatment with KRC-108 induced autophagy, apoptosis, and cell cycle arrest in KM12C cells (Fig. 3, 4). Several kinase inhibitors have been reported to activate autophagy, including imatinib, sorafenib, crizotinib, pazopanib, and sunitinib (Pan
Reports have suggested a relationship between autophagy and TrkA-overexpressing cells. Overexpression of TrkA in neuronal SK-N-MC or non-neuronal USOS cells resulted in cell death associated with autophagy (Dadakhujaev
TrkA transduces signals via Akt, ERK1/2, and PLCγ for neuronal survival and differentiation. Downstream signaling effectors of TrkA fusion proteins appear to be the same as those of the wild-type TrkA protein. In the case of the
The prevalence of NTRK fusion in malignancies has been previously investigated using the Cancer Genome Atlas database (Okamura
The results of this study suggest that KRC-108 is a promising small molecule for therapeutic application in cancers with TrkA fusion. Investigation of TrkA inhibitors such as KRC-108 compound will accelerate advances in tissue-agnostic agents and precision medicine. Identification of TrkA fusions in various types of cancers, and diagnostic techniques for the identification of chromosomal rearrangements and mutations in cancer patients should be accompanied by the development of TrkA-targeting agents.
This research was funded by the National Research Foundation, Government of Korea, grant number 2021R1A2C1007790 (S-Y.H.) and 2021R1A2C2010431 (J.L.).
The authors declare that they have no conflict of interests.
![]() |
![]() |