Purpose: In gastrointestinal stromal tumors (GIST), lipid-metabolizing enzymes remain underexplored, including fatty acid synthase (FASN).

Experimental Design: Forty GISTs were quantitated for FASN mRNA abundance. FASN immunoexpression was informative in 350 GISTs, including 213 with known KIT/PDGFRA/BRAF genotypes. In imatinib-resistant FASN-overexpressing GIST cells, the roles of overexpressed FASN and FASN-targeting C75 in tumor phenotypes, apoptosis and autophagy, KIT transcription, PI3K/AKT/mTOR activation, and imatinib resistance were analyzed by RNAi or myristoylated-AKT transfection. The therapeutic relevance of dual blockade of FASN and KIT was evaluated in vivo.

Results:FASN mRNA abundance significantly increased from very low/low-risk to high-risk levels of NCCN guidelines (P < 0.0001). FASN overexpression was associated with a nongastric location (P = 0.05), unfavorable genotype (P = 0.005), and increased risk level (P < 0.001) and independently predicted shorter disease-free survival (P < 0.001). In vitro, FASN knockdown inhibited cell growth and migration, inactivated the PI3K/AKT/mTOR pathway, and resensitized resistant GIST cells to imatinib. C75 transcriptionally repressed the KIT promoter, downregulated KIT expression and phosphorylation, induced LC3-II and myristoylated AKT-suppressible activity of caspases 3 and 7, attenuated the PI3K/AKT/mTOR/RPS6/4E-BP1 pathway activation, and exhibited dose-dependent therapeutic additivism with imatinib. Compared with both monotherapies, the C75/imatinib combination more effectively suppressed the growth of xenografts, exhibiting decreased KIT phosphorylation, Ki-67, and phosphorylated PI3K/AKT/mTOR levels and increased TUNEL labeling.

Conclusions: We have characterized the prognostic, biological, and therapeutic implications of overexpressed FASN in GISTs. C75 represses KIT transactivation, abrogates PI3K/AKT/mTOR activation, and provides a rationale for dual blockade of KIT and FASN in treating imatinib-resistant GISTs. Clin Cancer Res; 23(16); 4908–18. ©2017 AACR.

Translational Relevance

In this study, genetic and chemical approaches substantiated the critical roles of overexpressed FASN in aggressive GISTs, highlighting its growth-promoting and promigratory attributes, independent negative prognostic impact, and therapeutic implication in sustaining resistance to imatinib. In vitro, C75 transcriptionally repressed KIT and improved the sensitivity of resistant GIST cells toward imatinib therapy. Furthermore, interdependent signaling dynamics may exist among FASN, KIT-elicited signaling output, and the PI3K/AKT/mTOR pathway, coordinately forming a positive regulatory loop. When combined with imatinib, FASN-targeting C75 exhibited a significant growth-inhibiting effect on imatinib-resistant cell lines and derived xenografts. In GISTs, overexpressed FASN may serve as a poor prognosticator, and dual blockade of FASN and KIT emerges as a therapeutic strategy for imatinib-resistant cases.

The majority of gastrointestinal stromal tumors (GIST) harbor mutually exclusive KIT or PDGFRA mutations, which result in constitutive activation of the encoded receptor tyrosine kinases (RTK) to drive tumorigenesis and dictate treatment response to imatinib (1, 2). In adults, 10% to 15% of GISTs exhibit wild-type KIT and PDGFA genes but may bear mutated NF1, BRAF, or SDH genes (3, 4). Both the NIH (Bethesda, MD) and National Comprehensive Cancer Network (NCCN) schemes are deemed effective in risk stratification for GISTs, whereas their prognostic utility is not uniformly validated, and the NCCN scheme even lacks sufficient evidence-based data for some uncommon settings (5, 6). Another challenge in the current care of GIST patients is the inevitable resistance to imatinib that emerges following initial responses due to acquired secondary mutations (1, 7). Therefore, it is desirable to characterize aberrations in nonkinase biochemical pathways that may crosstalk with or regulate RTK-driven signaling to overcome the current limitations in prognosis and drug resistance, particularly for cases refractory to imatinib.

Lipid biosynthesis drastically increases to meet the needs of rapidly proliferating tumor cells (8). Being a multifunctional homodimeric enzyme in de novo lipogenesis, fatty acid synthase (FASN) is frequently overexpressed to catalyze palmitate synthesis to maintain the homeostasis of energy substrates and lipid rafts (9, 10). In most carcinomas, overexpressed FASN promotes aggressive phenotypes, predicts decreased patient survival, and has emerged as an attractive therapeutic target because normal tissues primarily import extracellular lipids without a strict requirement for FASN activity (10). However, the relevance of overexpressed FASN in GISTs remains to be clearly established; to date, only one pertinent study documented that FASN overexpression predominates in high-risk and metastatic GISTs (11).

In this study, mRNA and protein expression levels of FASN significantly increased in higher risk tumors. Furthermore, protein overexpression was strongly associated with unfavorable KIT/PDGFR/BRAF mutation types and independently predicted poor disease-free survival (DFS) in primary imatinib-naïve GISTs. In imatinib-resistant GIST cells, stable silencing against endogenous FASN decreased cell growth and migration and enabled resensitization to imatinib, concomitant with significant suppression of the activity and/or expression of kinases in the PI3K/AKT/mTOR/RPS6/4E-BP1 signaling cascade. Apart from inducing apoptosis and autophagy, FASN inhibition using C75 apparently inhibited total and phosphorylated KIT levels through transcriptional repression and inactivated downstream kinases. Compared with either drug alone, the therapeutic additivism of combined therapy of C75 and imatinib was substantiated in vitro and in vivo, with concomitant downregulation of KIT and the phosphorylated levels of PI3K/AKT/mTOR pathway. Taken together, we robustly characterized FASN as a progression-associated metabolic driver for accurate prognostication in GISTs and provided a rationale for dual blockade of KIT and FASN for nonkinase, metabolic derangement-targeting therapies.

Cell culture

GIST48 and GIST430 cells primarily harbor a homozygous V560D mutation and a heterozygous in-frame deletion in KIT exon 11, respectively. Following imatinib therapy, the GIST48 tumors acquired a heterozygous D820A mutation in KIT exon 17, and the GIST430 tumors developed a heterozygous missense mutation in exon 13 (12). Both cell lines were authenticated by short tandem repeat genotyping, periodically confirmed to be mycoplasma free using PlasmoTest (Invivogen) and maintained in IMDM (Invitrogen) supplemented with 15% FBS, 100 U/mL penicillin/streptomycin, and 4 mmol/L l-glutamine (Invitrogen) at 37°C in 5% CO2 (12, 13).

Tumor cohorts

The Institutional Review Boards approved this study (104-8432C). For IHC, we employed previously constructed tissue microarrays (TMA) of 370 primary GISTs resected before 2009 with triplicate cores in each case (13). Recut TMA sections yielded 350 cases informative for FASN immunoexpression status (Table 1), including 213 successfully genotyped for KIT/PDGFRA/BRAF. Independent of cases in the TMAs, 40 primary GISTs with available frozen samples (Supplementary Table S1) were used to determine FASN mRNA abundance by qRT-PCR. All cases evaluated for immunoexpression and mRNA abundance were imatinib naïve before disease relapse and unrelated to neurofibromatosis type I.

Table 1.

Associations of FASN expression with various clinicopathologic parameters in 350 GIST patients

FASN expression
Low exp.High exp.P
Sex   0.804 
 Male 132 45  
 Female 127 46  
Age (years) 58.9 ± 13.70 60.9 ± 11.70 0.137 
Location   0.050a 
 Gastric 164 47  
 Nongastric 95 44  
Histologic type   0.004a 
 Spindle 207 59  
 Epithelioid & mixed 52 32  
Tumor size (cm)b 5.4 ± 3.85 7.4 ± 4.35 <0.001a 
Mitotic count (50 HPFs)b 6.5 ± 18.94 12.0 ± 26.95 0.001a 
NIH risk   <0.001a 
 Low/very low 110 17  
 Intermediate 89 21  
 High 60 53  
NCCN guideline   <0.001a 
 None/very low 82  
 Low 78 22  
 Moderate 47 18  
 High 52 45  
Mutation type   0.005a 
 Favorable type 83 23  
 Unfavorable type 65 42  
FASN expression
Low exp.High exp.P
Sex   0.804 
 Male 132 45  
 Female 127 46  
Age (years) 58.9 ± 13.70 60.9 ± 11.70 0.137 
Location   0.050a 
 Gastric 164 47  
 Nongastric 95 44  
Histologic type   0.004a 
 Spindle 207 59  
 Epithelioid & mixed 52 32  
Tumor size (cm)b 5.4 ± 3.85 7.4 ± 4.35 <0.001a 
Mitotic count (50 HPFs)b 6.5 ± 18.94 12.0 ± 26.95 0.001a 
NIH risk   <0.001a 
 Low/very low 110 17  
 Intermediate 89 21  
 High 60 53  
NCCN guideline   <0.001a 
 None/very low 82  
 Low 78 22  
 Moderate 47 18  
 High 52 45  
Mutation type   0.005a 
 Favorable type 83 23  
 Unfavorable type 65 42  

Abbreviation: HPF, high power field.

aStatistically significant, P < 0.05.

bWilcoxon rank sum test.

Quantification of FASN and KIT mRNA levels

GIST cell lines stably transduced with shFASN or shLacZ or treated with C75, orlistat, or vehicle were extracted for total RNA using an RNeasy Mini kit (Qiagen). To isolate pure tumor cells from frozen tissues, laser capture microdissection (LCM) was applied on cryomold sections of 40 samples, from which total RNA was extracted as described previously (14). The RT, real-time PCR procedures using an ABI StepOnePlus System (Invitrogen), the reagents of a predesigned TaqMan assay (FASN, KIT, and POLR2A as a housekeeping gene for normalization), and the formulae for calculating FASN and KIT mRNA levels were described in Supplementary Methods S1.

Mutation analysis of KIT, PDGFRA, and BRAF genes

The mutational statuses of KIT exons 11, 9, 13, and 17, as well as of PDGFRA exons 12 and 18, were previously determined for 213 primary GISTs (13, 15). Of these, 22 cases with wild-type KIT and PDGFRA genes were screened for possible mutation in the BRAF hotspot exon 15 (16). The prognostically dichotomized grouping of genotypes is detailed in Supplementary Methods S2 and has been reported previously (13–16).

IHC

TMA sections were microwave heated for antigen retrieval, incubated with a primary antibody against FASN (clone 23, 1:100; BD Transduction), and detected for protein expression using a ChemMate EnVision Kit (Dako). FASN immunoexpression was recorded as the mean percentage of labeled cells in the tumoral cytoplasm, and overexpression was defined by 50% or more tumor cells with moderate or strong cytoplasmic staining. To screen for defects in the mitochondrial succinate dehydrogenase (SDH) complex, 22 GISTs without KIT or PDGFRA mutations in the TMA cohort were evaluated for immunoexpression of SDHA (1:750; Abcam) and SDHB (1:200; Abcam) on full sections (16).

FASN knockdown, myristoylated-AKT transfection, and KIT promoter assay

The methods used to establish stable clones of FASN-silenced GIST48 and GIST430 cells and myristoylated-AKT–transfected GIST430 cells are detailed in Supplementary Methods S3. With pLKO.1-shLacZ (TRCN0000072223) being the control, two lentiviral vectors of pLKO.1-shFASN (Taiwan National RNAi Core Facility), including shFASN#25 (TRCN0000003125) and shFASN#28 (TRCN0000003128), were used to silence FASN expression. The lentiviral vectors harboring constitutively active, myristoylated-AKT and the corresponding EGFP control were purchased from Addgene. The luciferase-tagged pKM2L-phKIT promoter construct was purchased from RIKEN Bioresource Center and transfected into GIST cell lines using Lipofectamine as detailed in Supplementary Methods S4.

Western blots

In two GIST cell lines, Western blots were performed to evaluate endogenous FASN expression, total and phosphorylated KIT, the efficiency of FASN knockdown and myristoylated AKT transfection, and the expression and activity of ERK, PI3K, AKT, and signaling molecules downstream of mTOR following genetic and pharmacologic manipulations, as described in Supplementary Methods S5.

Pharmacologic assays

The FASN inhibitors C75 (Sigma Aldrich) and orlistat (Roche) were obtained and suspended in DMSO. GIST48 and GIST430 cells were seeded in 96-well plates at a density of 5 × 103 cells per well the day before treatment at the indicated time points with vehicle control (0.9% saline), increasing concentrations of imatinib (Novartis) or C75, or varying combinations of imatinib and C75 at the indicated doses.

Functional assays

To evaluate effects associated with FASN knockdown, pharmacologic interventions, or myristoylated-AKT, we analyzed cancer phenotypic alterations using 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT), wound healing, and colorimetric caspase activity assays as appropriate and detailed in Supplementary Methods S6–S8.

Xenograft models

The animal welfare committee approved the experimental protocol (2013092401). GIST430 cells treated with C75 (50 and 100 μmol/L), imatinib (100 mg/kg), a combination of C75 (50 μmol/L) and imatinib (100 mg/kg), or vehicle controls were inoculated into the flanks of 8-week-old SCID mice to analyze the in vivo tumor growth-inhibiting effects of various therapies. Tumor volume was calculated using the following formula: V = π/6 × length (mm) × width (mm)2. Whole sections from formalin-fixed xenograft specimens were analyzed by TUNEL assay, and pertinent antibodies are described in Supplementary Methods S9.

Statistical analysis

For frozen samples, the Kruskal–Wallis and Mann–Whitney U tests were performed to examine differences in FASN mRNA levels between GISTs of various risk categories. In the TMA cohort, we evaluated associations between FASN immunoexpression and clinicopathologic factors using χ2 and Mann–Whitney U tests for categorical and continuous variables, respectively. As of April 2009 (median, 49.9 months; range, 1–247), follow-up data were available for 350 cases informative for FASN immunoexpression. The endpoint was DFS, which would not be confounded by imatinib therapy for relapsed or disseminated diseases. We used the log-rank test to compare univariate prognostic analyses. In multivariate analyses, significant prognosticators with univariate values P < 0.05 were generally introduced, including either the NIH scheme or NCCN guidelines. Size and mitosis, component factors of risk stratification, were not incorporated in the multivariate comparisons. Student t or one-way ANOVA test was used to analyze quantitative RT-PCR and functional or pharmacologic assays for cell and xenograft samples.

FASN mRNA and protein overexpression were associated with unfavorable clinicopathologic factors and poor outcomes

From 40 primary GISTs, their endogenous FASN mRNA levels in LCM-isolated tumor cells were quantified, revealing significant upregulation in higher risk categories defined by both NCCN guidelines (P < 0.0001, Fig. 1A) and the NIH scheme (P < 0.001, Supplementary Fig. S1). In regard to FASN mRNA levels, NCCN guidelines more effectively distinguished GISTs of various risk categories (high risk vs. moderate risk, P = 0.023; moderate risk vs. very low/low risk, P = 0.006). In contrast, no significant difference was observed between low-risk and intermediate-risk cases using the NIH scheme. Consistent with the frequent FASN transcriptional upregulation in carcinomas, FASN overexpression in the progression of GISTs likely occurs in part through transcriptional machinery following altered signaling stimuli (9, 10).

Figure 1.

The clinical relevance of increased FASN mRNA and protein expression in GISTs. A, Quantitative RT-PCR of GISTs demonstrated significantly increased FASN mRNA abundance in higher risk categories using NCCN guidelines (P < 0.0001), which effectively distinguished disparities in expression levels between cases at different risk categories, as indicated by significant P values. B, Gradually increased cellularity and mitosis were observed in representative low-, moderate-, and high-risk GISTs that exhibited correspondingly increased FASN cytoplasmic immunoexpression. C, In log-rank analyses, both FASN overexpression (left) and unfavorable KIT/PDGFR/BRAF genotypes (right) were highly predictive of poor disease-specific survival.

Figure 1.

The clinical relevance of increased FASN mRNA and protein expression in GISTs. A, Quantitative RT-PCR of GISTs demonstrated significantly increased FASN mRNA abundance in higher risk categories using NCCN guidelines (P < 0.0001), which effectively distinguished disparities in expression levels between cases at different risk categories, as indicated by significant P values. B, Gradually increased cellularity and mitosis were observed in representative low-, moderate-, and high-risk GISTs that exhibited correspondingly increased FASN cytoplasmic immunoexpression. C, In log-rank analyses, both FASN overexpression (left) and unfavorable KIT/PDGFR/BRAF genotypes (right) were highly predictive of poor disease-specific survival.

Close modal

To examine the translatability of upregulated mRNA, we analyzed the clinical relevance of overexpressed protein in a large set of primary imatinib-naïve GISTs. There were 350 GISTs informative for FASN immunoexpression status (Table 1) with clinical follow-up data, which comprised 88 no- or very low-risk cases, 100 low-risk cases, 65 moderate-risk cases, and 97 high-risk cases based on NCCN guidelines and corresponded to 127 very low/low-risk cases, 110 intermediate-risk cases, and 113 high-risk cases according to the NIH risk scheme. Of these, 22 cases with wild-type KIT and PDGFRA genes exhibited neither a BRAF hotspot mutation nor aberrant loss of SDHA or SDHB (16). FASN-overexpressing GISTs were frequently nongastric (P = 0.050) and were strongly associated with the presence of epithelioid cells, unfavorable KIT/PDGFR/BRAF genotypes, and increasing tumor size, mitotic rate, and risk levels by both NCCN and NIH criteria (all P ≤ 0.005). Univariately, poor DFS (Table 2; Fig. 1C) was associated with nongastric locations (P = 0.0023), unfavorable genotypes (P = 0.0005), epithelioid histology, increases in tumor size, mitosis, NCCN risk levels, and FASN overexpression (all P < 0.0001). In multivariate analysis, FASN overexpression (P < 0.001; HR, 2.448) independently predicted poor outcomes, along with higher NCCN risk levels (P < 0.001) and epithelioid histology (P = 0.007). However, unfavorable genotypes lost prognostic independence. Analogous to the NCCN guidelines, the alternative incorporation of NIH criteria in multivariate analysis demonstrated very similar influence and statistical significance (Supplementary Table S2). Specifically, FASN overexpression (P = 0.002; HR, 2.253) remained independent as an adverse prognosticator, together with higher NIH risk levels (P < 0.001) and epithelioid histology (P = 0.010).

Table 2.

Univariate and multivariate analyses for DFS according to FASN expression status, NCCN guidelines, and other prognostic factors

Univariate analysisMultivariate analysis
ParametersCases (n)Events (n)PHR (95% CI)P
Sex   0.4667   
 Male 177 43    
 Female 173 44    
Age (years)   0.0584   
 <70 259 59    
 ≥70 91 28    
Location   0.0023a  0.954 
 Gastric 211 40  1 (—)  
 Nongastric 139 47  1.015 (0.621–1.659)  
Histologic type   <0.0001a  0.007a 
 Spindle 266 51  1 (—)  
 Mixed/epithelioid 84 36  2.013 (1.213–3.340)  
Tumor size (cm)b   <0.0001a   
 ≤5 cm 161 16    
 >5; ≤10 cm 131 38    
 >10 cm 58 33    
Mitotic count (50 HPFs)b   <0.0001a   
 0–5 249 33    
 6–10 43 14    
 >10 58 40    
NCCN guideline   <0.0001a  <0.001a 
 None/very low 88  1 (—)  
 Low 100 10  3.773 (1.919–7.407)  
 Moderate 65 15  3.773 (1.783–7.937)  
 High 97 59  7.246 (1.653–31.25)  
Mutation type   0.0005a  0.142 
 Favorable type 106 22  1 (—)  
 Unfavorable type 107 45  1.480 (0.877–2.498)  
FASN expression   <0.0001a  <0.001a 
 Low expression 259 41  1 (—)  
 High expression 91 46  2.448 (1.482–4.043)  
Univariate analysisMultivariate analysis
ParametersCases (n)Events (n)PHR (95% CI)P
Sex   0.4667   
 Male 177 43    
 Female 173 44    
Age (years)   0.0584   
 <70 259 59    
 ≥70 91 28    
Location   0.0023a  0.954 
 Gastric 211 40  1 (—)  
 Nongastric 139 47  1.015 (0.621–1.659)  
Histologic type   <0.0001a  0.007a 
 Spindle 266 51  1 (—)  
 Mixed/epithelioid 84 36  2.013 (1.213–3.340)  
Tumor size (cm)b   <0.0001a   
 ≤5 cm 161 16    
 >5; ≤10 cm 131 38    
 >10 cm 58 33    
Mitotic count (50 HPFs)b   <0.0001a   
 0–5 249 33    
 6–10 43 14    
 >10 58 40    
NCCN guideline   <0.0001a  <0.001a 
 None/very low 88  1 (—)  
 Low 100 10  3.773 (1.919–7.407)  
 Moderate 65 15  3.773 (1.783–7.937)  
 High 97 59  7.246 (1.653–31.25)  
Mutation type   0.0005a  0.142 
 Favorable type 106 22  1 (—)  
 Unfavorable type 107 45  1.480 (0.877–2.498)  
FASN expression   <0.0001a  <0.001a 
 Low expression 259 41  1 (—)  
 High expression 91 46  2.448 (1.482–4.043)  

Abbreviation: HPF, high power field.

aStatistically significant, P < 0.05.

bTumor size and mitotic activity were not introduced in multivariate analysis, as these two parameters were component factors of NCCN risk scheme.

FASN knockdown attenuated cell growth, migration, imatinib resistance, and PI3K/mTOR/AKT activation

Given the challenge of imatinib resistance in GISTs (1), we focused on imatinib-resistant GIST48 and GIST430 cell lines to characterize the biological implications of FASN. As both cell lines overexpressed FASN, RNAi was applied, and two shFASN clones achieved stable silencing with different efficiency (Fig. 2A). Compared with shLacZ, shFASN significantly decreased viable cells in both GIST430 and GIST48 cells (Supplementary Fig. S2), consistent with the known growth-promoting function of FASN (9–11). Moreover, a wound healing assay exhibited significantly less migrating cells when FASN was silenced, indicating that FASN has a promigratory attribute (Fig. 2B; Supplementary Fig. S3). Depending on the efficiency of shFASN clones, FASN silencing resulted in a varying but significantly increased percentage of nonviable GIST48 and GIST430 cells treated with imatinib (5–1,000 nmol/L; Fig. 2C), implying that suppression of FASN expression may reduce imatinib resistance. Accumulating evidence has revealed that the PI3K/AKT/mTOR pathway plays important roles in relaying RTK signaling in GISTs and neoplastic lipogenesis of other cancers (12, 17). Therefore, we explored the potential regulatory basis of these kinases linked to overexpression of FASN in GIST cell models, demonstrating that phosphorylated forms of PI3K, AKT, and mTOR were all prominently downregulated by FASN knockdown (Fig. 2D). However, the expression and phosphorylation of KIT protein did not exhibit significantly altered levels in FASN-knockdown GIST48 and GIST430 cells (Fig. 2D), nor did fold changes of KIT mRNA expression and promoter activity as measured by qRT-PCR and luciferase-constructed reporter assays, respectively (Supplementary Fig. S4).

Figure 2.

FASN knockdown attenuated cell migration, imatinib resistance, and PI3K/AKT/mTOR signaling activity. A, GIST48 (left) and GIST430 (right) cell lines were FASN-overexpressing and knocked down by lentiviral vectors bearing shFASN#25 or shFASN#28 with different sequences. The silencing was confirmed by qRT-PCR and Western blots, with better efficiency seen for shFASN#28. The shLacZ plasmid, POLR2A transcript, and GAPDH protein were utilized as controls in RNAi, qRT-PCR, and Western blots, respectively. B, The percentages of wound closure were plotted, demonstrating that cell migration was significantly impaired by shFASN#25 and shFASN#28 in both cell lines. C, In XTT assays, shFASN#25 or shFASN#28, compared with shLacZ, significantly reduced viable cells in both GIST cell lines exposed to imatinib at 100 nmol/L. D, In both cell lines, shFASN#25 and shFASN#28 consistently reduced the phosphorylation levels of PI3K, AKT, and mTOR, whereas their total forms were varyingly affected. Notably, the level of total KIT remained constant, and phosphorylated KIT only slightly declined in GIST48 cells transduced with shFASN#28.

Figure 2.

FASN knockdown attenuated cell migration, imatinib resistance, and PI3K/AKT/mTOR signaling activity. A, GIST48 (left) and GIST430 (right) cell lines were FASN-overexpressing and knocked down by lentiviral vectors bearing shFASN#25 or shFASN#28 with different sequences. The silencing was confirmed by qRT-PCR and Western blots, with better efficiency seen for shFASN#28. The shLacZ plasmid, POLR2A transcript, and GAPDH protein were utilized as controls in RNAi, qRT-PCR, and Western blots, respectively. B, The percentages of wound closure were plotted, demonstrating that cell migration was significantly impaired by shFASN#25 and shFASN#28 in both cell lines. C, In XTT assays, shFASN#25 or shFASN#28, compared with shLacZ, significantly reduced viable cells in both GIST cell lines exposed to imatinib at 100 nmol/L. D, In both cell lines, shFASN#25 and shFASN#28 consistently reduced the phosphorylation levels of PI3K, AKT, and mTOR, whereas their total forms were varyingly affected. Notably, the level of total KIT remained constant, and phosphorylated KIT only slightly declined in GIST48 cells transduced with shFASN#28.

Close modal

C75 inhibited viability, exhibited additivism with imatinib, and induced apoptosis and autophagy in vitro

A synthetic analogue of cerulenin, C75, has been shown to potently inhibit FASN and suppress tumor growth of various human cancers in vitro and in vivo (9). However, the therapeutic potential and molecular underpinning of C75 remains unexplored in GISTS, especially in regard to crosstalk with KIT-elicited signal transduction. As measured by XTT assay, C75 had a deleterious effect on the survival of both cell lines, and the IC50 values, close to that reported in lung adenocarcinoma cells (18), were between 60 and 70 μmol/L (Fig. 3A, top). Another FASN inhibitor, orlistat, also dose dependently suppressed the cell viability of both GIST cell lines, although its potency was inferior to C75 (Supplementary Fig. S5). Accordingly, we selected C75 for further assessment of its potential efficacy in combination with imatinib. All four conditions of imatinib (100 and 200 nmol/L) and C75 (25 and 50 μmol/L) combination significantly improved the viability-suppressing capability in imatinib-resistant cell lines in a generally dose-dependent manner (Fig. 3A, bottom). This finding indicated an additive effect between C75 and imatinib and the need for validating the potential of a dual blockade of KIT and FASN in vivo (see below). To decipher the basis of C75 in growth inhibition of imatinib-resistant GIST cells, we examined whether C75 could induce cellular apoptosis, which was validated by significantly increased caspases 3 and 7 activity from 48 hours onward or by induced cleaved caspase-3 in GIST48 and GIST430 cells treated with C75 at both 50 and 100 μmol/L (Fig. 3B; Supplementary Fig. S6). In both GIST cell models, C75 at 50 μmol/L, similar to imatinib, also increased expression of the LC3-II isoform, indicating the role of C75 in triggering autophagy, and C75 at 100 μmol/L induced LC3-II more obviously than imatinib. Compared with monotherapy, GIST48 cells exhibited further increased levels of cleaved caspase-3 and LC3-II when treated with imatinib and 100 μmol/L C75. In contrast, GIST430 cells more readily showed additional increases of LC3-II when combing imatinib with C75 at either 50 or 100 μmol/L, albeit no apparent additive effect on induction of apoptosis. These findings implied an existent but variable additive effect of C75/imatinib combination on the induction of cellular autophagy and apoptosis in imatinib-resistant GIST cells (Supplementary Fig. S6).

Figure 3.

In vitro effects and mechanisms of C75 monotherapy versus C75/imatinib combination therapy. A, C75/imatinib combination therapy exhibited therapeutic additivism. Top, at the indicated doses, C75 monotherapy for 72 hours dose dependently decreased viable cells of GIST48 and GIST 430 cell lines; bottom, compared with imatinib at 100 or 200 nmol/L alone, both cell lines exhibited significantly decreased cell viability when combined with C75 at either 25 or 50 μmol/L. *, P < 0.05 for all effective combinations. B, C75 induced cellular apoptosis and autophagy. Top, in colorimetric quantitation, activated caspases 3 and 7 were significantly induced in both cell lines 48 hours after exposure to C75 at 50 and 100 μmol/L concentrations, respectively. *, P < 0.05. Bottom, upon exposure to C75 at 100 μmol/L for 48 or 72 hours, both cell lines exhibited significantly increased LC3-II isoform. C, In both cell lines, C75 consistently suppressed the activities of KIT and the PI3K/AKT/mTOR/RPS6/4E-BP1 cascade. The decreased phosphorylation of these kinases, including KIT, mostly occurred as early as 24 hours after treatment, except that RPS6 became inactivated at 72 hours. Notably, total KIT expression in both cell lines was concomitantly downregulated, attributable to transcriptional repression of the KIT promoter (Supplementary Figs. S5 and S6). Regarding expression levels and/or time of occurrence, the total forms of other kinases within the PI3K/AKT/mTOR cascade were varyingly affected by C75. D, In both cell lines, C75/imatinib combination treatment generally outperformed imatinib or C75 monotherapy in suppressing the activation of KIT, AKT, and mTOR. Compared with C75 monotherapy, the superiority of the combination therapy in inhibiting activated RPS6 and 4E-BP1 was only apparent in GIST48 cells. Paradoxically, the activation of ERK was induced by C75 or C75/imatinib combination therapy.

Figure 3.

In vitro effects and mechanisms of C75 monotherapy versus C75/imatinib combination therapy. A, C75/imatinib combination therapy exhibited therapeutic additivism. Top, at the indicated doses, C75 monotherapy for 72 hours dose dependently decreased viable cells of GIST48 and GIST 430 cell lines; bottom, compared with imatinib at 100 or 200 nmol/L alone, both cell lines exhibited significantly decreased cell viability when combined with C75 at either 25 or 50 μmol/L. *, P < 0.05 for all effective combinations. B, C75 induced cellular apoptosis and autophagy. Top, in colorimetric quantitation, activated caspases 3 and 7 were significantly induced in both cell lines 48 hours after exposure to C75 at 50 and 100 μmol/L concentrations, respectively. *, P < 0.05. Bottom, upon exposure to C75 at 100 μmol/L for 48 or 72 hours, both cell lines exhibited significantly increased LC3-II isoform. C, In both cell lines, C75 consistently suppressed the activities of KIT and the PI3K/AKT/mTOR/RPS6/4E-BP1 cascade. The decreased phosphorylation of these kinases, including KIT, mostly occurred as early as 24 hours after treatment, except that RPS6 became inactivated at 72 hours. Notably, total KIT expression in both cell lines was concomitantly downregulated, attributable to transcriptional repression of the KIT promoter (Supplementary Figs. S5 and S6). Regarding expression levels and/or time of occurrence, the total forms of other kinases within the PI3K/AKT/mTOR cascade were varyingly affected by C75. D, In both cell lines, C75/imatinib combination treatment generally outperformed imatinib or C75 monotherapy in suppressing the activation of KIT, AKT, and mTOR. Compared with C75 monotherapy, the superiority of the combination therapy in inhibiting activated RPS6 and 4E-BP1 was only apparent in GIST48 cells. Paradoxically, the activation of ERK was induced by C75 or C75/imatinib combination therapy.

Close modal

C75 repressed KIT transactivation and the C75/imatinib combination abrogated KIT and PI3K/mTOR/AKT signaling activities more potently than monotherapies

Unlike the observed RNAi results, total and phosphorylated KIT levels apparently diminished in C75-treated imatinib-resistant GIST cells (Fig. 3C), which were in line with significantly decreased KIT mRNA levels (Supplementary Fig. S7) and promoter activity (Supplementary Fig. S8). Similarly, orlistat induced significant repression in the KIT promoter activity in both cell lines, along with resultant decreases at the levels of mRNA, protein expression, and phosphorylation (Supplementary Figs. S7–S9). These findings indicate that transcriptional repression, at least partly, implicates the KIT-repressing effect of pharmacologic inhibition by C75, which became more evident when combined with imatinib (Fig. 3D; Supplementary Fig. S10). Furthermore, treatment with C75 or imatinib individually or in combination was examined to determine the modulatory effect on KIT phosphorylation and PI3K/AKT/mTOR signaling activity. Consistent with FASN silencing, FASN-targeting C75 abrogated the activation of PI3K, AKT, mTOR and, conceivably, their downstream kinases RPS6 and 4E-BP1 (Fig. 3C), with relatively slight or no decreases in the expression of the total forms of individual kinases. Generally, similar to C75, orlistat treatment inhibited the expression and activation of KIT as well as on downstream kinases of the AKT/mTOR signaling cascade in both GIST48 and GIST 430 cells (Supplementary Figs. S9 and S10). Paradoxically, in orlistat-treated GIST48 cells, the level of phosphorylated AKT increased between 24 and 48 hours but abated to a barely detectable level at 72 hours.

Compared with monotherapies or vehicle control, the C75/imatinib combination more effectively inactivated the kinases of the AKT/mTOR signaling cascade, except for phosphorylated RPS6 and 4E-BP1 in GIST430 cells. Instead, the activation status of ERK was accentuated by C75 and even more clearly by the C75/imatinib combination in both cell lines (Fig. 3D). Given the known antiapoptotic role of AKT (17), we transfected cells with myristoylated-AKT to elucidate its effect on FASN inhibition–induced apoptosis in GIST430 cells, which had more obvious C75-inactivated AKT than the GIST48 cells (Fig. 3C). The GIST430 cells with myristoylated-AKT exhibited significantly reduced activity of caspases 3 and 7 triggered by C75 (Supplementary Fig. S11A), implying a potential link between AKT inactivation and the C75-induced cytotoxicity. Moreover, myristoylated-AKT transfection increased the phosphorylated mTOR, RPS6, and 4E-BP1 and partially rescued the activities of upstream KIT and PI3K reduced by C75 (Supplementary Fig. S11B).

C75 monotherapy versus C75/imatinib combined therapy in vivo

Given the additivism between C75 and imatinib, a GIST430-derived xenograft model was established to analyze the in vivo therapeutic efficacy and antitumor mechanism of C75 and C75/imatinib combination therapy. The in vivo tumor-inhibiting effect of C75 was significant at both 50 and 100 μmol/L compared with the control group, although no dose-dependent difference was seen between the two doses tested on gross examination (Supplementary Fig. S12A). However, the group receiving 100 μmol/L C75 exhibited broader hyalinized stromal changes with fewer viable cells in the corresponding xenografts (Supplementary Fig. S12B). Next, we evaluated the therapeutic efficacy of the C75/imatinib combination, which from day 21 onward exhibited more prominent growth-suppressing capability than either C75 or imatinib alone (Fig. 4A), corroborating the additivism of the combined therapy in vitro. Notably, the gross fibrotic regression was only present in the xenograft specimens receiving combined therapy, and neither significant body weight loss nor impaired function of vital organs was observed in the treated mice. Microscopically, the PBS-treated xenografts displayed cellular proliferation of epithelioid to spindle cells without necrosis or fibrosis. Tumor cells of the control group exhibited diffuse and strong expression of p-cKIT, p-AKT, p-mTOR, pRPS6, and p-4E-BP1, high Ki-67 proliferative index, and few TUNEL-positive apoptotic cells. In contrast, the xenografts receiving combined therapy displayed remarkably lower cellularity and mitosis, prominent stromal fibrosis, and overt decreases in the levels of p-cKIT and phosphorylated kinases downstream of the AKT/mTOR pathway (all P < 0.001; Fig. 4B). However, these alterations in histomorphology, kinase expression and activity, and proliferative and apoptotic markers were mild to moderate in the monotherapy groups receiving imatinib or C75 alone.

Figure 4.

The in vivo therapeutic effect and mechanism of C75/imatinib combination treatment in GIST430-derived xenografts. A, Top, the tumor growth curves demonstrated that combined therapy with 50 μmol/L C75 and 100 mg/kg imatinib more significantly reduced the average tumor volume of xenografts than the PBS control (P < 0.001) or monotherapy with either C75 (P = 0.002) or imatinib (P = 0.002). Compared with the PBS controls, treatment with C75 (P = 0.001) or imatinib (P = 0.020) alone also resulted in significantly smaller tumors. Bottom, after sacrifice on day 28, images of GIST430 xenografts receiving various therapies illustrated the efficacy of combined therapy, with gross fibrotic regression only observed in this group. B, Microscopically, the representative xenografts exhibited the highest proportion of fibrotic regression to viable tumor cells, the most TUNEL-labeled apoptotic cells, and the fewest Ki67-labeled proliferative cells in the C75/imatinib combination group, followed by the C75 or imatinib monotherapy and then by the PBS control. The C75/imatinib combination group and the control group had the scarcest and the most copious expression of phosphorylated c-KIT (p-cKIT), respectively. Analogously, various groups correspondingly exhibited different expression patterns of phospho-AKT, phospho-mTOR, phospho-RPS6, and phospho-4EBP1, all of which were most prominently decreased in those receiving combined therapy.

Figure 4.

The in vivo therapeutic effect and mechanism of C75/imatinib combination treatment in GIST430-derived xenografts. A, Top, the tumor growth curves demonstrated that combined therapy with 50 μmol/L C75 and 100 mg/kg imatinib more significantly reduced the average tumor volume of xenografts than the PBS control (P < 0.001) or monotherapy with either C75 (P = 0.002) or imatinib (P = 0.002). Compared with the PBS controls, treatment with C75 (P = 0.001) or imatinib (P = 0.020) alone also resulted in significantly smaller tumors. Bottom, after sacrifice on day 28, images of GIST430 xenografts receiving various therapies illustrated the efficacy of combined therapy, with gross fibrotic regression only observed in this group. B, Microscopically, the representative xenografts exhibited the highest proportion of fibrotic regression to viable tumor cells, the most TUNEL-labeled apoptotic cells, and the fewest Ki67-labeled proliferative cells in the C75/imatinib combination group, followed by the C75 or imatinib monotherapy and then by the PBS control. The C75/imatinib combination group and the control group had the scarcest and the most copious expression of phosphorylated c-KIT (p-cKIT), respectively. Analogously, various groups correspondingly exhibited different expression patterns of phospho-AKT, phospho-mTOR, phospho-RPS6, and phospho-4EBP1, all of which were most prominently decreased in those receiving combined therapy.

Close modal

FASN, an enzyme critical for endogenous lipogenesis, catalyzes the condensation of malonyl-CoA and acetyl-CoA to synthesize palmitate de novo and converts excess carbon intake into long-chain fatty acids for storage (8, 10). In various carcinomas and their preinvasive precursors, the expression level of FASN is selectively increased and frequently associated with shorter DFS (10). Specifically, for GISTs, only one prior study on FASN reported its preferential overexpression in high-risk and metastatic tumors and confers a proproliferative function by upregulating cyclin A (11). In this study, GIST tissues exhibited a positive correlation between FASN mRNA abundance and risk levels, being statistically more robust when defined by NCCN guidelines than by NIH criteria. Although this observation suggested possible implications of different stratification criteria in the correlation with FASN mRNA expression, it indicated that FASN overexpression in GISTs is, at least in part, transcriptionally regulated. In prostate and breast cancers, FASN mRNA is known to be transcriptionally upregulated by activated AKT, which enhances the binding of sterol regulatory element–binding protein (SREBP)-1c to the FASN promoter (10, 19). The strong association between FASN overexpression and adverse clinicopathologic variables, such as higher risk levels, was also validated in our large cohort of GISTs. Moreover, we confirmed that FASN overexpression not only correlated with unfavorable genotypes but also conferred an independent negative prognostic impact, with a risk of shorter DFS that was two times higher.

Despite being complex, the molecular underpinning of FASN oncogenicity has been linked to its versatile roles in evading apoptosis, maintaining lipid rafts, and activating central oncogenic pathways, such as the PIK3/AKT/mTOR signaling cascade (9, 10). The biological implications of overexpressed FASN in GISTs were supported by FASN-silenced cell lines that demonstrated decreased cell growth and migration. Although overexpressed FASN has emerged as a potential therapeutic target in various cancer types (9, 20), knowledge about the biology of FASN inhibition in GIST cell and xenograft models is scant. In RTK-targeted therapies, such as imatinib targeting of mutant KIT in GISTs, acquired resistance frequently follows a short-lived treatment response (1, 7). This predicament urges a fundamental elucidation of interdependent KIT-eliciting downstream and collateral pathways essential for sustaining resistance and survival of tumor cells to develop novel therapeutic strategies. In GIST cells harboring refractory mutations, FASN knockdown indeed inhibited the activity of the PI3K/AKT/mTOR axis and downstream RPS6 and 4E-BP1 kinases and notably increased the susceptibility to imatinib. These results highlighted the therapeutic relevance of overexpressed FASN in imatinib-resistant GISTs and the potential of drug combinations for dual blockade of FASN and KIT.

Consequently inducing cellular apoptosis, the chemical anticancer mechanisms of C75 involve abrogation of de novo fatty acid synthesis, toxicity of accumulated malonyl-CoA, and increased reactive oxygen species associated with dissipated mitochondrial membrane potential (21–23). Similar to previously reported tumor types, the apoptosis-inducing effect of pharmacologic FASN inhibition was reproduced in GISTs (9, 24, 25). In imatinib-resistant GIST cells and xenografts, we observed significant additive efficacy with C75/imatinib combination therapy. Exposure to C75 alone or the C75/imatinib combination led to abrogation of AKT activity concomitant with increased caspase activity in cell lines and an increased TUNEL-labeling index in xenografts. In this context, C75-induced apoptosis in GIST cells is potentially linked to AKT inactivation, given that FASN inhibition may perturb membranous lipid rafts needed for the proper subcellular localization of proteins involved in signal transduction, including the antiapoptotic PI3K/AKT/mTOR pathway (17, 20). Moreover, transfection of C75-treated GIST cells with myristoylated-AKT counteracted the activity of caspases 3 and 7 and reactivated not only downstream mTOR, RPS6, and 4E-BP1 but also upstream KIT and PI3K. However, it should be added that the adverse weight-reducing side effect of C75 compromises its clinical utility as a cancer therapeutic (10).

Imatinib, which binds directly to the KIT receptor, was shown to have no appreciable effect on the KIT promoter (26). In contrast, C75 repressed KIT transactivation in imatinib-resistant GIST cells and eventually caused a prominent loss of total KIT protein that further exhausted available receptors for sustaining phosphorylated activation. These findings resonated with the well-characterized example of FASN inhibitors in alleviating the resistance to HER2-targeted therapy in breast carcinomas, given that C75 can inhibit the HER2 gene promoter through upregulation of the PEA3 transcriptional repressor (27, 28). Unlike C75 and orlistat, FASN knockdown did not significantly repress KIT at all levels of transcription, expression, and activity in imatinib-resistant GIST cell models. This discrepancy may pose a query whether C75 exerted a KIT promoter–repressing effect through an FASN-independent mechanism or it indirectly impeded the KIT signaling output through inhibiting docking proteins that interact with activated KIT. Considering the association of FASN overexpression with unfavorable RTK genotypes in GIST samples and the improved sensitivity toward imatinib by FASN knockdown, an interdependent positive feedback loop appears conceivable, which coordinately regulates FASN, KIT-elicited signaling output, and the PI3K/AKT/mTOR signaling pathway. Relevantly, FASN is required to sustain the level of diacylglycerol in the lipid rafts involving the recruitment of KIT-interacting docking proteins, such as protein kinase C (PKC) and Grb2 (29, 30). More intriguingly, PKC-θ isoform has been shown to mediate sustained mutant KIT activation in the Golgi complex in GISTs (31). In this context, both generalized inhibition by C75 and the resultant attenuation of KIT signaling relay might contribute to the inactivation of PI3K/AKT/mTOR and downstream kinases, with or without decreased expression of their total proteins. Collectively, FASN inhibition resulting from combined therapy minimized KIT dependency and improved the sensitivity to imatinib in resistant GISTs in vitro and in vivo.

Accumulating evidence indicates that apoptosis and autophagy share common regulatory elements within oncogenic pathways activated by RTKs, including the PI3K/AKT/mTOR pathway (32, 33). In GIST cells, the endogenous turnover of KIT protein is known to be involved in the autophagic process, and the novel HSP90AA1 inhibitor, NVP-AUY92, has been revealed to enhance the magnitude of autophagy for promoting KIT degradation (34). To date, few studies have described the autophagy-modulating role of FASN inhibition in tumor cells. One such study reported that glioma cells treated with C75 or orlistat revealed increased autophagic activity (24). In imatinib-resistant GIST cells, increased expression of LC3-II, a widely accepted autophagic marker (31–34), was inducible by C75 (also orlistat), and its level could be even higher upon combined treatment with C75 and imatinib. Although AKT inactivation–induced apoptosis directs cells to programmed death (17, 33), the significance of FASN inhibition–induced autophagy in GISTs requires further elucidation. Specifically, the relevant issues regarding C75-induced autophagy include its potential dependency on AKT, engagement in KIT expression and activation, and resultant cytoprotection to maintain energy homeostasis versus cytotoxicity to promote cellular death under nutrient deprivation. A better understanding of these aspects will help decipher the possible benefits or detriments of autophagic inhibition in GIST cell growth in combined therapy against FASN and KIT.

In short, we have characterized the clinical relevance and biological implications of FASN, as well as the potential of dual blockade of FASN and KIT in GISTs. With FASN mRNA being preferentially upregulated in aggressive GISTs, overexpressed FASN confers growth-promoting and promigratory oncometabolic phenotypes that are associated with adverse clinicopathologic factors and unfavorable RTK genotypes, and independently portends decreased DFS. Biologically, an interdependent positive feedback loop may exist to coordinately regulate FASN, KIT-elicited signaling output, and the PI3K/AKT/mTOR pathway in GISTs. Through disrupting this loop, FASN-targeting C75 represses KIT transactivation to exhaust KIT protein, induces apoptosis and autophagy, and inactivates signaling kinases downstream of PI3K/AKT. Therefore, FASN inhibition improves sensitivity to imatinib in resistant cell models and underpins the in vitro and in vivo additivism of C75/imatinib combination therapy. On the basis of the suppressive effects that C75 demonstrated, our study provides a rationale for future investigation of novel FASN inhibitors with a better therapeutic index in combination with imatinib in GISTs that acquire refractory mutations.

No potential conflicts of interest were disclosed.

Conception and design: C.-F. Li, L.-T. Chen, H.-Y. Huang

Development of methodology: C.-F. Li, T.-C. Chan, S.-C. Yu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C.-T. Li, T.-C. Chan, H.-Y. Huang

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): F.-M. Fang, Y.-Y. Chen, L.-T. Chen

Writing, review, and/or revision of the manuscript: C.-F. Li, S.-C. Yu, H.-Y. Huang

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.-F. Li, F.-M. Fang, Y.-Y. Chen, T.-C. Chan, S.-C. Yu, L.-T. Chen

Study supervision: L.-T. Chen and H.-Y. Huang supervised this work equally

Other (carried out the experiments): T.-T. Liu

The authors thank Dr. Jonathan Fletcher for providing GIST cell lines and Kaohsiung Chang Gung genomic (CLRPG8D0113) and tissue bank (CLRPG8B0033, CLRPG8E0161) core laboratories for technical assistance. The authors also thank Dr. Shiaw-Min Huang at Bioresource Collection and Research Center, Food Industry Research and Development Institute for the authentication of GIST48 and GIST430 cells.

H.-Y. Huang was funded by Taiwan National Ministry of Science and Technology (NSC99-2628-B-182A-064-MY3) and Chang Gung Hospital (CMRPG8C0983). C.-F. Li was funded by Taiwan National Ministry of Science and Technology (NSC99-2320-B-384-001-MY2 and MOST104-2314-B-384 -009-MY3), Ministry of Health and Welfare (MOHW103-TDU-M-221-123017 and MOHW104-TDU-M-212-133004), and Taiwan National Health Research Institutes (CA-106-PP-36 and CA-106-SP-03).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Heinrich
MC
,
Corless
CL
,
Blanke
CD
,
Demetri
GD
,
Joensuu
H
,
Roberts
PJ
, et al
Molecular correlates of imatinib resistance in gastrointestinal stromal tumors
.
J Clin Oncol
2006
;
24
:
4764
74
.
2.
Joensuu
H
,
Rutkowski
P
,
Nishida
T
,
Steigen
SE
,
Brabec
P
,
Plank
L
, et al
KIT and PDGFRA mutations and the risk of GI stromal tumor recurrence
.
J Clin Oncol
2015
;
33
:
634
42
.
3.
Gasparotto
D
,
Rossi
S
,
Polano
M
,
Tamborini
E
,
Lorenzetto
E
,
Sbaraglia
M
, et al
Quadruple-negative GIST is a sentinel for unrecognized Neurofibromatosis Type 1 syndrome
.
Clin Cancer Res
2017
;
23
:
273
82
.
4.
Rossi
S
,
Sbaraglia
M
,
Dell'Orto
MC
,
Gasparotto
D
,
Cacciatore
M
,
Boscato
E
, et al
Concomitant KIT/BRAF and PDGFRA/BRAF mutations are rare events in gastrointestinal stromal tumors
.
Oncotarget
2016
;
7
:
30109
18
.
5.
Demetri
GD
,
von Mehren
M
,
Antonescu
CR
,
DeMatteo
RP
,
Ganjoo
KN
,
Maki
RG
, et al
NCCN Task Force report: update on the management of patients with gastrointestinal stromal tumors
.
J Natl Compr Canc Netw
2010
;
8
Suppl 2
:
S1
S41
.
6.
Fletcher
CD
,
Berman
JJ
,
Corless
C
,
Gorstein
F
,
Lasota
J
,
Longley
BJ
, et al
Diagnosis of gastrointestinal stromal tumors: a consensus approach
.
Hum Pathol
2002
;
33
:
459
65
.
7.
Antonescu
CR
,
Besmer
P
,
Guo
T
,
Arkun
K
,
Hom
G
,
Koryotowski
B
, et al
Acquired resistance to imatinib in gastrointestinal stromal tumor occurs through secondary gene mutation
.
Clin Cancer Res
2005
;
11
:
4182
90
.
8.
Liu
R
,
Huang
Y
. 
Lipid Signaling in Tumorigenesis
.
Mol Cell Pharmacol
2014
;
6
:
1
9
.
9.
Flavin
R
,
Peluso
S
,
Nguyen
PL
,
Loda
M
. 
Fatty acid synthase as a potential therapeutic target in cancer
.
Future Oncol
2010
;
6
:
551
62
.
10.
Menendez
JA
,
Lupu
R
. 
Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis
.
Nat Rev Cancer
2007
;
7
:
763
77
.
11.
Rossi
S
,
Ou
W
,
Tang
D
,
Bhattacharya
N
,
Dei Tos
AP
,
Fletcher
JA
, et al
Gastrointestinal stromal tumours overexpress fatty acid synthase
.
J Pathol
2006
;
209
:
369
75
.
12.
Bauer
S
,
Duensing
A
,
Demetri
GD
,
Fletcher
JA
. 
KIT oncogenic signaling mechanisms in imatinib-resistant gastrointestinal stromal tumor: PI3-kinase/AKT is a crucial survival pathway
.
Oncogene
2007
;
26
:
7560
8
.
13.
Li
CF
,
Chen
LT
,
Lan
J
,
Chou
FF
,
Lin
CY
,
Chen
YY
, et al
AMACR amplification and overexpression in primary imatinib-naive gastrointestinal stromal tumors: a driver of cell proliferation indicating adverse prognosis
.
Oncotarget
2014
;
5
:
11588
603
.
14.
Li
CF
,
Huang
WW
,
Wu
JM
,
Yu
SC
,
Hu
TH
,
Uen
YH
, et al
Heat shock protein 90 overexpression independently predicts inferior disease-free survival with differential expression of the alpha and beta isoforms in gastrointestinal stromal tumors
.
Clin Cancer Res
2008
;
14
:
7822
31
.
15.
Huang
HY
,
Li
SH
,
Yu
SC
,
Chou
FF
,
Tzeng
CC
,
Hu
TH
, et al
Homozygous deletion of MTAP gene as a poor prognosticator in gastrointestinal stromal tumors
.
Clin Cancer Res
2009
;
15
:
6963
72
.
16.
Li
CF
,
Liu
TT
,
Chuang
IC
,
Chen
YY
,
Fang
FM
,
Chan
TC
, et al
PLCB4 copy gain and PLCß4 overexpression in primary gastrointestinal stromal tumors: Integrative characterization of a lipid-catabolizing enzyme associated with worse disease-free survival
.
Oncotarget
2017
;
8
:
19997
20010
.
17.
Calvisi
DF
,
Wang
C
,
Ho
C
,
Ladu
S
,
Lee
SA
,
Mattu
S
, et al
Increased lipogenesis, induced by AKT-mTORC1-RPS6 signaling, promotes development of human hepatocellular carcinoma
.
Gastroenterology
2011
;
140
:
1071
83
.
18.
Relat
J
,
Blancafort
A
,
Oliveras
G
,
Cufi
S
,
Haro
D
,
Marrero
PF
, et al
Different fatty acid metabolism effects of (-)-epigallocatechin-3-gallate and C75 in adenocarcinoma lung cancer
.
BMC Cancer
2012
;
12
:
280
.
19.
Lewis
CA
,
Griffiths
B
,
Santos
CR
,
Pende
M
,
Schulze
A
. 
Regulation of the SREBP transcription factors by mTORC1
.
Biochem Soc Trans
2011
;
39
:
495
9
.
20.
Menendez
JA
,
Vellon
L
,
Lupu
R
. 
Targeting fatty acid synthase-driven lipid rafts: a novel strategy to overcome trastuzumab resistance in breast cancer cells
.
Med Hypotheses
2005
;
64
:
997
1001
.
21.
Pizer
ES
,
Thupari
J
,
Han
WF
,
Pinn
ML
,
Chrest
FJ
,
Frehywot
GL
, et al
Malonyl-coenzyme-A is a potential mediator of cytotoxicity induced by fatty-acid synthase inhibition in human breast cancer cells and xenografts
.
Cancer Res
2000
;
60
:
213
8
.
22.
Chen
C
,
Han
X
,
Zou
X
,
Li
Y
,
Yang
L
,
Cao
K
, et al
4-methylene-2-octyl-5-oxotetrahydrofuran-3-carboxylic acid (C75), an inhibitor of fatty-acid synthase, suppresses the mitochondrial fatty acid synthesis pathway and impairs mitochondrial function
.
J Biol Chem
2014
;
289
:
17184
94
.
23.
Impheng
H
,
Pongcharoen
S
,
Richert
L
,
Pekthong
D
,
Srisawang
P
. 
The selective target of capsaicin on FASN expression and de novo fatty acid synthesis mediated through ROS generation triggers apoptosis in HepG2 cells
.
PLoS One
2014
;
9
:
e107842
.
24.
Grube
S
,
Dunisch
P
,
Freitag
D
,
Klausnitzer
M
,
Sakr
Y
,
Walter
J
, et al
Overexpression of fatty acid synthase in human gliomas correlates with the WHO tumor grade and inhibition with Orlistat reduces cell viability and triggers apoptosis
.
J Neurooncol
2014
;
118
:
277
87
.
25.
Patel
AV
,
Johansson
G
,
Colbert
MC
,
Dasgupta
B
,
Ratner
N
. 
Fatty acid synthase is a metabolic oncogene targetable in malignant peripheral nerve sheath tumors
.
Neuro Oncol
2015
;
17
:
1599
608
.
26.
Sambol
EB
,
Ambrosini
G
,
Geha
RC
,
Kennealey
PT
,
Decarolis
P
,
O'Connor
R
, et al
Flavopiridol targets c-KIT transcription and induces apoptosis in gastrointestinal stromal tumor cells
.
Cancer Res
2006
;
66
:
5858
66
.
27.
Menendez
JA
,
Vellon
L
,
Mehmi
I
,
Oza
BP
,
Ropero
S
,
Colomer
R
, et al
Inhibition of fatty acid synthase (FAS) suppresses HER2/neu (erbB-2) oncogene overexpression in cancer cells
.
Proc Natl Acad Sci U S A
2004
;
101
:
10715
20
.
28.
Vazquez-Martin
A
,
Colomer
R
,
Brunet
J
,
Menendez
JA
. 
Pharmacological blockade of fatty acid synthase (FASN) reverses acquired autoresistance to trastuzumab (Herceptin by transcriptionally inhibiting ‘HER2 super-expression’ occurring in high-dose trastuzumab-conditioned SKBR3/Tzb100 breast cancer cells
.
Int J Oncol
2007
;
31
:
769
76
.
29.
Wagner
R
,
Stübiger
G
,
Veigel
D
,
Wuczkowski
M
,
Lanzerstorfer
P
,
Weghuber
J
, et al
Multi-level suppression of receptor-PI3K-mTORC1 by fatty acid synthase inhibitors is crucial for their efficacy against ovarian cancer cells
.
Oncotarget
2017
;
8
:
11600
13
.
30.
Benjamin
DI
,
Li
DS
,
Lowe
W
,
Heuer
T
,
Kemble
G
,
Nomura
DK
. 
Diacylglycerol metabolism and signaling is a driving force underlying FASN inhibitor sensitivity in cancer cells
.
ACS Chem Biol
2015
;
10
:
1616
23
.
31.
Kim
WK
,
Yun
S
,
Park
CK
,
Bauer
S
,
Kim
J
,
Lee
MG
, et al
Sustained mutant KIT activation in the Golgi complex is mediated by PKC-θ in gastrointestinal Stromal Tumors
.
Clin Cancer Res
2017
;
23
:
845
56
.
32.
Chen
S
,
Rehman
SK
,
Zhang
W
,
Wen
A
,
Yao
L
,
Zhang
J
. 
Autophagy is a therapeutic target in anticancer drug resistance
.
Biochim Biophys Acta
2010
;
1806
:
220
9
.
33.
Maiuri
MC
,
Zalckvar
E
,
Kimchi
A
,
Kroemer
G
. 
Self-eating and self-killing: crosstalk between autophagy and apoptosis
.
Nat Rev Mol Cell Biol
2007
;
8
:
741
52
.
34.
Hsueh
YS
,
Yen
CC
,
Shih
NY
,
Chiang
NJ
,
Li
CF
,
Chen
LT
. 
Autophagy is involved in endogenous and NVP-AUY922-induced KIT degradation in gastrointestinal stromal tumors
.
Autophagy
2013
;
9
:
220
33
.