Purpose:

This work aimed to explore in depth the genomic and molecular underpinnings of hepatocellular carcinoma (HCC) with increased 2[18F]fluoro-2-deoxy-d-glucose (FDG) uptake in PET and to identify therapeutic targets based on this imaging-genomic surrogate.

Experimental Design:

We used RNA sequencing and whole-exome sequencing data obtained from 117 patients with HCC who underwent hepatic resection with preoperative FDG-PET/CT imaging as a discovery cohort. The primary radiogenomic results were validated with transcriptomes from a second cohort of 81 patients with more advanced tumors. All patients were allocated to an FDG-avid or FDG–non-avid group according to the PET findings. We also screened potential drug candidates targeting FDG-avid HCCs in vitro and in vivo.

Results:

High FDG avidity conferred worse recurrence-free survival after HCC resection. Whole transcriptome analysis revealed upregulation of mTOR pathway signals in the FDG-avid tumors, together with higher abundance of associated mutations. These clinical and genomic findings were replicated in the validation set. A molecular signature of FDG-avid HCCs identified in the discovery set consistently predicted poor prognoses in the public-access datasets of two cohorts. Treatment with an mTOR inhibitor resulted in decreased FDG uptake followed by effective tumor control in both the hyperglycolytic HCC cell lines and xenograft mouse models.

Conclusions:

Our PET-based radiogenomic analysis indicates that mTOR pathway genes are markedly activated and altered in HCCs with high FDG retention. This nuclear imaging biomarker may stimulate umbrella trials and tailored treatments in precision care of patients with HCC.

Translational Relevance

Radiogenomics has been mainly used to create imaging biomarkers mirroring the genetic and molecular makeup of malignant disease without tissue sampling. The heterogeneity of 2[18F]fluoro-2-deoxy-d-glucose (FDG) uptake and its biological correlates in PET imaging for hepatocellular carcinoma (HCC) could facilitate radiogenomic approaches to the disease. HCC tumors with increased FDG uptake, which are associated with worse outcomes, are selectively enriched for expression and mutation of genes in the mTOR signaling pathway. A molecular signature relevant to FDG-avid HCCs in public-access cohorts is consistently correlated with unfavorable prognoses. In vitro and in vivo studies showed that the mTOR inhibitor, omipalisib, had antitumor activity via suppression of glucose metabolism in FDG-avid HCC cells. PET-based radiogenomics has identified aberrant mTOR activity as a potential target in hyperglycolytic HCCs. Our findings could warrant a novel focus on clinical trials of HCC based on the FDG-PET findings, and help tailor future treatments.

2[18F]fluoro-2-deoxy-d-glucose PET and CT (FDG-PET/CT) are diagnostic tools for imaging selective uptake of the radiolabeled glucose analog, FDG, by cancer cells, which are characterized by enhanced glucose metabolism (1). In general, FDG-PET/CT plays a practical role in detecting metastatic foci, rather than identifying intrahepatic tumors, in the staging of patients with hepatocellular carcinoma (HCC; ref. 2–4). Increased PET uptake is likely to be most useful as an oncologic marker for selecting resection or transplant candidates and predicting posttreatment outcomes (5–8).

Radiogenomics has been mainly used to create imaging biomarkers mirroring the genomolecular makeup of malignant disease without needing tissue sampling (9–11). FDG-PET/CT imaging combined with anatomic and functional properties could be an excellent tool for radiogenomic approaches in oncology care (12). Despite the widespread clinical utility of FDG-PET imaging in multiple cancer types, the underlying cellular or molecular bases have only been investigated in a few types such as breast, colon, and lung cancers (13–15). The relevant issues still remain unclear in HCC. Fortunately, the intensity of FDG-PET uptake in HCC lesions varies markedly across tumors and patients (7, 16–18), and this could facilitate radiogenomic studies of the disease.

To address this matter, we examined the prognostic significance of preoperative FDG-PET uptake by the target lesion in patients undergoing surgical resection for HCC, and analyzed the molecular and genotypic correlates of high intensity FDG-PET uptake. The ultimate goal of this study was to identify therapeutic targets based on stratifying patients with HCC in terms of radiogenomic surrogates. To this end, we designed in vitro and in vivo experiment to characterize FDG-avid HCC cell lines.

Patient and sample data collection

The current study was approved by the Institutional Review Board (IRB) of Asan Medical Center (No. 2019–1321), and the study was conducted in accordance with the Declaration of Helsinki. Specimens provided by the Bio-resource center were obtained from patients who gave written informed consent. A flow diagram of the overall study design is displayed in Fig. 1.

Figure 1.

Flow diagram of the overall study design.

Figure 1.

Flow diagram of the overall study design.

Close modal

Discovery and validation cohorts

We first screened a preconstructed panel of 206 patients who received curative hepatectomy for HCC between 2005 and 2011 at Asan Medical Center and for whom there were both whole-exome sequencing and RNA sequencing (RNA-seq) data on resected tumor samples (19–21). We finally curated 117 individuals in whom FDG-PET/CT scans obtained within 4 weeks before surgery were available, and used them as a discovery set (Supplementary Table S1). No patients received prior local or systemic anti-HCC treatment. Most were infected with hepatitis B virus (HBV; 77.8%) and had early-stage disease [i.e., Barcelona Clinic Liver Cancer (BCLC)-0/A; 95.7%]. To confirm the robustness and generality of the results from this set, we gathered a validation set of 81 patients with HCC of different composition who underwent primary hepatic resection between 2009 and 2013, and for whom we had surgical specimens archived in our own Bio-resource center (http://brc.amc.seoul.kr), in addition to preoperative FDG PET/CT scans. The archive is an IRB-approved human biorepository in Asan Medical Center. Over half of the validation cohort had non–early stages of HCC (i.e., BCLC-B/C; 55.6%), and 39.5% of the tumors were of non-HBV origin (Supplementary Table S2). RNA-seq data were newly generated for each surgical sample in this set. See Supplementary Data for details.

External public cohorts

To examine the external consistency and retest reliability of the prognostic power of the molecular signatures relevant to FDG-avid tumors, we used the Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) and Liver Cancer-RIKEN, Japan (LIRI-JP) datasets comprising 262 and 191 samples, respectively, with pathologically confirmed HCC and full outcome data (22, 23). The two public-access gene expression datasets were downloaded from the cBioPortal (RRID:SCR_014555) and International Cancer Genome Consortium data portal (https://dcc.icgc.org), respectively. In terms of outcome data, both overall survival (OS) and recurrence-free survival (RFS) were available in the TCGA-LIHC database, but only OS was available for the LIRI-JP samples. Details of the demographic and clinical characteristics of each sample contained in the two datasets are provided in Supplementary Tables S3 and S4.

PET/CT acquisition and extraction of FDG uptake features

For details of the PET/CT imaging protocols, see Supplementary Data. All PET/CT images were analyzed using a dedicated workstation (Syngo.via VB20A, Siemens, Erlangen, Germany) by two board-certified nuclear medicine physicians working in consensus (J.S. Kim and M. Oh). For quantitative analysis, maximum standardized uptake values (SUVmax) were measured by drawing a volume-of-interest (VOI) for the tumor in the liver (24). In cases of multiple lesions, the SUVmax that showed the highest FDG uptake was recorded. Tumor to non-tumor SUV ratios (TNR) were calculated as the ratios between the SUVmax of tumors and the mean SUV of liver backgrounds determined by averaging the SUVs of three 1-cm spherical lesion-free VOIs– two in the right lobe and one in the left lobe (25).

On the basis of visual analysis of the largest tumor, tumors were classified as FDG-avid or FDG–non-avid depending on whether FDG-PET uptake was higher than background hepatic activity or similar to or lower than background hepatic activity (26). Representative FDG-PET/CT images of each group are presented in Fig. 2A.

Figure 2.

Prognostic and transcriptomic underpinnings of FDG-PET avidity in HCC. A, Representative images of FDG-avid (left) and FDG–non-avid (right) HCC tumors. B, RFS according to FDG-PET avidity in the discovery (left) and validation (right) cohorts. The FDG-avid groups in both cohorts had significantly lower RFS rates than the FDG–non-avid subjects. C, Distribution of the five predefined HCC molecular classes according to FDG uptake in the discovery set. D, Enrichment of gene sets related to poor prognosis and stem cell features in the FDG-avid HCCs in the discovery set.

Figure 2.

Prognostic and transcriptomic underpinnings of FDG-PET avidity in HCC. A, Representative images of FDG-avid (left) and FDG–non-avid (right) HCC tumors. B, RFS according to FDG-PET avidity in the discovery (left) and validation (right) cohorts. The FDG-avid groups in both cohorts had significantly lower RFS rates than the FDG–non-avid subjects. C, Distribution of the five predefined HCC molecular classes according to FDG uptake in the discovery set. D, Enrichment of gene sets related to poor prognosis and stem cell features in the FDG-avid HCCs in the discovery set.

Close modal

Genomic and transcriptomic datasets and analyses

DNA- or RNA-seq data were obtained from the largest tumor in subjects in both the discovery and validation cohorts with ≥2 intrahepatic lesions. To identify functional sets of genes associated with FDG uptake, single-sample scores for each gene set were calculated from the transcriptomic data by the single-sample Gene Set Enrichment Analysis (GSEA) method implemented in Genepattern (27, 28). Differentially expressed genes in the FDG-avid group and their protein–protein interaction networks were identified and deduced using the STRING (RRID:SCR_005223) database (https://string-db.org; ref. 29).

We developed an “FDG-avid signature”, which consisted of the top hundred genes significantly upregulated in the FDG-avid group of the discovery cohort in order of P value (Supplementary Table S5), and tested its prognostic value in the external public datasets. See Supplementary Data for the further transcriptomic analyses.

In vitro and in vivo biological evaluation of the radiogenomic findings

Five well-characterized human HCC cell lines (Huh7, PLC/PRF/5, Hep3B, HepG2, and SNU182) were obtained from the Korean Cell Line Bank, Seoul, Republic of Korea. Sorafenib-resistant Huh7 cells (Huh7-SR) were generated by exposure of Huh7 cells to stepwise increments of sorafenib concentration (Supplementary Figs. S1 and S2). The cell lines were authenticated upon receipt by short tandem repeat (STR) profiling, using an AmpFlSTR Identifier Kit (Applied Biosystems, Foster City, California, Catalog No. 4322288), and based on available STR profiles. Before use, all cells were screened for the presence of Mycoplasma by PCR using a Mycoplasma detection kit (Myco-ReadTM, BioMAX). The reported experiments were performed less than 2 months after thawing early passage cells. Details of in vitro studies, including 18F-FDG uptake testing, cell viability assays, and Western blot analysis are given in the Supplementary Data. PLC/PRF/5 cell line-based xenograft NRG mouse models were used for animal studies. Experimental designs and procedures, including animal FDG-PET imaging and IHC staining, are presented in the Supplementary Data. Omipalisib (GSK2126458; Sellckchem) was used to block the mTOR pathway in both in vitro and in vivo settings. All animal study protocols were approved by the Institutional Animal Care and Use Committee of Asan Institute for Life Sciences (No. 2019–02–341).

Statistical analysis

For statistical comparisons, the χ2 test, Fisher exact test, Student t test, or Mann–Whitney U test was performed when appropriate. RFS was calculated from the date of tumor resection until detection of the first HCC recurrence, death, or the last follow-up. Survival curves were analyzed by the Kaplan–Meier method and compared using the log-rank test. Independent factors for RFS were evaluated by multivariate Cox regression analysis. Significance was defined as two-tailed P < 0.05. All statistical analyses were performed with R Version 3.4.1.

Data availability statement

Normalized gene expression data for the discovery and validation cohorts have been deposited in GEO (https://www.ncbi.nlm.nih.gov/geo; accession nos. GSE124751 and GSE164121, respectively).

Clinical implications of FDG-PET avidity in the HCC cohorts

Of the 117 patients in the discovery cohort, 57 (48.7%) had FDG-avid tumors. The FDG-avid group was associated with worse tumor biology in terms of poorly differentiated tumors, microvascular invasion, and serum α-fetoprotein (AFP) elevation (Supplementary Table S1); similar trends were also observed in the validation set of 81 patients (Supplementary Table S2). RFS time was shorter in the FDG-avid group, with 5-year RFS rates of 48% in the avid group and 68% in the non-avid group, and this difference remained significant after adjusting for all clinical covariates including traditional predictors such as microvascular invasion and serum AFP level [adjusted hazard ratio (HR), 1.69; 95% confidence interval (CI), 1.01–2.83; P = 0.045; Fig. 2B; Supplementary Table S6). The difference was even clearer in the validation set of more advanced tumors [adjusted HR, 2.45; 95% CI, 1.33–4.51; P = 0.004 for the FDG-avid group (60.5%; n = 49) vs. the FDG–non-avid group (39.5%; n = 32); Fig. 2B; Supplementary Table S6].

In terms of the predefined molecular classifications of HCC (30–34), Hoshida S1 (43.9%), Boyault G1-G3 (56.1%), Chiang's proliferation (57.9%), and Lee's (73.7%), and Roessler's A (64.9%) classes, along with proliferation properties, were the most prevalent subclasses in the FDG-avid group of the discovery set (Fig. 2C). In contrast, the FDG–non-avid group members were mainly Hoshida S3 (58.3%) and Boyault G4 (45.0%) tumors, which are known to be well differentiated. In addition, the FDG–non-avid tumors were significantly enriched in the Lee's (70.0%) and Roessler's B (65.0%) clusters associated with favorable survival (32, 34). Gene sets representing poor prognostic signatures for HCC (33–36) were also significantly associated with intense FDG uptake (Fig. 2D).

Associations between the expression of metabolic markers and FDG uptake levels

To see whether HCC-driven FDG uptake reflected the extent of glucose metabolism, we measured the expression of glycolysis-related genes and examined their relationships with SUVs on PET in the discovery samples (Supplementary Fig. S3). The expression of the glycolytic genes, SLC2A1 and HK2, encoding glucose transporter 1 (GLUT1) and hexokinase 2 (37), respectively, was found to be positively correlated with TNR expression (r = 0.50, P < 0.001; and r = 0.32, P < 0.001, respectively). In contrast, an inverse relationship with TNR was observed for the G6PC gene, which is involved in gluconeogenesis, the opposite process to glycolysis (37). As anticipated, expression of both glycolytic genes was significantly higher in the FDG-avid group (Ps < 0.01). These observations demonstrate that the FDG-avid HCCs are metabolically active cells with increased uptake and trapping of FDG, possibly due to upregulation of GLUT1 and hexokinase 2 and downregulation of glucose 6-phosphatase (37).

Identification of gene sets related to FDG-PET avidity

Single-sample GSEA in the discovery set based on Hallmark databases revealed that FDG-avid tumors were significantly enriched for gene sets related to glycolysis and its regulatory partners in the cell cycle (Fig. 3A). Strikingly, both the MTORC1 and PI3K_AKT_MTOR pathways and their core components such as TSC2, MTOR, RHEB, and EIF-4EB1 were strongly upregulated in PET-positive cases, suggesting that activation of mTOR signaling could be a key element of the glycolytic phenotype in tumor cells (Fig. 3A and B). In addition to the individual involvement in mTOR and glycolysis-related genotypes detected by GSEA, high biological connectivity of key components in the two genetic pathways was also observed by visualizing the protein-networks of differentially expressed genes in FDG-avid HCCs, as expressed by a high average local clustering coefficient of 0.83 (Fig. 3C). We next examined somatic mutations and DNA copy-number alterations, focusing on a list of 17 genes in the canonical PI3K/AKT/MTOR pathway. A number of genes in the pathway were found significantly altered in the FDG-avid group, including PIK3CA, PTEN, TSC2, RICTOR, and MTOR (36.8% vs. 18.3%, P = 0.042; Fig. 3D). On the other hand, genes associated with hepatic physiology including bile acid and fatty acid metabolism, adipogenesis, and coagulation were downregulated in samples with high FDG uptake. Other gene sets including TGFβ signaling- and immune response–related pathways were not significantly associated with FDG-PET avidity (Supplementary Fig. S4). These molecular findings were confirmed in the validation set of more advanced-stage tumors, which might become potential major candidates for mTOR pathway inhibitors (Fig. 3E).

Figure 3.

Genetic activation and mutations of the mTOR pathway in FDG-avid HCC. A, Heat map using single-sample GSEA based on the Hallmark database. B, Significant dysregulation of representative mTOR signaling–related genes in the FDG-avid group. C, STRING-generated visualization of the functional protein association network involving mTOR and the glycolysis pathway components differentially expressed in FDG-avid tumors. The red and green nodes indicate mTOR signaling and glycolytic genes, respectively; and the red and blue borders indicate upregulated and downregulated genes, respectively. D, Landscape and frequency of somatic mutations in 17 genes (PIK3CA, PIK3R1, PIK3C2B, PTEN, INPP4B, MAPKAP1, PDK1, TSC1, TSC2, RHEB, MTOR, RPTOR, RICTOR, DEPTOR, DEPDC5, NPRL3, and STK11) involved in the PI3K/AKT/mTOR pathway according to FDG uptake. E, GSEA results for the validation dataset. MTORC1, PI3K_AKT_MTOR, and cell-cycle signaling were also significantly upregulated in the FDG-avid group in the validation set.

Figure 3.

Genetic activation and mutations of the mTOR pathway in FDG-avid HCC. A, Heat map using single-sample GSEA based on the Hallmark database. B, Significant dysregulation of representative mTOR signaling–related genes in the FDG-avid group. C, STRING-generated visualization of the functional protein association network involving mTOR and the glycolysis pathway components differentially expressed in FDG-avid tumors. The red and green nodes indicate mTOR signaling and glycolytic genes, respectively; and the red and blue borders indicate upregulated and downregulated genes, respectively. D, Landscape and frequency of somatic mutations in 17 genes (PIK3CA, PIK3R1, PIK3C2B, PTEN, INPP4B, MAPKAP1, PDK1, TSC1, TSC2, RHEB, MTOR, RPTOR, RICTOR, DEPTOR, DEPDC5, NPRL3, and STK11) involved in the PI3K/AKT/mTOR pathway according to FDG uptake. E, GSEA results for the validation dataset. MTORC1, PI3K_AKT_MTOR, and cell-cycle signaling were also significantly upregulated in the FDG-avid group in the validation set.

Close modal

Prognostic validation of the FDG-avid signature in different cohorts

We divided the levels of expression of the FDG-avid signature based on the single-sample GSEA enrichment scores: high for the top 25%, mid for the 25%–75% group, and low for the bottom 25% group. We next examined the prognostic relevance of this grouping in the two public HCC sets, of multiethnic (TCGA-LIHC; n = 262) and Japanese origin (LIRI-JP, n = 191), respectively, as shown in Fig. 4. Tumors with high FDG-avid signatures were significantly associated with the poorest survival outcomes, followed by the mid FDG-avid signature samples in both cohorts. These correlations point to a genetic contribution to the FDG-PET uptake that reflects the level of glucose utilization in cancer cells.

Figure 4.

Kaplan–Meier curves for survival outcomes according to expression of the FDG-avid signature based on the two public-use HCC datasets. The expression levels of the FDG-avid signature displayed significant stepwise correlations with the RFS or OS of patients after HCC resection in the TCGA-LIHC cohort (n = 262; A) and the LIRI-JP cohort (n = 191; B).

Figure 4.

Kaplan–Meier curves for survival outcomes according to expression of the FDG-avid signature based on the two public-use HCC datasets. The expression levels of the FDG-avid signature displayed significant stepwise correlations with the RFS or OS of patients after HCC resection in the TCGA-LIHC cohort (n = 262; A) and the LIRI-JP cohort (n = 191; B).

Close modal

Functional changes of glucose metabolism and cellular viability after mTOR inhibition

Based on the radiogenomic data identified in clinical settings, we next investigated the changes in metabolic and proliferative behavior upon inhibition of the mTOR pathway in in vitro models of HCC. We first screened six representative HCC cell lines, and, among these, we selected for further experiments PLC/PRF/5, Huh7, and Huh-SR cells, which displayed increased glucose uptake (Fig. 5A). To evaluate the potential of mTOR inhibition for treating FDG-avid HCC, we tested the effect of omipalisib, a second-generation dual PI3K/mTOR inhibitor, in phase I clinical trials against solid tumors (38), in the three selected cell lines, that showed the strongest anticancer efficacy against those cell lines after screening a library of 17 anticancer drugs targeting the PI3K–mTOR pathway (Supplementary Fig. S5). The IC50 values for omipalisib ranged from 0.07 to 1.3 μmol/L in both the parental and sorafenib-resistant cell lines, while those for sorafenib, lenvatinib, and regorafenib, which are first-line or rescue drugs for sorafenib failure currently used to treat HCC in clinical practice (6), were mostly >4.0 μmol/L (Fig. 5B). Interestingly, omipalisib was much more effective in Huh7-SR cells than regorafenib, a standard rescue drug for sorafenib (39). Treatment with increasing concentrations of omipalisib for 72 hours reduced the viability of FDG-avid PLC/PRF/5 and Huh7 to a greater extent than that of non-avid cells such as Hep3B and HepG2 (Supplementary Fig. S6). Glucose uptake was already reduced in all three cell lines shortly after 24 hours of exposure to 0.5 μmol/L omipalisib, and this was accompanied by decreased expression of major modulators of glycolysis such as GLUT1 and hexokinase 2, and these findings were restricted to the FDG-avid cell lines (Fig. 5C and D; Supplementary Figs. S7 and S8). However, a significant drop in cell viability in all these cell lines was evident from 48 hours of treatment (Fig. 5E). Western blotting also showed that omipalisib resulted in decreased phosphorylation of mTOR and its downstream targets in the early phase (24 hours) of treatment (Fig. 5F).

Figure 5.

Effects of mTOR inhibition in hyperglycolytic HCC cells. A,18F-FDG retention in HCC cell lines. Error bars indicate SEM. B, Dose–response curves for omipalisib, sorafenib, lenvatinib, and regorafenib in FDG-avid cell lines, Huh7, Huh7-SR, and PLC/PRF/5. The corresponding IC50 values for each cell line are shown. C, Changes of glucose uptake in FDG-avid cells exposed to 0.5 μmol/L omipalisib. Glucose Uptake-Glo was used to measure glucose uptake rates, as described in Supplementary Data. Error bars, SEM. Significance was assessed by Student t test. *, P < 0.05. D, Western blots of hexokinase 2 and GLUT1 after treatment with omipalisib. E, Proliferation of FDG-avid cells after exposure to 0.5 μmol/L omipalisib. Cell viability was measured by CellTiter-Glo. Error bars, SEM. Significance was determined by Student t test. *, P < 0.05. F, Western blots of mTOR pathway proteins at baseline, 24 hours, and 48 hours after treatment of the three FDG-avid cell lines with the indicated doses of omipalisib.

Figure 5.

Effects of mTOR inhibition in hyperglycolytic HCC cells. A,18F-FDG retention in HCC cell lines. Error bars indicate SEM. B, Dose–response curves for omipalisib, sorafenib, lenvatinib, and regorafenib in FDG-avid cell lines, Huh7, Huh7-SR, and PLC/PRF/5. The corresponding IC50 values for each cell line are shown. C, Changes of glucose uptake in FDG-avid cells exposed to 0.5 μmol/L omipalisib. Glucose Uptake-Glo was used to measure glucose uptake rates, as described in Supplementary Data. Error bars, SEM. Significance was assessed by Student t test. *, P < 0.05. D, Western blots of hexokinase 2 and GLUT1 after treatment with omipalisib. E, Proliferation of FDG-avid cells after exposure to 0.5 μmol/L omipalisib. Cell viability was measured by CellTiter-Glo. Error bars, SEM. Significance was determined by Student t test. *, P < 0.05. F, Western blots of mTOR pathway proteins at baseline, 24 hours, and 48 hours after treatment of the three FDG-avid cell lines with the indicated doses of omipalisib.

Close modal

This antitumor effect of mTOR inhibition via suppression of glucose metabolism was next examined in vivo using tumor xenografts derived from PLC/PRF/5 cells (Fig. 6A). Two weeks after injecting the cells, when tumor volume was around 150 mm3, we performed baseline FDG-PET imaging and then randomized the mice to receive either 1.5 mg/kg omipalisib (n = 7) or control vehicle (n = 7). After 3 days, FDG uptake began to decrease dramatically in the omipalisib-treated animals (Fig. 6B and C), and this was eventually followed by a decline in tumor volume (Fig. 6D and E), and remained low over the entire 3-week duration of treatment. IHC staining indicated that omipalisib exerted its antitumor effect by targeting mTORC1-dependent phosphorylation of 4E-BP1 and S6 ribosomal protein rather than phosphorylation of Akt, a sensitive substrate of mTORC2 (Fig. 6F; Supplementary Fig. S9). Collectively, these data suggest that the FDG-PET avidity per se of HCC tumors could dictate their susceptibility to the mTOR inhibitor.

Figure 6.

In vivo confirmation of therapeutic effects of mTOR inhibition on FDG-avid HCC cells. A, Schematic of the FDG-avid HCC xenograft mouse model treated with omipalisib (n = 7) and vehicle (n = 7). B, Effect of omipalisib (1.5 mg/kg orally once a day; red) versus vehicle (black) on FDG-PET uptake by the HCC tumor xenografts. C, Representative FDG-PET images before and after treatment. D, Effect of omipalisib (red) versus vehicle (black) on tumor growth in HCC xenograft mice. E, Appearance of ex vivo tumors 25 days after treatment with omipalisib versus vehicle. F, Representative IHC staining for phospho-4E-BP1, and phospho-S6 ribosomal protein (S6RP) expressed in HCC cancer cells in tissues obtained from tumors (x200). Omipalisib treatment attenuated phosphorylation of both 4E-BP1 and S6RP in tumors. *, P < 0.05; **, P < 0.01; ***, P < 0.001, for comparison of omipalisib with vehicle. Error bars represent SEM.

Figure 6.

In vivo confirmation of therapeutic effects of mTOR inhibition on FDG-avid HCC cells. A, Schematic of the FDG-avid HCC xenograft mouse model treated with omipalisib (n = 7) and vehicle (n = 7). B, Effect of omipalisib (1.5 mg/kg orally once a day; red) versus vehicle (black) on FDG-PET uptake by the HCC tumor xenografts. C, Representative FDG-PET images before and after treatment. D, Effect of omipalisib (red) versus vehicle (black) on tumor growth in HCC xenograft mice. E, Appearance of ex vivo tumors 25 days after treatment with omipalisib versus vehicle. F, Representative IHC staining for phospho-4E-BP1, and phospho-S6 ribosomal protein (S6RP) expressed in HCC cancer cells in tissues obtained from tumors (x200). Omipalisib treatment attenuated phosphorylation of both 4E-BP1 and S6RP in tumors. *, P < 0.05; **, P < 0.01; ***, P < 0.001, for comparison of omipalisib with vehicle. Error bars represent SEM.

Close modal

Since the glycolytic switch of cancer cells is controlled by nonmetabolic oncogenic regulators (40), FDG uptake in PET scans could serve as an oncologic surrogate bridging the phenomics and genomics of malignant diseases (1, 41). Our nuclear imaging-molecular correlative study found that aside from cell-cycle checkpoints reciprocally interconnected with metabolic pathways, the signaling network of mTOR cascades was robustly activated in FDG-avid HCCs—which have a poor prognosis—and this did not appear to depend on tumor stage. The same findings were noted for gene mutations in the PI3K/AKT/mTOR circuitry, which are known to be more prevalent in Asians with HCC (42, 43). In addition, the prognostic correlations of a nucleogenomic signature related to FDG-positive lesions in various racial and regional cohorts lend support to the idea that the molecular components involved play key roles in the aberrant glucose metabolism in HCC; these findings also avoid a limitation of the current study, namely that most of the HCC examined were due to HBV (Supplementary Tables S3 and S4; refs. 22, 23). Subsequent investigation showed that the reduction in FDG uptake shortly after inhibiting mTOR was followed by a significant decrease of cell viability and tumor regression specifically in the FDG-avid cell lines, which in turn may point to mTOR-dependent metabolic reprogramming of the HCC.

As a radiogenomic tool, PET has the advantage of allowing assessment of functional and physiologic activity with greater accuracy than anatomical imaging modalities such as CT or magnetic resonance imaging (41). 18F-FDG, an 18F-labeled glucose analogue, is the tracer of choice for detecting the increased glucose turnover in cancerous tissues (27). Features of tumor images extracted from FDG-PET/CT have been directly associated with type-specific genetic alterations in cancers, including epithelial–mesenchymal transition-related genes in lung cancer, MYC target genes in breast cancer, and KRAS/BRAF genes in colon cancer (14, 15, 44). The current study revealed the central metabolic effects of mTOR underpinning the intense FDG uptake in HCC tumors. The same correlation between (phosphorylated) mTOR expression and positive FDG-PET scans has been reported in IHC studies of non–small cell lung cancer and mesothelioma neoplasia (45, 46). The mTOR signaling pathway is actively involved in major aspects of HCC behavior including cell proliferation and spreading, which are energetically fueled by metabolic reprogramming (42). Such aggressive effects of deregulated mTOR linked to FDG uptake could be at least partly responsible for the early tumor recurrence in patients with PET-positive HCC.

Unfortunately, a representative randomized clinical trial of an mTOR inhibitor (i.e., everolimus) failed to demonstrate a survival advantage in the second-line setting of advanced HCC (47). In addition, a recent pan-cancer basket trial including 3 liver cancer cases did not demonstrate a predictive role for response to everolimus of genetic alterations in TSC1, TSC2, or MTOR related to mTOR activation, which were detected in only 6 samples out of our 57 FDG-avid series (48). However, an exploratory study of breast cancer samples from the TAMRAD trial indicated that activation of mTORC1 and its later effectors could be a potential predictive biomarker of everolimus efficacy (49). Indeed, the recent positive results from the REACH-2 trial of ramucirumab after sorafenib in a selected HCC population with serum AFP ≥400 ng/mL have encouraged biomarker-driven approaches to clinical trials in hepatic oncology (50). Based on our translational results, nuclear imaging criteria-based umbrella designs evaluating next-generation mTOR inhibitors (e.g., omipalisib) in patients chosen on the basis of FDG-PET features of HCC would be worth further consideration: these noninvasive protocols have the additional advantage that HCC is usually diagnosed from imaging studies alone without tissue sampling (6). On the other hand, PET imaging has emerged as a useful prognostic tool for evaluating HCC before liver transplantation (LT) due to its ability to predict aggressive biological behavior, as observed in our surgically resected cases (7, 8, 17). An updated meta-analysis demonstrated that mTOR inhibitor–based regimens improved post-LT outcomes, mainly in terms of HCC recurrence, compared with standard immunosuppression protocols, but the difference was only significant during the first few years after LT (51). Most recently, an exploratory analysis of the SiLVER-trial showed that the survival advantage of sirolimus treatment after LT for HCC was most pronounced in patients with active tumors producing AFP, a tumor group which is quite closely related to our FDG-avid series (52). Given the present radiogenomic findings, incorporating mTOR inhibitors into selected recipients with high FDG-avid signatures as measured by our method, or with increased FDG uptake in actual PET scans, is more likely to highlight or boost the anti-HCC effect of immunosuppressants.

In conclusion, the present combined imaging-genetic data indicate that aberrant signaling by the mTOR cascade is the key driving force in activating glucose metabolism in HCC as manifested by FDG uptake in PET images. Use of this nucleogenomic biomarker is likely to facilitate PET-based target curation in clinical trials, as well as precision pharmaceutical care employing tailored medicines.

No disclosures were reported.

J. An: Conceptualization, data curation, software, formal analysis, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing. M. Oh: Resources, data curation, software, formal analysis, supervision, visualization, methodology, writing–original draft. S.-Y. Kim: Software, formal analysis, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing. Y.-J. Oh: Resources, data curation, software, formal analysis, investigation, visualization, methodology, writing–review and editing. B. Oh: Data curation, validation, investigation, visualization, methodology, writing–review and editing. J.-H. Oh: Validation, visualization, methodology, writing–review and editing. W. Kim: Validation, investigation, visualization, writing–review and editing. J.H. Jung: Data curation, formal analysis, investigation. H.I. Kim: Data curation, supervision, validation, writing–review and editing. J.-S. Kim: Data curation, validation, investigation, methodology, writing–review and editing. C.O. Sung: Conceptualization, formal analysis, validation, investigation, visualization, methodology, writing–review and editing. J.H. Shim: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing.

This study was supported by grants from the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Science, (ICT grant no. NRF-2017R1E1A1A01074298, to J.H. Shim), and the research fund of Hanyang University (grant no. HY-201900000002619, to J. An).

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.
Juweid
ME
,
Cheson
BD
.
Positron emission tomography and assessment of cancer therapy
.
N Engl J Med
2006
;
354
:
496
507
.
2.
Park
JW
,
Kim
JH
,
Kim
SK
,
Kang
KW
,
Park
KW
,
Choi
JI
, et al
.
A prospective evaluation of 18F-FDG and 11C-acetate PET/CT for detection of primary and metastatic hepatocellular carcinoma
.
J Nucl Med
2008
;
49
:
1912
21
.
3.
Lee
JE
,
Jang
JY
,
Jeong
SW
,
Lee
SH
,
Kim
SG
,
Cha
SW
, et al
.
Diagnostic value for extrahepatic metastases of hepatocellular carcinoma in positron emission tomography/computed tomography scan
.
World J Gastroenterol
2012
;
18
:
2979
87
.
4.
Sugiyama
M
,
Sakahara
H
,
Torizuka
T
,
Kanno
T
,
Nakamura
F
,
Futatsubashi
M
, et al
.
18F-FDG PET in the detection of extrahepatic metastases from hepatocellular carcinoma
.
J Gastroenterol
2004
;
39
:
961
8
.
5.
Cho
Y
,
Lee
DH
,
Lee
YB
,
Lee
M
,
Yoo
JJ
,
Choi
WM
, et al
.
Does 18F-FDG positron emission tomography-computed tomography have a role in initial staging of hepatocellular carcinoma?
PLoS One
2014
;
9
:
e105679
.
6.
European Association for the Study of the Liver
.
Electronic address EEE, European Association for the study of the L. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma
.
J Hepatol
2018
;
69
:
182
236
.
7.
Hong
G
,
Suh
KS
,
Suh
SW
,
Yoo
T
,
Kim
H
,
Park
MS
, et al
.
Alpha-fetoprotein and (18)F-FDG positron emission tomography predict tumor recurrence better than Milan criteria in living donor liver transplantation
.
J Hepatol
2016
;
64
:
852
9
.
8.
Lin
CY
,
Liao
CW
,
Chu
LY
,
Yen
KY
,
Jeng
LB
,
Hsu
CN
, et al
.
Predictive value of 18F-FDG PET/CT for vascular invasion in patients with hepatocellular carcinoma before liver transplantation
.
Clin Nucl Med
2017
;
42
:
e183
7
.
9.
Bai
HX
,
Lee
AM
,
Yang
L
,
Zhang
P
,
Davatzikos
C
,
Maris
JM
, et al
.
Imaging genomics in cancer research: limitations and promises
.
Br J Radiol
2016
;
89
:
20151030
.
10.
Pinker
K
,
Shitano
F
,
Sala
E
,
Do
RK
,
Young
RJ
,
Wibmer
AG
, et al
.
Background, current role, and potential applications of radiogenomics
.
J Magn Reson Imaging
2018
;
47
:
604
20
.
11.
Jeong
WK
,
Jamshidi
N
,
Felker
ER
,
Raman
SS
,
Lu
DS
.
Radiomics and radiogenomics of primary liver cancers
.
Clin Mol Hepatol
2019
;
25
:
21
9
.
12.
Park
YJ
,
Shin
MH
,
Moon
SH
.
Radiogenomics based on PET imaging
.
Nucl Med Mol Imaging
2020
;
54
:
128
38
.
13.
Choi
H
,
Paeng
JC
,
Kim
DW
,
Lee
JK
,
Park
CM
,
Kang
KW
, et al
.
Metabolic and metastatic characteristics of ALK-rearranged lung adenocarcinoma on FDG PET/CT
.
Lung Cancer
2013
;
79
:
242
7
.
14.
Palaskas
N
,
Larson
SM
,
Schultz
N
,
Komisopoulou
E
,
Wong
J
,
Rohle
D
, et al
.
18F-fluorodeoxy-glucose positron emission tomography marks MYC-overexpressing human basal-like breast cancers
.
Cancer Res
2011
;
71
:
5164
74
.
15.
Kawada
K
,
Nakamoto
Y
,
Kawada
M
,
Hida
K
,
Matsumoto
T
,
Murakami
T
, et al
.
Relationship between 18F-fluorodeoxyglucose accumulation and KRAS/BRAF mutations in colorectal cancer
.
Clin Cancer Res
2012
;
18
:
1696
703
.
16.
Lee
JH
,
Park
JY
,
Kim
DY
,
Ahn
SH
,
Han
KH
,
Seo
HJ
, et al
.
Prognostic value of 18F-FDG PET for hepatocellular carcinoma patients treated with sorafenib
.
Liver Int
2011
;
31
:
1144
9
.
17.
Asman
Y
,
Evenson
AR
,
Even-Sapir
E
,
Shibolet
O
. [
18F]fludeoxyglucose positron emission tomography and computed tomography as a prognostic tool before liver transplantation, resection, and loco-ablative therapies for hepatocellular carcinoma
.
Liver Transpl
2015
;
21
:
572
80
.
18.
Kornberg
A
,
Kupper
B
,
Tannapfel
A
,
Buchler
P
,
Krause
B
,
Witt
U
, et al
.
Patients with non–[18 F]fludeoxyglucose-avid advanced hepatocellular carcinoma on clinical staging may achieve long-term recurrence-free survival after liver transplantation
.
Liver Transpl
2012
;
18
:
53
61
.
19.
Ahn
SM
,
Jang
SJ
,
Shim
JH
,
Kim
D
,
Hong
SM
,
Sung
CO
, et al
.
Genomic portrait of resectable hepatocellular carcinomas: implications of RB1 and FGF19 aberrations for patient stratification
.
Hepatology
2014
;
60
:
1972
82
.
20.
Hwang
HS
,
An
J
,
Kang
HJ
,
Oh
B
,
Oh
YJ
,
Oh
JH
, et al
.
Prognostic molecular indices of resectable hepatocellular carcinoma: Implications of S100P for early recurrence
.
Ann Surg Oncol
2021
;
28
:
6466
78
.
21.
Kang
HJ
,
Oh
JH
,
Chun
SM
,
Kim
D
,
Ryu
YM
,
Hwang
HS
, et al
.
Immunogenomic landscape of hepatocellular carcinoma with immune cell stroma and EBV-positive tumor-infiltrating lymphocytes
.
J Hepatol
2019
;
71
:
91
103
.
22.
Cancer Genome Atlas Research Network
.
Comprehensive and integrative genomic characterization of hepatocellular carcinoma
.
Cell
2017
;
169
:
1327
41
.
23.
Fujimoto
A
,
Furuta
M
,
Totoki
Y
,
Tsunoda
T
,
Kato
M
,
Shiraishi
Y
, et al
.
Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer
.
Nat Genet
2016
;
48
:
500
9
.
24.
Shiomi
S
,
Nishiguchi
S
,
Ishizu
H
,
Iwata
Y
,
Sasaki
N
,
Tamori
A
, et al
.
Usefulness of positron emission tomography with fluorine-18-fluorodeoxyglucose for predicting outcome in patients with hepatocellular carcinoma
.
Am J Gastroenterol
2001
;
96
:
1877
80
.
25.
Na
SJ
,
Oh
JK
,
Hyun
SH
,
Lee
JW
,
Hong
IK
,
Song
BI
, et al
.
(18)F-FDG PET/CT can predict survival of advanced hepatocellular carcinoma patients: a multicenter retrospective cohort study
.
J Nucl Med
2017
;
58
:
730
6
.
26.
Kim
YI
,
Paeng
JC
,
Cheon
GJ
,
Suh
KS
,
Lee
DS
,
Chung
JK
, et al
.
Prediction of posttransplantation recurrence of hepatocellular carcinoma using metabolic and volumetric indices of 18F-FDG PET/CT
.
J Nucl Med
2016
;
57
:
1045
51
.
27.
Gallagher
BM
,
Fowler
JS
,
Gutterson
NI
,
MacGregor
RR
,
Wan
CN
,
Wolf
AP
.
Metabolic trapping as a principle of oradiopharmaceutical design: some factors responsible for the biodistribution of [18F] 2-deoxy-2-fluoro-D-glucose
.
J Nucl Med
1978
;
19
:
1154
61
.
28.
Barbie
DA
,
Tamayo
P
,
Boehm
JS
,
Kim
SY
,
Moody
SE
,
Dunn
IF
, et al
.
Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1
.
Nature
2009
;
462
:
108
12
.
29.
Szklarczyk
D
,
Gable
AL
,
Lyon
D
,
Junge
A
,
Wyder
S
,
Huerta-Cepas
J
, et al
.
STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets
.
Nucleic Acids Res
2019
;
47
:
D607
D13
.
30.
Hoshida
Y
,
Nijman
SM
,
Kobayashi
M
,
Chan
JA
,
Brunet
JP
,
Chiang
DY
, et al
.
Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma
.
Cancer Res
2009
;
69
:
7385
92
.
31.
Boyault
S
,
Rickman
DS
,
de Reynies
A
,
Balabaud
C
,
Rebouissou
S
,
Jeannot
E
, et al
.
Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets
.
Hepatology
2007
;
45
:
42
52
.
32.
Lee
JS
,
Chu
IS
,
Heo
J
,
Calvisi
DF
,
Sun
Z
,
Roskams
T
, et al
.
Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling
.
Hepatology
2004
;
40
:
667
76
.
33.
Chiang
DY
,
Villanueva
A
,
Hoshida
Y
,
Peix
J
,
Newell
P
,
Minguez
B
, et al
.
Focal gains of VEGFA and molecular classification of hepatocellular carcinoma
.
Cancer Res
2008
;
68
:
6779
88
.
34.
Roessler
S
,
Jia
HL
,
Budhu
A
,
Forgues
M
,
Ye
QH
,
Lee
JS
, et al
.
A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients
.
Cancer Res
2010
;
70
:
10202
12
.
35.
Villanueva
A
,
Hoshida
Y
,
Battiston
C
,
Tovar
V
,
Sia
D
,
Alsinet
C
, et al
.
Combining clinical, pathology, and gene expression data to predict recurrence of hepatocellular carcinoma
.
Gastroenterology
2011
;
140
:
1501
12
.
36.
Wang
SM
,
Ooi
LL
,
Hui
KM
.
Identification and validation of a novel gene signature associated with the recurrence of human hepatocellular carcinoma
.
Clin Cancer Res
2007
;
13
:
6275
83
.
37.
Hay
N
.
Reprogramming glucose metabolism in cancer: can it be exploited for cancer therapy?
Nat Rev Cancer
2016
;
16
:
635
49
.
38.
Munster
P
,
Aggarwal
R
,
Hong
D
,
Schellens
JH
,
van der Noll
R
,
Specht
J
, et al
.
First-in-human phase I study of GSK2126458, an oral pan-class I phosphatidylinositol-3-kinase inhibitor, in patients with advanced solid tumor malignancies
.
Clin Cancer Res
2016
;
22
:
1932
9
.
39.
Bruix
J
,
Qin
S
,
Merle
P
,
Granito
A
,
Huang
YH
,
Bodoky
G
, et al
.
Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomized, double-blind, placebo-controlled, phase III trial
.
Lancet
2017
;
389
:
56
66
.
40.
Levine
AJ
,
Puzio-Kuter
AM
.
The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes
.
Science
2010
;
330
:
1340
4
.
41.
Weber
WA
.
Positron emission tomography as an imaging biomarker
.
J Clin Oncol
2006
;
24
:
3282
92
.
42.
Matter
MS
,
Decaens
T
,
Andersen
JB
,
Thorgeirsson
SS
.
Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends
.
J Hepatol
2014
;
60
:
855
65
.
43.
Zhu
AX
,
Chen
D
,
He
W
,
Kanai
M
,
Voi
M
,
Chen
LT
, et al
.
Integrative biomarker analyses indicate etiological variations in hepatocellular carcinoma
.
J Hepatol
2016
;
65
:
296
304
.
44.
Yamamoto
S
,
Huang
D
,
Du
L
,
Korn
RL
,
Jamshidi
N
,
Burnette
BL
, et al
.
Radiogenomic analysis demonstrates associations between (18)F-Fluoro-2-Deoxyglucose PET, prognosis, and epithelial–mesenchymal transition in non–small cell lung cancer
.
Radiology
2016
;
280
:
261
70
.
45.
Kaira
K
,
Serizawa
M
,
Koh
Y
,
Takahashi
T
,
Yamaguchi
A
,
Hanaoka
H
, et al
.
Biological significance of 18F-FDG uptake on PET in patients with non—small cell lung cancer
.
Lung Cancer
2014
;
83
:
197
204
.
46.
Kaira
K
,
Serizawa
M
,
Koh
Y
,
Takahashi
T
,
Hanaoka
H
,
Oriuchi
N
, et al
.
Relationship between 18F-FDG uptake on positron emission tomography and molecular biology in malignant pleural mesothelioma
.
Eur J Cancer
2012
;
48
:
1244
54
.
47.
Zhu
AX
,
Kudo
M
,
Assenat
E
,
Cattan
S
,
Kang
YK
,
Lim
HY
, et al
.
Effect of everolimus on survival in advanced hepatocellular carcinoma after failure of sorafenib: the EVOLVE-1 randomized clinical trial
.
JAMA
2014
;
312
:
57
67
.
48.
Adib
E
,
Klonowska
K
,
Giannikou
K
,
Do
KT
,
Pruitt-Thompson
S
,
Bhushan
K
, et al
.
Phase II clinical trial of everolimus in a pan-cancer cohort of patients with mTOR pathway alterations
.
Clin Cancer Res
2021
;
27
:
3845
53
.
49.
Treilleux
I
,
Arnedos
M
,
Cropet
C
,
Wang
Q
,
Ferrero
JM
,
Abadie-Lacourtoisie
S
, et al
.
Translational studies within the TAMRAD randomized GINECO trial: evidence for mTORC1 activation marker as a predictive factor for everolimus efficacy in advanced breast cancer
.
Ann Oncol
2015
;
26
:
120
5
.
50.
Zhu
AX
,
Kang
YK
,
Yen
CJ
,
Finn
RS
,
Galle
PR
,
Llovet
JM
, et al
.
Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased alpha-fetoprotein concentrations (REACH-2): a randomized, double-blind, placebo-controlled, phase III trial
.
Lancet Oncol
2019
;
20
:
282
96
.
51.
Grigg
SE
,
Sarri
GL
,
Gow
PJ
,
Yeomans
ND
.
Systematic review with meta-analysis: sirolimus- or everolimus-based immunosuppression following liver transplantation for hepatocellular carcinoma
.
Aliment Pharmacol Ther
2019
;
49
:
1260
73
.
52.
Schnitzbauer
AA
,
Filmann
N
,
Adam
R
,
Bachellier
P
,
Bechstein
WO
,
Becker
T
, et al
.
mTOR inhibition is most beneficial after liver transplantation for hepatocellular carcinoma in patients with active tumors
.
Ann Surg
2020
;
272
:
855
62
.