Skip Navigation
Skip to contents

JLC : Journal of Liver Cancer

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Liver Cancer > Volume 25(2); 2025 > Article
Original Article
Association between metabolites and hepatocellular carcinoma: findings from a two-sample Mendelian randomization study
Tung Hoang1orcid, Van Mai Truong2orcid, Tho Thi Anh Tran3,4orcid, Bao Le Thai Tran1orcid, Ngoc Hong Cao1orcid
Journal of Liver Cancer 2025;25(2):251-265.
DOI: https://doi.org/10.17998/jlc.2025.08.26
Published online: September 2, 2025

1Faculty of Pharmacy, University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam

2Faculty of Odonto-Stomatology, University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam

3Department of Gastroenterology and Hepatology, Nghe An Oncology Hospital, Nghe An, Vietnam

4Department of Oncology, Hanoi Medical University, Ha Noi, Vietnam

Corresponding author: Tung Hoang, Faculty of Pharmacy, University of Health Sciences, Vietnam National University Ho Chi Minh City, Y.A1 Administrative Building, Hai Thuong Lan Ong Street, Dong Hoa Ward, Ho Chi Minh City 700000, Vietnam E-mail: htung@uhsvnu.edu.vn
• Received: July 24, 2025   • Revised: August 14, 2025   • Accepted: August 26, 2025

© 2025 The Korean Liver Cancer Association.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 698 Views
  • 34 Downloads
prev next
  • Backgrounds/Aims
    Identifying metabolic biomarkers can enhance early detection and risk stratification of hepatocellular carcinoma (HCC). We conducted a two-sample Mendelian randomization (MR) study to assess the potential causal effects of metabolites on HCC risk.
  • Methods
    We performed meta-analyses to pool the effects of genetic instruments from 64 previously published genome-wide association studies. Summary statistics for HCC were obtained from a meta-analysis of the UK BioBank and FinnGen cohorts. MR analyses for the association between 3,275 metabolites and HCC risk were performed using inverse variance weighted, weighted median, MR-Egger, and MR-PRESSO methods to estimate the association. Enrichment analyses were performed on the significant metabolites to identify biological pathways associated with macronutrient intake.
  • Results
    We identified 99 metabolites that were positively and 36 metabolites that were negatively associated with HCC risk. Methyl glucopyranoside and phosphatidylcholine C38:3 were positively associated with HCC risk, whereas while 3-dehydrocarnitine and 10-undecenoate were inversely associated, with no evidence of heterogeneity, pleiotropy, or outlier effects for any of these associations. Pathway enrichment analysis showed that metabolites associated with increased HCC risk were primarily related to amino acid transport and solute carrier transporter disorders, whereas those linked to reduced risk were mainly involved in inositol and phosphatidylinositol metabolism, glycerophospholipid catabolism, and MeCP2-related regulatory processes.
  • Conclusions
    This comprehensive MR study identified several metabolites with potential causal roles in HCC development. Our findings highlight nutrient transport, lipid metabolism, and related regulatory mechanisms as key components of HCC pathogenesis, offering new avenues for biomarker discovery and therapeutic intervention.
Liver cancer is the sixth most common malignant tumor and the third leading cause of cancer-related mortality worldwide, with 865,269 new cases and 757,948 deaths reported in 2022.1 Hepatocellular carcinoma (HCC) accounts for approximately 90% of primary liver cancers, followed by intrahepatic cholangiocarcinoma and other less common liver malignancies.2,3 Nearly 90% of HCC cases are linked to chronic viral hepatitis, excessive alcohol consumption, and non-alcoholic steatohepatitis.2,3
Recent evidence highlights the critical role of metabolites, which are key substrates and products of metabolism essential for cellular processes, such as energy production, signal transduction, and apoptosis, in tumor development.4-6 Metabolic alterations were reported to contribute to cancer-associated metabolic reprogramming, influencing gene expression, cellular differentiation, and the tumor microenvironment through six key mechanisms: 1) dysregulated glucose and amino acid uptake, 2) alternative nutrient acquisition pathways, 3) utilization of glycolysis/tricarboxylic acid cycle intermediates for biosynthesis and nicotinamide adenine dinucleotide phosphate production, 4) increased nitrogen demand, 5) metabolite-driven gene regulation, and 6) metabolic interactions within the tumor microenvironment.5,6 Given their role in cancer progression, metabolites, particularly those detectable in plasma and serum, are promising biomarkers for screening, diagnosis, and assessment of therapeutic responses owing to their convenience and minimally invasive nature.7 However, metabolic heterogeneity is influenced by genetic mutations, cell and tissue origin, and epigenetic regulation.8
A previous observational study identified 150 metabolites that were significantly altered in patients with HCC compared with controls.9 Notably, a panel of six metabolites, including alpha-fetoprotein, 6-bromotryptophan, N-acetylglycine, salicyluric glucuronide, testosterone sulfate, and age, demonstrated strong discriminatory performance in distinguishing early stage HCC cases from controls.9 Furthermore, a recent meta-analysis of 68 cohort studies identified nearly 600 metabolites showing significant variations across different stages of liver disease progression from healthy controls to liver cirrhosis and HCC.10 Despite these findings, most studies remain observational, limiting their ability to establish causal relationships owing to confounding factors, reverse causation, and selection bias.11 Furthermore, conventional epidemiological approaches and randomized controlled trials (RCTs) are yet to provide conclusive evidence on the role of metabolic markers in reducing the incidence of HCC. In this context, genome-wide association studies (GWAS) of metabolites offer an opportunity to identify genetic instruments for Mendelian randomization (MR), a method that can strengthen causal inference in the absence of large-scale RCTs.12 Therefore, this study aimed to screen for genetically predicted metabolites associated with HCC development and progression, using robust genetic and metabolomic approaches.
Study design
This study employed a two-sample MR design that relied on three key assumptions.13 First, the relevance assumption requires that the selected genetic variants are strongly associated with the metabolites.13 Second, the independence assumption states that there should be no unmeasured confounders affecting the relationship between genetic variants and HCC risk.13 Third, the exclusion restriction assumption ensures that genetic variants influence HCC risk solely through their effect on metabolites, with no alternative pathways involved.13
Data source
Exposure data summary statistics were retrieved from published GWAS on metabolites using the MetaboAnalyst database (https://www.mgwas.ca/mGWAS/upload/MGBrowseView.xhtml). We collected data for 337,881 genetic variants significantly associated with over 4,000 metabolite levels from 64 GWAS, excluding those from the UK Biobank. After removing variants with missing allele information and a zero-standard error, we performed a random-effects meta-analysis across 64 independent GWAS to derive pooled effects of each variant on metabolites, accounting for between-study variability. This integration across multiple cohorts, rather than relying on a single study, provided stronger and more reliable instrumental variables by increasing the sample size and enhancing the statistical power. Accordingly, summary statistics were obtained for 186,348 significant variants from which F-statistics were calculated; 183,631 strong instruments remained after excluding weak instruments (F-statistics ≤10). Additionally, the HCC meta- analysis integrated 32,342,897 variants from FinnGen and UK Biobank. By harmonizing the metabolite summary statistics with those for HCC through single nucleotide polymorphism (SNP)-based merging, we identified 147,562 variants that served as instrumental variables for assessing the MR association between 3,275 metabolites and HCC (Fig. 1).
Statistical analysis
For the primary analysis, we employed the inverse-variance weighted (IVW) method under the assumption of balanced pleiotropy.14 This approach utilized SNP-specific Wald estimates, which were calculated by dividing the SNP-outcome association by the SNP-exposure association. These estimates were then combined using a random effects model to account for potential heterogeneity. To ensure the robustness of our findings, we conducted additional sensitivity analyses using the weighted median method15 and the MR-Egger regression.16 While the IVW method provides the most precise estimates when all instrumental variables are valid, the weighted median method offers a compromise between efficiency and robustness, allowing for invalid instruments. Additionally, we applied MR-Egger regression to estimate the pleiotropy-adjusted causal effect estimate, which remains valid even when all instruments have direct effects on the outcome, although this method is less efficient than IVW.14-16
We used multiple complementary approaches to assess horizontal pleiotropy. First, we calculated the MR-Egger intercept and its associated P-value, where a statistically significant deviation from zero indicated the presence of unbalanced horizontal pleiotropy.16 Second, we performed MR pleiotropy residual sum and outlier (MR-PRESSO) analyses, which detect and correct for horizontal pleiotropy by identifying outlier genetic variants.17 For this analysis, we reported the raw P-value for pleiotropy detection, outlier-corrected P-value after removing pleiotropic SNPs, and global test P-value, which evaluates the overall presence of horizontal pleiotropy in the instrument set. All MR analyses were performed using R statistical Software (R Foundation, Vienna, Austria).18
To further investigate the mechanisms linking metabolites to HCC, we conducted enrichment analyses of metabolic and lipid pathways related to HCC using an integrated KEGG pathway via the Human Metabolome Database, Reactome, and WikiPathways Databases. The analyses were performed using the online platform MetaboAnalyst (version 6.0). By utilizing a set of significant metabolites, we identified critical pathways that may play a role in HCC development and progression, offering valuable biological insights into the underlying processes.19
Metabolites positively associated with HCC risk and underlying pathways
Table 1 presents the associations between genetic predisposition to metabolites and increased risk of HCC. Using the IVW method, 257 metabolites were identified, 99 of which remained significant after sensitivity analyses using the median-weighted and MR-Egger methods.
Among the most significant metabolites, methyl glucopyranoside (α+β) showed a strong association with HCC risk (odds ratio [OR], 30.24; 95% confidence interval [CI], 19.65-46.54; false discovery rate [FDR] P=3.76×10-54 by the IVW method) and demonstrated high instrument validity, with no evidence of heterogeneity (I2=0%, Cochran’s Q P>0.999) and no indication of directional horizontal pleiotropy (MR-Egger intercept P=0.087; MR-PRESSO global P>0.99). Similarly, phosphatidylcholine (acyl-alkyl) C38:3 was also strongly associated with increased HCC risk (OR, 1.62; 95% CI, 1.50-1.74; FDR P=1.07×10-35), with no heterogeneity (I2=0%, Cochran’s Q P=0.994) and no evidence of directional horizontal pleiotropy (MR-Egger intercept P=0.101; MR-PRESSO global P=0.998) (Supplementary Table 1).
Enrichment analysis highlighted RaMB-DB pathways related to amino acid transport defects as the most significant pathways, followed by solute carrier (SLC) transporter disorders, amino acid transport across the plasma membrane, and Na+/Cl- dependent neurotransmitter transporter pathways (Fig. 2).
Metabolites negatively associated with HCC risk and underlying pathways
Table 2 summarizes the associations between genetic predisposition to metabolites and a reduced risk of HCC. The IVW method identified 218 metabolites, with 36 remaining significant after sensitivity analyses using the median-weighted and MR-Egger methods.
Among the most significant metabolites, cholesteryl ester (20:4) showed a strong inverse association with HCC risk (OR, 0.02; 95% CI, 0.01-0.03; FDR P=7.77×10-63 by the IVW method) and demonstrated high instrument validity, with no evidence of heterogeneity (I2=0%, Cochran’s Q P=0.996) and no indication of directional horizontal pleiotropy (MR-Egger intercept P=0.101; MR-PRESSO global P>0.999). Similarly, 10-undecenoate was also strongly associated with decreased HCC risk (OR, 0.93; 95% CI, 0.91-0.95; FDR P=4.36×10-8), with no heterogeneity (I2=0%, Cochran’s Q P>0.999) and no evidence of directional horizontal pleiotropy (MR-Egger intercept P=0.166; MR-PRESSO global P>0.999) (Supplementary Table 2).
Enrichment analysis revealed that inositol transporters were the most significant RaMB-DB pathway, followed by the synthesis of phosphatidylinositol, glycerophospholipid catabolism, and MeCP2 and associated Rett syndrome pathways (Fig. 3).
This study identified genetic predispositions to specific 135 metabolites that were significantly associated with either an increased or decreased risk of HCC. Among the most significant metabolites, methyl glucopyranoside (α+β) and phosphatidylcholine (acyl-alkyl) C38:3 were positively associated with HCC risk, whereas 3-dehydrocarnitine and 10-undecenoate were inversely associated, with all associations showing no evidence of heterogeneity, pleiotropy, or outlier influence. Enrichment analysis revealed that the shared significant metabolites associated with increased HCC risk were primarily related to amino acid transport and SLC transporter disorders, whereas those linked to reduced risk were mainly involved in inositol and phosphatidylinositol metabolism, glycerophospholipid catabolism, and MeCP2-related regulatory processes.
Recently, several studies have employed MR methods to investigate the relationship between metabolites and HCC. These studies utilized summary statistics data for metabolites derived from publicly available GWAS. For instance, Tang et al.20 conducted an MR analysis using GWAS data encompassing 1,400 plasma metabolites and HCC. Their findings revealed that 36 metabolites were significantly associated with HCC in East Asians, including 21 that were linked to an increased incidence risk, while 15 were associated with overall HCC risk.20 Similarly, a two-sample MR analysis was performed to examine 137 metabolites as potential causal mediators of five common gastrointestinal cancers.21 Their study showed that higher levels of isovalerylcarnitine were associated with an elevated risk of liver cancer in Europeans.21 More recently, Lin Ning conducted a comprehensive MR analysis to examine the associations between 1,400 systemic metabolites and the risk of HCC and cholangiocarcinoma.22 In this study, we identified 19 metabolites with significant links to these cancers. Among them, nine metabolites were associated with increased risk, including bilirubin (E, Z, Z, E), bilirubin (Z, Z) to taurocholate ratio, dimethylarginine (sdma+adma), N-methyltaurine, 4-vinylguaiacol sulfate, cholate-to-cAMP ratio, glycohyocholate, cholesterol, and 4-methylguaiacol sulfate.22 Additionally, ten metabolites, including ursodeoxycholate, 3-hydroxybutyroylglycine, linoleoylcholine, nonanoylcarnitine (C9), pristanate, heptenedioate (C7:1-DC), mannonate, N-acetyl-L-glutamine, sphinganine, and N-lactoyl isoleucine have been reported as protective metabolites.22 In contrast, a study by Ma used a bidirectional MR approach to assess the causal relationships between 21 amino acids and the risk of primary liver cancer.23 These findings indicated no causal association between amino acids and primary liver cancer (PLC) risk, although PLC was found to influence serine concentration.23 However, previous studies have notable limitations, primarily due to their reliance on summary statistics derived from a single GWAS for either metabolites or HCC. This approach may reduce the statistical power and limit the generalizability of the findings, especially when the sample sizes are relatively small. In contrast, our study addressed these limitations by integrating data from 64 independent GWASs for metabolites, thereby capturing a broader and more diverse range of genetic variation. Additionally, we utilized pooled GWAS data for HCC from both the UK Biobank and Finngen, which enhances the statistical power and reliability. This comprehensive approach strengthens the robustness of our findings and allows for more accurate identification of metabolite-HCC associations across populations.
Metabolic reprogramming has emerged as a central topic in cancer biology, reflecting dynamic alterations in cellular metabolism that support tumor growth and progression.24-28 One of the earliest and most well-established examples is the Warburg effect, where cancer cells preferentially convert glucose into lactate through aerobic glycolysis, bypassing mitochondrial oxidative phosphorylation even in the presence of sufficient oxygen.29,30 This metabolic shift not only facilitates rapid energy production, but also supports the biosynthetic and redox needs of proliferating tumor cells, underscoring its critical role in oncogenesis. In our study, methyl glucopyranoside level was positively associated with the risk of HCC. This metabolite has been previously linked to cancer biology, with significant differences observed between non-small cell lung cancer patients with poor and good survival outcomes in a cohort of 220 advanced-stage Caucasian patients,31 and MR analyses indicated a positive association with malignant neoplasms of bone and joint cartilage.32 Experimental evidence further supports its biological relevance, as methyl 6-O-cinnamoyl-α-D-glucopyranoside has been shown to ameliorate acute liver injury by inhibiting oxidative stress via Nrf2 signaling activation.33 Similarly, our finding of a positive association between phosphatidylcholine (acyl-alkyl) C38:3 and HCC aligns with prior reports of elevated phosphatidylcholine levels in HCC patients compared with cirrhotic controls.34 This observation was supported by the dysregulation of phosphatidylcholine synthesis enzymes in HCC, including the upregulation of choline kinase alpha and downregulation of phosphatidylethanolamine N-methyltransferase, the latter preferentially producing long-chain polyunsaturated phosphatidylcholines known for their anti-inflammatory properties.34 A specific decrease in phosphatidylcholines in HCC tissues may reflect a disruption of protective lipid-mediated pathways, further highlighting the potential mechanistic links between altered lipid metabolism and hepatocarcinogenesis.34
In contrast, our findings indicate that elevated 3-dehydrocarnitine levels are linked to a reduced likelihood of HCC. As a breakdown product of long-chain carnitines, higher 3-dehydrocarnitine levels may signal enhanced catabolism of these molecules, which normally transport fatty acids into mitochondria for energy generation.35 The reduced availability of long-chain carnitines could limit the mitochondrial energy supply in cancer cells, potentially slowing their growth. This pattern of carnitine alteration aligns with reports that disturbances in fatty acid transport and metabolism are involved in HCC development.35 In addition, our study found an inverse association between 10-undecenoate and HCC. Although direct evidence in the literature is limited, its related compound, undecylenic acid, an 11-carbon monounsaturated fatty acid derived from the distillation of castor oil via pyrolysis,36 has been linked to both the Healthy Eating Index and Dietary Approaches to Stop Hypertension dietary indices,37 which are associated with a lower risk of HCC.38
In this study, enrichment analysis revealed that the top positively enriched metabolite pathways were largely associated with impaired SLC-mediated transport, a mechanism previously implicated in metabolic imbalance and liver tumorigenesis in earlier studies. SLC transporters are essential for maintaining the metabolic balance in the liver, and their dysregulation contributes to tumor progression.39 Several SLCs are implicated in HCC: SLC26A6 is overexpressed and linked to cancer-related pathways, hOSCP1 variants increase non-viral liver cancer risk, SLC2A1 is upregulated while SLC2A2 is downregulated, correlating with prognosis, SLC13A5 supports energy homeostasis and hepatoma proliferation, SLCO2A1 is elevated in HCC, while SLCO1B1 expression decreases with cancer grade, and SLC46A3 shows increased expression in HCC.40 In contrast, our study revealed that the metabolites negatively associated with HCC were mainly enriched in pathways related to inositol transport and phospholipid metabolism. This is supported by biological evidence showing that myo-inositol and its derivatives regulate cell signaling, lipid homeostasis, and hepatic insulin sensitivity, thereby exerting anti-inflammatory and antitumor effects in the liver.41 Epidemiological studies have also linked higher dietary inositol intake and better phospholipid profile with a reduced risk of non-alcoholic fatty liver disease,42,43 and may thus lead to HCC development.
Despite its strengths, the main limitation is that our MR analyses mainly relied on GWAS summary statistics from European-ancestry populations, which may limit the generalizability to other ethnic groups given differences in metabolite profiles and HCC epidemiology. For example, with population-attributable fractions of more than 40% for hepatic B virus and 19% for alcohol consumption, chronic hepatitis B virus infection is more prevalent in East Asian populations (69%), whereas non-alcoholic fatty liver disease and alcohol-related liver disease predominate in Western cohorts (up to 35%), leading to different baseline HCC risks.44-46 In addition, genetic variants influencing metabolite levels (e.g., SRD5A2 gene) may differ in allele frequency and linkage disequilibrium patterns across ancestries, potentially modifying SNP-metabolite effect estimates and altering the strength or direction of MR pathways.47 Likewise, associations between genetic variants and HCC may vary with population-specific exposures, such as hepatic B virus prevalence, aflatoxin contamination, dietary patterns, and gut microbiome composition, affecting SNP-outcome estimates and the derived causal effects.48 These cross-population differences in both the SNP-metabolite and SNP-HCC associations highlight the need for validation in non-European cohorts. However, large-scale metabolite and HCC GWAS in non-European populations remains scarce; future multi-ethnic and cross-ancestry MR studies are warranted to confirm and extend our findings. Finally, of the 475 metabolites identified as significant by the IVW after excluding weak instruments, only 135 remained in concordance with the weighted median and MR-Egger analyses. This stringent cross-method confirmation, while increasing confidence by reducing potential bias from horizontal pleiotropy, may also have excluded true associations with weaker or less consistent signals, potentially underestimating the full spectrum of metabolite-HCC relationships.
In summary, this study uncovered genetic predispositions to specific metabolites that were significantly associated with either an elevated or a reduced risk of HCC. Enrichment analysis indicated that metabolites associated with higher HCC risk were mainly involved in oxidative pathways, whereas those linked to lower risk were primarily related to transporter-mediated functions.

Acknowledgement

Tho Thi Anh Tran was funded by the Master’s, PhD Scholarship Program of Vingroup Innovation Foundation (VINIF), code VINIF.2023.TS.119.

Conflicts of Interest

The authors declare no competing interests.

Ethics Statement

This study utilized publicly available summary-level data and therefore did not require ethical approval.

Funding Statement

This research was also funded by Vietnam National University Ho Chi Minh City (VNU-HCM) under the grant number C2024-44-34.

Data Availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Author Contributions

Conceptualization: TH, VMT, TTAT, BLTT, NHC

Data Curation: TH

Formal Analysis: TH

Methodology: TH, VMT, TTAT, BLTT, NHC

Writing - original draft: TH

Writing - review & editing: TH, BLTT, NHC

Supplementary data can be found with this article online https://doi.org/10.17998/jlc.2025.08.26.
Figure 1.
Flowchart for identification of instrumental variables.
jlc-2025-08-26f1.jpg
Figure 2.
Enriched pathways of metabolites positively associated with hepatocellular carcinoma. IEM, inborn errors of metabolism; SLC, solute carrier.
jlc-2025-08-26f2.jpg
Figure 3.
Enriched pathways of metabolites negatively associated with hepatocellular carcinoma. PI, phosphatidylinositol; MECP2, methyl- CpG-binding protein 2; SLC, solute carrier.
jlc-2025-08-26f3.jpg
jlc-2025-08-26f4.jpg
Table 1.
Genetically metabolites positively associated with hepatocellular carcinoma risk
Metabolite Number of instruments Median F-statistics (range) Inverse-variance weighted
Heterogeneity
OR (95% CI) Raw P-value FDR P-value I2 (%) P-value
Valylarginine 65 57.89 (42.74-67.40) 1.27 (1.15-1.40) 1.81E-06 2.84E-05 0.0 >0.999
Free carnitine (C0) 80 56.90 (41.30-67.40) 1.15 (1.11-1.19) 6.48E-15 2.33E-13 0.0 >0.999
Propionyl-carnitine 7 30.82 (30.12-32.51) 1.45 (1.23-1.70) 9.49E-06 0.0001 37.3 0.144
X-23314 1,100 35.00 (21.57-67.40) 1.67 (1.55-1.79) 4.63E-44 6.32E-42 74.0 <0.001
Methyl glucopyranoside (α+β) 69 39.80 (30.20-67.40) 30.24 (19.65-46.54) 3.76E-54 8.8E-52 0.0 >0.999
Tyrosine 67 58.00 (29.75-67.40) 4.29 (1.94-9.46) 0.0003 0.003 37.5 0.001
Large VLDL particles 63 47.26 (33.21-62.34) 1.15 (1.05-1.26) 0.003 0.024 0.0 0.972
Small VLDL-cholesterol 25 58.77 (39.14-67.40) 2.94 (2.02-4.30) 2.26E-08 4.27E-07 19.4 0.193
Large VLDL-triglycerides 20 61.76 (37.74-64.22) 3.84 (2.55-5.78) 1.3E-10 2.9E-09 0.0 0.650
Double bond protons of mobile lipids (MobCH) 104 34.93 (29.82-60.98) 3.98 (2.70-5.86) 2.69E-12 6.99E-11 94.1 <0.001
Medium VLDL particles 273 47.41 (34.11-126.55) 1.16 (1.11-1.21) 1.82E-10 3.99E-09 5.4 0.250
Serum total triglycerides 481 35.34 (29.73-77.93) 1.48 (1.35-1.61) 4.61E-18 2.1E-16 73.3 <0.001
Small VLDL particles 43 42.76 (32.69-60.98) 1.35 (1.18-1.56) 2.39E-05 <0.001 36.3 0.011
Large VLDL total lipids 20 52.33 (34.62-62.67) 1.56 (1.26-1.93) 4.52E-05 0.001 49.4 0.007
Small VLDL-free cholesterol 143 42.22 (10.65-67.40) 1.47 (1.37-1.57) 1.17E-29 7.8E-28 14.4 0.084
Extra-small VLDL phospholipids 1,570 40.10 (10.06-67.40) 1.15 (1.12-1.18) 1.29E-29 8.47E-28 0.0 0.998
Medium VLDL total lipids 31 28.74 (19.57-57.77) 12.15 (6.58-22.44) 1.49E-15 6.03E-14 0.0 0.712
Large VLDL-free cholesterol 1,181 40.04 (16.06-137.87) 1.27 (1.23-1.32) 1.04E-38 1.06E-36 19.8 <0.001
FAw67 1,165 40.90 (11.04-118.53) 1.25 (1.21-1.30) 1.18E-31 8.05E-30 21.1 <0.001
Large VLDL-cholesterol 1,143 40.62 (13.55-108.54) 1.37 (1.30-1.44) 1.17E-33 9.35E-32 58.6 <0.001
Large VLDL-cholesteryl esters 1,085 40.98 (13.24-107.47) 1.43 (1.35-1.52) 8.56E-33 6.37E-31 67.2 <0.001
Very-large VLDL total lipids 1,142 39.13 (11.45-107.18) 1.34 (1.28-1.40) 3.48E-36 3.36E-34 41.0 <0.001
Medium VLDL-cholesterol 941 38.10 (11.60-121.37) 1.43 (1.32-1.55) 4.59E-19 2.15E-17 79.5 <0.001
IDL-triglycerides 1,166 39.37 (29.72-111.42) 1.32 (1.27-1.37) 1.84E-49 3.17E-47 13.1 <0.001
Extra-small VLDL particles 1,073 40.82 (29.72-108.62) 1.34 (1.29-1.38) 2.2E-55 5.53E-53 9.3 <0.001
Extra-small VLDL total lipids 1,214 38.80 (21.07-118.66) 1.32 (1.27-1.37) 6.2E-50 1.13E-47 13.5 <0.001
Very-large VLDL-triglycerides 1,076 41.22 (24.59-110.08) 1.32 (1.28-1.37) 8.26E-53 1.69E-50 9.2 0.011
Linoleic acid 1,059 41.48 (22.73-108.46) 1.35 (1.30-1.40) 7.66E-54 1.67E-51 17.4 <0.001
Medium VLDL-triglycerides 1,092 40.29 (19.99-105.04) 1.32 (1.27-1.38) 1.16E-41 1.31E-39 22.3 <0.001
Medium VLDL phospholipids 869 45.19 (11.43-98.92) 1.3 (1.19-1.42) 5.24E-09 1.05E-07 82.2 <0.001
Apolipoprotein B 806 40.14 (15.58-108.71) 1.43 (1.31-1.57) 2.92E-15 1.12E-13 81.4 <0.001
Extremely-large VLDL particles 919 48.03 (21.68-90.05) 1.28 (1.17-1.40) 4.75E-08 8.69E-07 82.7 <0.001
Very-large VLDL phospholipids 586 41.69 (12.37-87.83) 1.35 (1.20-1.51) 4.54E-07 7.59E-06 84.7 <0.001
Small VLDL phospholipids 842 45.81 (18.71-88.74) 1.36 (1.25-1.48) 2.65E-12 6.94E-11 81.1 <0.001
Extra-small VLDL-triglycerides 782 41.10 (22.89-74.27) 1.35 (1.21-1.50) 5.09E-08 9.25E-07 84.8 <0.001
Medium VLDL-free cholesterol 728 40.11 (17.74-99.09) 1.27 (1.15-1.40) 3.02E-06 4.6E-05 81.8 <0.001
Large VLDL phospholipids 1,207 41.11 (29.73-219.46) 1.27 (1.23-1.32) 1.92E-38 1.9E-36 16.8 <0.001
Very-large VLDL particles 741 42.30 (24.57-132.06) 1.27 (1.21-1.32) 4.08E-27 2.52E-25 29.8 <0.001
Extremely-large VLDL-triglycerides 387 41.21 (19.57-77.15) 1.21 (1.14-1.28) 6.21E-12 1.57E-10 0.0 >0.999
Extremely-large VLDL total lipids 1,091 38.00 (29.72-108.96) 1.31 (1.26-1.36) 8.28E-44 1.08E-41 11.4 0.002
Very-large HDL-triglycerides 979 39.35 (29.73-109.47) 1.32 (1.28-1.38) 2.83E-48 4.64E-46 10.4 0.007
Medium VLDL-cholesteryl esters 980 39.57 (29.72-106.04) 1.31 (1.26-1.36) 1.82E-44 2.6E-42 11.2 0.004
Small VLDL-triglycerides 972 39.59 (29.75-103.96) 1.32 (1.27-1.37) 4.5E-46 6.69E-44 12.4 0.001
Small VLDL total lipids 943 36.68 (29.74-91.84) 1.37 (1.32-1.43) 1.4E-50 2.7E-48 11.8 0.003
C-glycosyltryptophan/hydroxyisovalerylcarnitine 1,205 44.00 (13.96-108.59) 1.44 (1.32-1.56) 2.87E-18 1.32E-16 84.9 <0.001
Valine/isovalerylcarnitine 1,058 43.27 (17.25-125.91) 1.73 (1.59-1.88) 7.76E-39 8.2E-37 84.0 <0.001
Large LDL phospholipids 1,009 50.52 (13.56-101.08) 1.29 (1.19-1.40) 6.37E-10 1.35E-08 83.5 <0.001
Small LDL total lipids 831 40.87 (16.22-97.78) 1.39 (1.27-1.52) 9.66E-13 2.73E-11 83.6 <0.001
Large LDL-cholesterol 849 41.31 (15.17-115.54) 1.36 (1.25-1.49) 7.29E-12 1.82E-10 83.5 <0.001
IDL total lipids 998 49.19 (15.36-99.44) 1.25 (1.15-1.36) 7.44E-08 1.33E-06 83.1 <0.001
Medium LDL-cholesterol 726 40.11 (13.12-99.04) 1.26 (1.15-1.39) 1.92E-06 2.96E-05 83.3 <0.001
LDL-cholesterol 147 44.23 (29.76-67.40) 1.05 (1.03-1.07) 9.75E-06 0.0001 0.0 >0.99
IDL phospholipids 198 34.26 (29.76-67.40) 2.02 (1.50-2.72) 3.74E-06 5.64E-05 94.0 <0.001
Large LDL-cholesteryl esters 114 41.58 (29.74-111.54) 4.29 (2.96-6.22) 1.26E-14 4.37E-13 95.1 <0.001
IDL-free cholesterol 402 41.14 (12.25-107.19) 2.15 (1.85-2.49) 9.3E-24 5.25E-22 91.0 <0.001
Medium LDL particles 54 39.20 (10.72-60.98) 1.18 (1.08-1.30) 0.001 0.005 0.0 0.991
Large LDL particles 57 41.05 (29.92-49.76) 1.33 (1.21-1.48) 2.17E-08 4.13E-07 0.0 >0.999
IDL particles 35 60.18 (39.03-63.56) 1.59 (1.28-1.98) 2.41E-05 0.0003 0.0 0.476
Medium LDL-cholesteryl esters 179 38.34 (29.73-66.03) 1.11 (1.03-1.20) 0.006 0.041 0.0 0.999
Serum total cholesterol 965 41.03 (17.31-112.42) 1.32 (1.27-1.38) 1.12E-43 1.41E-41 13.3 0.001
Medium LDL total lipids 819 40.89 (10.49-102.57) 1.41 (1.33-1.49) 1.08E-34 9.09E-33 50.8 <0.001
Small LDL-cholesterol 788 40.27 (10.30-96.91) 1.71 (1.57-1.86) 2.26E-35 2.06E-33 78.4 <0.001
IDL-cholesterol 943 39.89 (21.32-108.34) 1.61 (1.49-1.74) 2.16E-33 1.68E-31 75.6 <0.001
Small LDL particles 1,033 42.13 (27.79-114.84) 1.58 (1.46-1.70) 1.13E-31 7.91E-30 78.8 <0.001
Esterified cholesterol 1,115 40.94 (13.42-111.12) 1.47 (1.36-1.59) 4.38E-23 2.39E-21 81.8 <0.001
Large LDL total lipids 1,092 42.67 (10.93-109.13) 1.40 (1.30-1.51) 4.01E-19 1.9E-17 80.7 <0.001
Medium LDL phospholipids 1,178 44.53 (11.83-121.88) 1.37 (1.27-1.47) 3.95E-17 1.77E-15 80.3 <0.001
Large LDL-free cholesterol 927 44.84 (12.34-98.35) 1.35 (1.24-1.48) 1.76E-11 4.18E-10 84.0 <0.001
Total fatty acids 1,039 45.18 (13.94-104.31) 1.42 (1.30-1.55) 1.29E-14 4.43E-13 86.8 <0.001
Glycine 966 39.39 (29.72-97.33) 1.63 (1.50-1.76) 1.11E-32 8.07E-31 77.7 <0.001
3-ureidopropionate 6 55.11 (38.93-67.40) 1.99 (1.33-2.97) 0.001 0.008 11.0 0.345
Lysine 22 60.29 (40.51-67.40) 3.24 (2.05-5.14) 5.37E-07 8.84E-06 26.2 0.128
Monounsaturated fatty acids 18 53.15 (41.84-58.72) 5.27 (3.46-8.02) 8.39E-15 2.95E-13 0.0 0.999
1-ribosyl-imidazoleacetate 17 66.03 (45.08-67.40) 5.21 (3.39-8.01) 5.34E-14 1.72E-12 0.0 0.999
otPUFA 19 61.40 (37.60-67.40) 4.17 (2.84-6.13) 3.11E-13 9.02E-12 0.0 0.834
X-21343 18 59.92 (52.26-67.40) 4.53 (3.08-6.66) 1.68E-14 5.67E-13 0.0 0.995
Phosphatidylcholine 37:4/Phosphatidylethanolamine 40:3 [M+H]+ 268 36.25 (29.78-109.10) 2.00 (1.64-2.45) 1.35E-11 3.26E-10 91.5 0.000
Phosphatidylcholine (acyl-alkyl) C38:3 (PC ae C38:3) 301 50.26 (29.83-67.40) 1.62 (1.50-1.74) 1.07E-35 9.98E-34 0.0 0.994
Phosphatidylcholine (diacyl) C32:2 (PC aa C32:2) 126 47.19 (29.72-67.40) 4.62 (2.56-8.35) 3.7E-07 6.25E-06 56.6 <0.001
Omega-3 fatty acids 61 32.22 (29.73-66.03) 1.18 (1.12-1.25) 2.75E-09 5.63E-08 18.2 0.115
Docosahexaenoic acid 57 33.03 (29.74-46.17) 1.10 (1.03-1.17) 0.003 0.025 0.0 0.959
X-14473 121 48.64 (29.81-67.40) 1.49 (1.18-1.89) 0.001 0.009 21.6 0.022
X-11538 167 56.20 (29.88-66.03) 1.07 (1.05-1.10) 1.09E-07 1.91E-06 0.0 0.921
X-21849 47 43.56 (31.35-47.48) 38.75 (26.11-57.52) 1.2E-73 6.57E-71 0.0 >0.999
X-02249 48 32.58 (29.82-67.40) 1.14 (1.08-1.20) 6.3E-06 9.34E-05 0.0 0.986
Glutamine 245 30.29 (29.78-43.89) 1.18 (1.15-1.21) 3.74E-41 4.09E-39 0.0 >0.999
C-glycosyltryptophan/succinylcarnitine 98 32.49 (29.85-67.40) 1.08 (1.05-1.12) 3.64E-07 6.18E-06 0.0 >0.999
Triacylglycerol 51:3 [M+NH4]+ 1,339 40.59 (10.31-122.11) 1.43 (1.39-1.48) 1.8E-99 2.01E-96 17.2 <0.001
Triacylglycerol 53:4 [M+NH4]+ 582 35.16 (14.83-65.23) 1.63 (1.43-1.85) 9.4E-14 2.87E-12 84.3 <0.001
Diacylglycerol 36:3 [M+NH4]+ 528 37.27 (14.46-65.23) 1.43 (1.28-1.61) 1.17E-09 2.43E-08 79.7 <0.001
Triacylglycerol 50:4 [M+NH4]+ 719 43.86 (26.58-61.81) 1.39 (1.25-1.55) 1.12E-09 2.33E-08 82.6 <0.001
Triacylglycerol 54:5 [M+NH4]+ 791 43.45 (26.61-65.23) 1.21 (1.10-1.33) <0.001 0.002 80.2 <0.001
Triacylglycerol 50:3 [M+NH4]+ 979 44.61 (13.35-128.45) 1.43 (1.33-1.54) 2.42E-21 1.24E-19 79.4 <0.001
Diacylglycerol 34:3 [M+H-H2O]+ 981 44.44 (26.80-134.77) 1.40 (1.30-1.51) 3.04E-19 1.46E-17 79.8 <0.001
FAw9S 1,158 36.25 (18.80-111.57) 1.43 (1.34-1.52) 3.54E-28 2.27E-26 73.5 <0.001
X-12822 1,173 41.50 (16.12-111.13) 1.41 (1.32-1.50) 5.56E-26 3.37E-24 78.1 <0.001
Glycolate 160 36.16 (29.72-67.40) 1.54 (1.26-1.88) 2.65E-05 0.0003 75.9 <0.001
7-methylguanine 354 31.90 (29.72-66.03) 1.44 (1.21-1.71) 4.46E-05 0.0006 85.7 <0.001
Sphingomyelin 32:1 [M+H]+ 660 37.26 (29.72-67.40) 1.17 (1.05-1.29) 0.004 0.030 76.5 <0.001

OR, odds ratio; CI, confidence interval; FDR, false discovery rate; VLDL, very low density lipoprotein; IDL, intermediate-density lipoprotein; HDL, high-density lipoprotein; LDL, low-density lipoprotein.

Table 2.
Genetically metabolites negatively associated with hepatocellular carcinoma risk
Metabolite Number of instruments Median F-statistics (range) Inverse-variance weighted
Heterogeneity
OR (95% CI) Raw P-value FDR P-value I2 (%) P-value
1-stearoylglycero-phosphoethanolamine 24 46.34 (29.93-67.40) 0.63 (0.47-0.83) 0.001 0.009 25.6 0.125
10-undecenoate (11:1n1) 220 44.42 (29.72-67.40) 0.93 (0.91-0.95) 2.12E-09 4.36E-08 0.0 1.000
3-dehydrocarnitine 155 39.31 (29.72-67.40) 0.02 (0.01-0.03) 7.77E-63 3.18E-60 0.0 0.996
Alanine/tyrosine 290 47.64 (41.40-66.03) 0.76 (0.73-0.80) 5.22E-35 4.5E-33 0.0 1.000
Androstenediol 131 45.29 (30.30-64.22) 0.97 (0.95-0.99) 0.001 0.009 0.0 0.998
Decenoylcarnitine (C10.1) 174 53.23 (29.96-63.30) 0.91 (0.86-0.95) >0.001 0.003 0.0 1.000
Cholesteryl ester CE(16:1) [M+NH4]1+ 18 64.66 (37.74-67.40) 0.28 (0.20-0.41) 4.86E-11 1.12E-09 0.0 0.790
Cholesteryl ester CE(17:1) [M+NH4]1+ 18 55.83 (47.79-66.03) 0.28 (0.19-0.41) 1.9E-11 4.48E-10 0.0 0.657
Cholesteryl ester CE(18:2) [M+NH4]1+ 16 63.18 (37.58-67.40) 0.10 (0.05-0.19) 8.38E-11 1.88E-09 0.0 0.852
Cholesteryl ester (20:4) 42 38.10 (31.01-43.22) 0.93 (0.89-0.97) >0.001 0.005 40.8 0.004
Cis-4-decenoyl-carnitine 284 45.12 (29.91-67.40) 0.85 (0.82-0.89) 1.48E-14 5.06E-13 31.2 9.4E-07
Dehydroisoandrosterone sulfate/4-androsten-3β,17β-diol disulfate 89 37.80 (30.83-58.60) 0.36 (0.23-0.56) 6.61E-06 9.67E-05 0.0 0.979
Diglyceride DG(44:6) [M+H-H2O]1+ 20 55.26 (37.81-65.23) 0.41 (0.32-0.54) 1.63E-11 3.89E-10 0.0 0.512
Glucose/taurine 3 22.17 (20.36-25.08) 0.52 (0.33-0.80) 0.003 0.026 75.6 0.017
Glycolithocholate sulfate 65 53.70 (30.64-55.53) 0.67 (0.61-0.75) 7.63E-14 2.43E-12 19.7 0.089
Glycylphenylalanine 92 44.00 (29.73-67.40) 0.88 (0.85-0.91) 1.43E-13 4.27E-12 0.0 0.993
Glycerophospholipid/serum-triacylglycerol 216 45.81 (29.76-110.87) 0.82 (0.78-0.86) 1.74E-15 6.94E-14 0.0 1.000
Hexanoyl-carnitine 409 46.26 (29.97-67.40) 0.90 (0.87-0.94) 1.68E-06 2.64E-05 31.5 3.5E-09
Hydroxyisovaleroyl carnitine 79 36.74 (29.83-67.40) 0.05 (0.03-0.08) 1.34E-43 1.62E-41 0.0 1.000
Myo-inositol 40 49.54 (34.34-56.57) 0.04 (0.02-0.12) 1.01E-09 2.13E-08 0.0 1.000
PC plasmalogen (ether-linked) C32:2 50 33.37 (29.76-66.03) 0.82 (0.73-0.93) 0.003 0.020 0.0 1.000
Phosphatidylethanolamine PE(36:4) 33 34.87 (30.31-60.51) 0.92 (0.88-0.97) 0.001 0.012 31.7 0.044
Phenylalanylserine 57 33.80 (29.81-67.40) 0.89 (0.85-0.94) 1.4E-05 0.000 21.8 0.078
Sphingomyelin SM(34:2) [M+H]1+ 19 53.82 (38.23-65.23) 0.06 (0.03-0.14) 1.5E-11 3.62E-10 0.0 0.621
Sphingomyelin SM(36:2) [M+H]1+ 18 59.69 (37.87-64.22) 0.09 (0.05-0.17) 1.05E-12 2.93E-11 0.0 0.906
Sphingomyelin SM(41:1) [M+H]1+ 17 50.91 (38.57-62.34) 0.13 (0.07-0.23) 1.04E-11 2.56E-10 0.0 0.722
Triglyceride TG(51:3) [M+NH4]1+ 7 42.55 (39.88-47.94) 0.57 (0.41-0.79) 0.001 0.009 15.3 0.313
Triglyceride TG(53:3) [M+NH4]1+ 12 40.97 (39.32-51.85) 0.67 (0.52-0.87) 0.002 0.018 16.8 0.279
Urate/histidine 322 52.04 (30.11-67.40) 0.07 (0.05-0.09) 2.07E-65 9.67E-63 0.0 1.000
X-08402 64 34.02 (29.82-67.40) 0.11 (0.06-0.20) 5.67E-13 1.62E-11 0.0 0.927
X-08402/cholesterol 84 42.63 (29.72-67.40) 0.02 (0.01-0.04) 5.75E-34 4.71E-32 0.0 1.000
X-11247 38 46.83 (31.27-50.93) 0.38 (0.29-0.50) 2.38E-12 6.34E-11 0.0 1.000
X-17178 102 43.31 (31.02-63.87) 0.80 (0.77-0.83) 3.2E-33 2.44E-31 0.0 0.903
Cholesteryl ester (20:4) 447 33.75 (30.12-65.23) 0.84 (0.80-0.89) 6.48E-11 1.47E-09 0.0 1.000
2-aminooctanoate 117 49.35 (30.49-67.40) 0.82 (0.76-0.89) 5.69E-07 9.28E-06 0.0 1.000
3-hydroxyisovalerate 134 26.23 (19.55-35.74) 0.18 (0.11-0.29) 1.31E-12 3.57E-11 40.2 1.88E-06

OR, odds ratio; CI, confidence interval; FDR, false discovery rate; PC, phosphatidylcholine.

  • 1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229−263.ArticlePubMed
  • 2. Oh JH, Jun DW. The latest global burden of liver cancer: a past and present threat. Clin Mol Hepatol 2023;29:355−357.ArticlePubMedPMCPDF
  • 3. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:6. ArticlePubMedPDF
  • 4. Johnson CH, Ivanisevic J, Siuzdak G. Metabolomics: beyond biomarkers and towards mechanisms. Nat Rev Mol Cell Biol 2016;17:451−459.ArticlePubMedPMCPDF
  • 5. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab 2016;23:27−47.ArticlePubMedPMC
  • 6. Elia I, Haigis MC. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab 2021;3:21−32.ArticlePubMedPMCPDF
  • 7. Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, et al. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024;9:132. ArticlePubMedPMCPDF
  • 8. Kim J, DeBerardinis RJ. Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab 2019;30:434−446.ArticlePubMedPMC
  • 9. Sanchez JI, Fontillas AC, Kwan SY, Sanchez CI, Calderone TL, Lee JL, et al. Metabolomics biomarkers of hepatocellular carcinoma in a prospective cohort of patients with cirrhosis. JHEP Rep 2024;6:101119. ArticlePubMedPMC
  • 10. Anh NH, Long NP, Min YJ, Ki Y, Kim SJ, Jung CW, et al. Molecular and metabolic phenotyping of hepatocellular carcinoma for biomarker discovery: a meta-analysis. Metabolites 2023;13:1112. ArticlePubMedPMC
  • 11. Hammerton G, Munafò MR. Causal inference with observational data: the need for triangulation of evidence. Psychol Med 2021;51:563−578.ArticlePubMedPMC
  • 12. Richmond RC, Smith GD. Mendelian randomization: concepts and scope. Cold Spring Harb Perspect Med 2022;12:a040501. ArticlePubMedPMC
  • 13. Davies NM, Holmes MV, Smith GD. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ 2018;362:k601. ArticlePubMedPMC
  • 14. Bowden J, Del Greco MF, Minelli C, Smith GD, Sheehan N, Thompson J. A framework for the investigation of pleiotropy in two-sample summary data Mendelian randomization. Stat Med 2017;36:1783−1802.ArticlePubMedPMCPDF
  • 15. Bowden J, Smith GD, Haycock PC, Burgess S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol 2016;40:304−314.ArticlePubMedPMCPDF
  • 16. Bowden J, Smith GD, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol 2015;44:512−525.ArticlePubMedPMC
  • 17. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet 2018;50:693−698.ArticlePubMedPMCPDF
  • 18. R Foundation. The R Project for statistical computing [Internet]. Vienna: R Foundation; [cited 2025 Feb 14]. Available from: https://www.rproject.org/
  • 19. Pang Z, Lu Y, Zhou G, Hui F, Xu L, Viau C, et al. MetaboAnalyst 6.0: towards a unified platform for metabolomics data processing, analysis and interpretation. Nucleic Acids Res 2024;52:W398−W406.ArticlePubMedPMCPDF
  • 20. Tang X, Xue J, Zhang J, Zhou J. Causal effect of immunocytes, plasma metabolites, and hepatocellular carcinoma: a bidirectional two-sample Mendelian randomization study and mediation analysis in East Asian populations. Genes (Basel) 2024;15:1183. ArticlePubMedPMC
  • 21. Lu J, Feng Y, Guo K, Sun L, Ruan S, Zhang K. Association between human blood metabolome and the risk of gastrointestinal tumors. PLoS One 2024;19:e0304574.ArticlePubMedPMC
  • 22. Ning L, Gao Z, Chen D, Han J, Xie G, Sun J. Causality of blood metabolites on hepatocellular carcinoma and cholangiocarcinoma: a metabolome-wide mendelian randomization study. BMC Cancer 2025;25:389. ArticlePubMedPMCPDF
  • 23. Ma C, Tang L, Yao J, Tan G. Identifying proteins and amino acids associated with liver cancer risk: a study utilizing Mendelian randomization and bulk RNA sequencing analysis. J Pers Med 2024;14:262. ArticlePubMedPMC
  • 24. Zhang Y, Tang J, Jiang C, Yi H, Guang S, Yin G, et al. Metabolic reprogramming in cancer and senescence. MedComm (2020) 2025;6:e70055.ArticlePubMedPMC
  • 25. Xu X, Peng Q, Jiang X, Tan S, Yang Y, Yang W, et al. Metabolic reprogramming and epigenetic modifications in cancer: from the impacts and mechanisms to the treatment potential. Exp Mol Med 2023;55:1357−1370.ArticlePubMedPMCPDF
  • 26. Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, et al. Metabolic reprogramming in cancer: mechanisms and therapeutics. MedComm (2020) 2023;4:e218.ArticlePubMedPMC
  • 27. Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 2012;21:297−308.ArticlePubMedPMC
  • 28. Park S, Hall MN. Metabolic reprogramming in hepatocellular carcinoma: mechanisms and therapeutic implications. Exp Mol Med 2025;57:515−523.ArticlePubMedPMCPDF
  • 29. Barba I, Carrillo-Bosch L, Seoane J. Targeting the Warburg effect in cancer: where do we stand? Int J Mol Sci 2024;25:3142. ArticlePubMedPMC
  • 30. Vaupel P, Schmidberger H, Mayer A. The Warburg effect: essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol 2019;95:912−919.ArticlePubMed
  • 31. Shen J, Ye Y, Chang DW, Huang M, Heymach JV, Roth JA, et al. Circulating metabolite profiles to predict overall survival in advanced non-small cell lung cancer patients receiving first-line chemotherapy. Lung Cancer 2017;114:70−78.ArticlePubMedPMC
  • 32. Du Y, Xiao X, Liu F, Zhu W, Mo J, Liu Z. Causal effects of metabolites on malignant neoplasm of bone and articular cartilage: a mendelian randomization study. Front Genet 2025;16:1366743. ArticlePubMedPMC
  • 33. Xu Q, Deng Y, Ming J, Luo Z, Chen X, Chen T, et al. Methyl 6-O-cinnamoyl-α-d-glucopyranoside ameliorates acute liver injury by inhibiting oxidative stress through the activation of Nrf2 signaling pathway. Front Pharmacol 2022;13:873938. ArticlePubMedPMC
  • 34. Cotte AK, Cottet V, Aires V, Mouillot T, Rizk M, Vinault S, et al. Phospholipid profiles and hepatocellular carcinoma risk and prognosis in cirrhotic patients. Oncotarget 2019;10:2161−2172.ArticlePubMedPMC
  • 35. Lv Y, Hou X, Zhang Q, Li R, Xu L, Chen Y, et al. Untargeted metabolomics study of the in vitro anti-hepatoma effect of saikosaponin D in combination with NRP-1 knockdown. Molecules 2019;24:1423. ArticlePubMedPMC
  • 36. Day ZI, Mayfosh AJ, Giel MC, Hong Y, Williams SA, Santavanond JP, et al. Novel formulation of undecylenic acid induces tumor cell apoptosis. Int J Mol Sci 2022;23:14170. ArticlePubMedPMC
  • 37. Costello E, Goodrich JA, Patterson WB, Walker DI, Chen JC, Baumert BO, et al. Proteomic and metabolomic signatures of diet quality in young adults. Nutrients 2024;16:429. ArticlePubMedPMC
  • 38. Shu W, Liu L, Jiang J, Yao Q. Dietary patterns and hepatocellular carcinoma risk: a systematic review and meta-analysis of cohort and case-control studies. Nutr Metab (Lond) 2024;21:47. ArticlePubMedPMCPDF
  • 39. Hushmandi K, Einollahi B, Saadat SH, Lee EHC, Farani MR, Okina E, et al. Amino acid transporters within the solute carrier superfamily: underappreciated proteins and novel opportunities for cancer therapy. Mol Metab 2024;84:101952. ArticlePubMedPMC
  • 40. Bharadwaj R, Jaiswal S, de la Cruz EEV, Thakare RP. Targeting solute carrier transporters (SLCs) as a therapeutic target in different cancers. Diseases 2024;12:63. ArticlePubMedPMC
  • 41. Arefhosseini S, Roshanravan N, Tutunchi H, Rostami S, Khoshbaten M, Ebrahimi-Mameghani M. Myo-inositol supplementation improves cardiometabolic factors, anthropometric measures, and liver function in obese patients with non-alcoholic fatty liver disease. Front Nutr 2023;10:1092544. ArticlePubMedPMC
  • 42. Dajani AI, Popovic B. Essential phospholipids for nonalcoholic fatty liver disease associated with metabolic syndrome: a systematic review and network meta-analysis. World J Clin Cases 2020;8:5235−5249.ArticlePubMedPMC
  • 43. Pani A, Giossi R, Menichelli D, Fittipaldo VA, Agnelli F, Inglese E, et al. Inositol and non-alcoholic fatty liver disease: a systematic review on deficiencies and supplementation. Nutrients 2020;12:3379. ArticlePubMedPMC
  • 44. Kim DY. Changing etiology and epidemiology of hepatocellular carcinoma: Asia and worldwide. J Liver Cancer 2024;24:62−70.ArticlePubMedPMCPDF
  • 45. Baecker A, Liu X, La Vecchia C, Zhang ZF. Worldwide incidence of hepatocellular carcinoma cases attributable to major risk factors. Eur J Cancer Prev 2018;27:205−212.ArticlePubMedPMC
  • 46. Maucort-Boulch D, de Martel C, Franceschi S, Plummer M. Fraction and incidence of liver cancer attributable to hepatitis B and C viruses worldwide. Int J Cancer 2018;142:2471−2477.ArticlePubMedPDF
  • 47. Surendran P, Stewart ID, Au Yeung VPW, Pietzner M, Raffler J, Wörheide MA, et al. Rare and common genetic determinants of metabolic individuality and their effects on human health. Nat Med 2022;28:2321−2332.ArticlePubMedPMCPDF
  • 48. Ghouse J, Gellert-Kristensen H, O'Rourke CJ, Seidelin AS, Thorleifsson G, Sveinbjörnsson G, et al. Genome-wide meta-analysis identifies nine loci associated with higher risk of hepatocellular carcinoma development. JHEP Rep 2025;7:101485. ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • ePub LinkePub Link
      • XML DownloadDownload Citation
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        Association between metabolites and hepatocellular carcinoma: findings from a two-sample Mendelian randomization study
        J Liver Cancer. 2025;25(2):251-265.   Published online September 2, 2025
        Close
      • XML DownloadXML Download
      Related articles
      Association between metabolites and hepatocellular carcinoma: findings from a two-sample Mendelian randomization study
      Image Image Image Image
      Figure 1. Flowchart for identification of instrumental variables.
      Figure 2. Enriched pathways of metabolites positively associated with hepatocellular carcinoma. IEM, inborn errors of metabolism; SLC, solute carrier.
      Figure 3. Enriched pathways of metabolites negatively associated with hepatocellular carcinoma. PI, phosphatidylinositol; MECP2, methyl- CpG-binding protein 2; SLC, solute carrier.
      Graphical abstract
      Association between metabolites and hepatocellular carcinoma: findings from a two-sample Mendelian randomization study
      Metabolite Number of instruments Median F-statistics (range) Inverse-variance weighted
      Heterogeneity
      OR (95% CI) Raw P-value FDR P-value I2 (%) P-value
      Valylarginine 65 57.89 (42.74-67.40) 1.27 (1.15-1.40) 1.81E-06 2.84E-05 0.0 >0.999
      Free carnitine (C0) 80 56.90 (41.30-67.40) 1.15 (1.11-1.19) 6.48E-15 2.33E-13 0.0 >0.999
      Propionyl-carnitine 7 30.82 (30.12-32.51) 1.45 (1.23-1.70) 9.49E-06 0.0001 37.3 0.144
      X-23314 1,100 35.00 (21.57-67.40) 1.67 (1.55-1.79) 4.63E-44 6.32E-42 74.0 <0.001
      Methyl glucopyranoside (α+β) 69 39.80 (30.20-67.40) 30.24 (19.65-46.54) 3.76E-54 8.8E-52 0.0 >0.999
      Tyrosine 67 58.00 (29.75-67.40) 4.29 (1.94-9.46) 0.0003 0.003 37.5 0.001
      Large VLDL particles 63 47.26 (33.21-62.34) 1.15 (1.05-1.26) 0.003 0.024 0.0 0.972
      Small VLDL-cholesterol 25 58.77 (39.14-67.40) 2.94 (2.02-4.30) 2.26E-08 4.27E-07 19.4 0.193
      Large VLDL-triglycerides 20 61.76 (37.74-64.22) 3.84 (2.55-5.78) 1.3E-10 2.9E-09 0.0 0.650
      Double bond protons of mobile lipids (MobCH) 104 34.93 (29.82-60.98) 3.98 (2.70-5.86) 2.69E-12 6.99E-11 94.1 <0.001
      Medium VLDL particles 273 47.41 (34.11-126.55) 1.16 (1.11-1.21) 1.82E-10 3.99E-09 5.4 0.250
      Serum total triglycerides 481 35.34 (29.73-77.93) 1.48 (1.35-1.61) 4.61E-18 2.1E-16 73.3 <0.001
      Small VLDL particles 43 42.76 (32.69-60.98) 1.35 (1.18-1.56) 2.39E-05 <0.001 36.3 0.011
      Large VLDL total lipids 20 52.33 (34.62-62.67) 1.56 (1.26-1.93) 4.52E-05 0.001 49.4 0.007
      Small VLDL-free cholesterol 143 42.22 (10.65-67.40) 1.47 (1.37-1.57) 1.17E-29 7.8E-28 14.4 0.084
      Extra-small VLDL phospholipids 1,570 40.10 (10.06-67.40) 1.15 (1.12-1.18) 1.29E-29 8.47E-28 0.0 0.998
      Medium VLDL total lipids 31 28.74 (19.57-57.77) 12.15 (6.58-22.44) 1.49E-15 6.03E-14 0.0 0.712
      Large VLDL-free cholesterol 1,181 40.04 (16.06-137.87) 1.27 (1.23-1.32) 1.04E-38 1.06E-36 19.8 <0.001
      FAw67 1,165 40.90 (11.04-118.53) 1.25 (1.21-1.30) 1.18E-31 8.05E-30 21.1 <0.001
      Large VLDL-cholesterol 1,143 40.62 (13.55-108.54) 1.37 (1.30-1.44) 1.17E-33 9.35E-32 58.6 <0.001
      Large VLDL-cholesteryl esters 1,085 40.98 (13.24-107.47) 1.43 (1.35-1.52) 8.56E-33 6.37E-31 67.2 <0.001
      Very-large VLDL total lipids 1,142 39.13 (11.45-107.18) 1.34 (1.28-1.40) 3.48E-36 3.36E-34 41.0 <0.001
      Medium VLDL-cholesterol 941 38.10 (11.60-121.37) 1.43 (1.32-1.55) 4.59E-19 2.15E-17 79.5 <0.001
      IDL-triglycerides 1,166 39.37 (29.72-111.42) 1.32 (1.27-1.37) 1.84E-49 3.17E-47 13.1 <0.001
      Extra-small VLDL particles 1,073 40.82 (29.72-108.62) 1.34 (1.29-1.38) 2.2E-55 5.53E-53 9.3 <0.001
      Extra-small VLDL total lipids 1,214 38.80 (21.07-118.66) 1.32 (1.27-1.37) 6.2E-50 1.13E-47 13.5 <0.001
      Very-large VLDL-triglycerides 1,076 41.22 (24.59-110.08) 1.32 (1.28-1.37) 8.26E-53 1.69E-50 9.2 0.011
      Linoleic acid 1,059 41.48 (22.73-108.46) 1.35 (1.30-1.40) 7.66E-54 1.67E-51 17.4 <0.001
      Medium VLDL-triglycerides 1,092 40.29 (19.99-105.04) 1.32 (1.27-1.38) 1.16E-41 1.31E-39 22.3 <0.001
      Medium VLDL phospholipids 869 45.19 (11.43-98.92) 1.3 (1.19-1.42) 5.24E-09 1.05E-07 82.2 <0.001
      Apolipoprotein B 806 40.14 (15.58-108.71) 1.43 (1.31-1.57) 2.92E-15 1.12E-13 81.4 <0.001
      Extremely-large VLDL particles 919 48.03 (21.68-90.05) 1.28 (1.17-1.40) 4.75E-08 8.69E-07 82.7 <0.001
      Very-large VLDL phospholipids 586 41.69 (12.37-87.83) 1.35 (1.20-1.51) 4.54E-07 7.59E-06 84.7 <0.001
      Small VLDL phospholipids 842 45.81 (18.71-88.74) 1.36 (1.25-1.48) 2.65E-12 6.94E-11 81.1 <0.001
      Extra-small VLDL-triglycerides 782 41.10 (22.89-74.27) 1.35 (1.21-1.50) 5.09E-08 9.25E-07 84.8 <0.001
      Medium VLDL-free cholesterol 728 40.11 (17.74-99.09) 1.27 (1.15-1.40) 3.02E-06 4.6E-05 81.8 <0.001
      Large VLDL phospholipids 1,207 41.11 (29.73-219.46) 1.27 (1.23-1.32) 1.92E-38 1.9E-36 16.8 <0.001
      Very-large VLDL particles 741 42.30 (24.57-132.06) 1.27 (1.21-1.32) 4.08E-27 2.52E-25 29.8 <0.001
      Extremely-large VLDL-triglycerides 387 41.21 (19.57-77.15) 1.21 (1.14-1.28) 6.21E-12 1.57E-10 0.0 >0.999
      Extremely-large VLDL total lipids 1,091 38.00 (29.72-108.96) 1.31 (1.26-1.36) 8.28E-44 1.08E-41 11.4 0.002
      Very-large HDL-triglycerides 979 39.35 (29.73-109.47) 1.32 (1.28-1.38) 2.83E-48 4.64E-46 10.4 0.007
      Medium VLDL-cholesteryl esters 980 39.57 (29.72-106.04) 1.31 (1.26-1.36) 1.82E-44 2.6E-42 11.2 0.004
      Small VLDL-triglycerides 972 39.59 (29.75-103.96) 1.32 (1.27-1.37) 4.5E-46 6.69E-44 12.4 0.001
      Small VLDL total lipids 943 36.68 (29.74-91.84) 1.37 (1.32-1.43) 1.4E-50 2.7E-48 11.8 0.003
      C-glycosyltryptophan/hydroxyisovalerylcarnitine 1,205 44.00 (13.96-108.59) 1.44 (1.32-1.56) 2.87E-18 1.32E-16 84.9 <0.001
      Valine/isovalerylcarnitine 1,058 43.27 (17.25-125.91) 1.73 (1.59-1.88) 7.76E-39 8.2E-37 84.0 <0.001
      Large LDL phospholipids 1,009 50.52 (13.56-101.08) 1.29 (1.19-1.40) 6.37E-10 1.35E-08 83.5 <0.001
      Small LDL total lipids 831 40.87 (16.22-97.78) 1.39 (1.27-1.52) 9.66E-13 2.73E-11 83.6 <0.001
      Large LDL-cholesterol 849 41.31 (15.17-115.54) 1.36 (1.25-1.49) 7.29E-12 1.82E-10 83.5 <0.001
      IDL total lipids 998 49.19 (15.36-99.44) 1.25 (1.15-1.36) 7.44E-08 1.33E-06 83.1 <0.001
      Medium LDL-cholesterol 726 40.11 (13.12-99.04) 1.26 (1.15-1.39) 1.92E-06 2.96E-05 83.3 <0.001
      LDL-cholesterol 147 44.23 (29.76-67.40) 1.05 (1.03-1.07) 9.75E-06 0.0001 0.0 >0.99
      IDL phospholipids 198 34.26 (29.76-67.40) 2.02 (1.50-2.72) 3.74E-06 5.64E-05 94.0 <0.001
      Large LDL-cholesteryl esters 114 41.58 (29.74-111.54) 4.29 (2.96-6.22) 1.26E-14 4.37E-13 95.1 <0.001
      IDL-free cholesterol 402 41.14 (12.25-107.19) 2.15 (1.85-2.49) 9.3E-24 5.25E-22 91.0 <0.001
      Medium LDL particles 54 39.20 (10.72-60.98) 1.18 (1.08-1.30) 0.001 0.005 0.0 0.991
      Large LDL particles 57 41.05 (29.92-49.76) 1.33 (1.21-1.48) 2.17E-08 4.13E-07 0.0 >0.999
      IDL particles 35 60.18 (39.03-63.56) 1.59 (1.28-1.98) 2.41E-05 0.0003 0.0 0.476
      Medium LDL-cholesteryl esters 179 38.34 (29.73-66.03) 1.11 (1.03-1.20) 0.006 0.041 0.0 0.999
      Serum total cholesterol 965 41.03 (17.31-112.42) 1.32 (1.27-1.38) 1.12E-43 1.41E-41 13.3 0.001
      Medium LDL total lipids 819 40.89 (10.49-102.57) 1.41 (1.33-1.49) 1.08E-34 9.09E-33 50.8 <0.001
      Small LDL-cholesterol 788 40.27 (10.30-96.91) 1.71 (1.57-1.86) 2.26E-35 2.06E-33 78.4 <0.001
      IDL-cholesterol 943 39.89 (21.32-108.34) 1.61 (1.49-1.74) 2.16E-33 1.68E-31 75.6 <0.001
      Small LDL particles 1,033 42.13 (27.79-114.84) 1.58 (1.46-1.70) 1.13E-31 7.91E-30 78.8 <0.001
      Esterified cholesterol 1,115 40.94 (13.42-111.12) 1.47 (1.36-1.59) 4.38E-23 2.39E-21 81.8 <0.001
      Large LDL total lipids 1,092 42.67 (10.93-109.13) 1.40 (1.30-1.51) 4.01E-19 1.9E-17 80.7 <0.001
      Medium LDL phospholipids 1,178 44.53 (11.83-121.88) 1.37 (1.27-1.47) 3.95E-17 1.77E-15 80.3 <0.001
      Large LDL-free cholesterol 927 44.84 (12.34-98.35) 1.35 (1.24-1.48) 1.76E-11 4.18E-10 84.0 <0.001
      Total fatty acids 1,039 45.18 (13.94-104.31) 1.42 (1.30-1.55) 1.29E-14 4.43E-13 86.8 <0.001
      Glycine 966 39.39 (29.72-97.33) 1.63 (1.50-1.76) 1.11E-32 8.07E-31 77.7 <0.001
      3-ureidopropionate 6 55.11 (38.93-67.40) 1.99 (1.33-2.97) 0.001 0.008 11.0 0.345
      Lysine 22 60.29 (40.51-67.40) 3.24 (2.05-5.14) 5.37E-07 8.84E-06 26.2 0.128
      Monounsaturated fatty acids 18 53.15 (41.84-58.72) 5.27 (3.46-8.02) 8.39E-15 2.95E-13 0.0 0.999
      1-ribosyl-imidazoleacetate 17 66.03 (45.08-67.40) 5.21 (3.39-8.01) 5.34E-14 1.72E-12 0.0 0.999
      otPUFA 19 61.40 (37.60-67.40) 4.17 (2.84-6.13) 3.11E-13 9.02E-12 0.0 0.834
      X-21343 18 59.92 (52.26-67.40) 4.53 (3.08-6.66) 1.68E-14 5.67E-13 0.0 0.995
      Phosphatidylcholine 37:4/Phosphatidylethanolamine 40:3 [M+H]+ 268 36.25 (29.78-109.10) 2.00 (1.64-2.45) 1.35E-11 3.26E-10 91.5 0.000
      Phosphatidylcholine (acyl-alkyl) C38:3 (PC ae C38:3) 301 50.26 (29.83-67.40) 1.62 (1.50-1.74) 1.07E-35 9.98E-34 0.0 0.994
      Phosphatidylcholine (diacyl) C32:2 (PC aa C32:2) 126 47.19 (29.72-67.40) 4.62 (2.56-8.35) 3.7E-07 6.25E-06 56.6 <0.001
      Omega-3 fatty acids 61 32.22 (29.73-66.03) 1.18 (1.12-1.25) 2.75E-09 5.63E-08 18.2 0.115
      Docosahexaenoic acid 57 33.03 (29.74-46.17) 1.10 (1.03-1.17) 0.003 0.025 0.0 0.959
      X-14473 121 48.64 (29.81-67.40) 1.49 (1.18-1.89) 0.001 0.009 21.6 0.022
      X-11538 167 56.20 (29.88-66.03) 1.07 (1.05-1.10) 1.09E-07 1.91E-06 0.0 0.921
      X-21849 47 43.56 (31.35-47.48) 38.75 (26.11-57.52) 1.2E-73 6.57E-71 0.0 >0.999
      X-02249 48 32.58 (29.82-67.40) 1.14 (1.08-1.20) 6.3E-06 9.34E-05 0.0 0.986
      Glutamine 245 30.29 (29.78-43.89) 1.18 (1.15-1.21) 3.74E-41 4.09E-39 0.0 >0.999
      C-glycosyltryptophan/succinylcarnitine 98 32.49 (29.85-67.40) 1.08 (1.05-1.12) 3.64E-07 6.18E-06 0.0 >0.999
      Triacylglycerol 51:3 [M+NH4]+ 1,339 40.59 (10.31-122.11) 1.43 (1.39-1.48) 1.8E-99 2.01E-96 17.2 <0.001
      Triacylglycerol 53:4 [M+NH4]+ 582 35.16 (14.83-65.23) 1.63 (1.43-1.85) 9.4E-14 2.87E-12 84.3 <0.001
      Diacylglycerol 36:3 [M+NH4]+ 528 37.27 (14.46-65.23) 1.43 (1.28-1.61) 1.17E-09 2.43E-08 79.7 <0.001
      Triacylglycerol 50:4 [M+NH4]+ 719 43.86 (26.58-61.81) 1.39 (1.25-1.55) 1.12E-09 2.33E-08 82.6 <0.001
      Triacylglycerol 54:5 [M+NH4]+ 791 43.45 (26.61-65.23) 1.21 (1.10-1.33) <0.001 0.002 80.2 <0.001
      Triacylglycerol 50:3 [M+NH4]+ 979 44.61 (13.35-128.45) 1.43 (1.33-1.54) 2.42E-21 1.24E-19 79.4 <0.001
      Diacylglycerol 34:3 [M+H-H2O]+ 981 44.44 (26.80-134.77) 1.40 (1.30-1.51) 3.04E-19 1.46E-17 79.8 <0.001
      FAw9S 1,158 36.25 (18.80-111.57) 1.43 (1.34-1.52) 3.54E-28 2.27E-26 73.5 <0.001
      X-12822 1,173 41.50 (16.12-111.13) 1.41 (1.32-1.50) 5.56E-26 3.37E-24 78.1 <0.001
      Glycolate 160 36.16 (29.72-67.40) 1.54 (1.26-1.88) 2.65E-05 0.0003 75.9 <0.001
      7-methylguanine 354 31.90 (29.72-66.03) 1.44 (1.21-1.71) 4.46E-05 0.0006 85.7 <0.001
      Sphingomyelin 32:1 [M+H]+ 660 37.26 (29.72-67.40) 1.17 (1.05-1.29) 0.004 0.030 76.5 <0.001
      Metabolite Number of instruments Median F-statistics (range) Inverse-variance weighted
      Heterogeneity
      OR (95% CI) Raw P-value FDR P-value I2 (%) P-value
      1-stearoylglycero-phosphoethanolamine 24 46.34 (29.93-67.40) 0.63 (0.47-0.83) 0.001 0.009 25.6 0.125
      10-undecenoate (11:1n1) 220 44.42 (29.72-67.40) 0.93 (0.91-0.95) 2.12E-09 4.36E-08 0.0 1.000
      3-dehydrocarnitine 155 39.31 (29.72-67.40) 0.02 (0.01-0.03) 7.77E-63 3.18E-60 0.0 0.996
      Alanine/tyrosine 290 47.64 (41.40-66.03) 0.76 (0.73-0.80) 5.22E-35 4.5E-33 0.0 1.000
      Androstenediol 131 45.29 (30.30-64.22) 0.97 (0.95-0.99) 0.001 0.009 0.0 0.998
      Decenoylcarnitine (C10.1) 174 53.23 (29.96-63.30) 0.91 (0.86-0.95) >0.001 0.003 0.0 1.000
      Cholesteryl ester CE(16:1) [M+NH4]1+ 18 64.66 (37.74-67.40) 0.28 (0.20-0.41) 4.86E-11 1.12E-09 0.0 0.790
      Cholesteryl ester CE(17:1) [M+NH4]1+ 18 55.83 (47.79-66.03) 0.28 (0.19-0.41) 1.9E-11 4.48E-10 0.0 0.657
      Cholesteryl ester CE(18:2) [M+NH4]1+ 16 63.18 (37.58-67.40) 0.10 (0.05-0.19) 8.38E-11 1.88E-09 0.0 0.852
      Cholesteryl ester (20:4) 42 38.10 (31.01-43.22) 0.93 (0.89-0.97) >0.001 0.005 40.8 0.004
      Cis-4-decenoyl-carnitine 284 45.12 (29.91-67.40) 0.85 (0.82-0.89) 1.48E-14 5.06E-13 31.2 9.4E-07
      Dehydroisoandrosterone sulfate/4-androsten-3β,17β-diol disulfate 89 37.80 (30.83-58.60) 0.36 (0.23-0.56) 6.61E-06 9.67E-05 0.0 0.979
      Diglyceride DG(44:6) [M+H-H2O]1+ 20 55.26 (37.81-65.23) 0.41 (0.32-0.54) 1.63E-11 3.89E-10 0.0 0.512
      Glucose/taurine 3 22.17 (20.36-25.08) 0.52 (0.33-0.80) 0.003 0.026 75.6 0.017
      Glycolithocholate sulfate 65 53.70 (30.64-55.53) 0.67 (0.61-0.75) 7.63E-14 2.43E-12 19.7 0.089
      Glycylphenylalanine 92 44.00 (29.73-67.40) 0.88 (0.85-0.91) 1.43E-13 4.27E-12 0.0 0.993
      Glycerophospholipid/serum-triacylglycerol 216 45.81 (29.76-110.87) 0.82 (0.78-0.86) 1.74E-15 6.94E-14 0.0 1.000
      Hexanoyl-carnitine 409 46.26 (29.97-67.40) 0.90 (0.87-0.94) 1.68E-06 2.64E-05 31.5 3.5E-09
      Hydroxyisovaleroyl carnitine 79 36.74 (29.83-67.40) 0.05 (0.03-0.08) 1.34E-43 1.62E-41 0.0 1.000
      Myo-inositol 40 49.54 (34.34-56.57) 0.04 (0.02-0.12) 1.01E-09 2.13E-08 0.0 1.000
      PC plasmalogen (ether-linked) C32:2 50 33.37 (29.76-66.03) 0.82 (0.73-0.93) 0.003 0.020 0.0 1.000
      Phosphatidylethanolamine PE(36:4) 33 34.87 (30.31-60.51) 0.92 (0.88-0.97) 0.001 0.012 31.7 0.044
      Phenylalanylserine 57 33.80 (29.81-67.40) 0.89 (0.85-0.94) 1.4E-05 0.000 21.8 0.078
      Sphingomyelin SM(34:2) [M+H]1+ 19 53.82 (38.23-65.23) 0.06 (0.03-0.14) 1.5E-11 3.62E-10 0.0 0.621
      Sphingomyelin SM(36:2) [M+H]1+ 18 59.69 (37.87-64.22) 0.09 (0.05-0.17) 1.05E-12 2.93E-11 0.0 0.906
      Sphingomyelin SM(41:1) [M+H]1+ 17 50.91 (38.57-62.34) 0.13 (0.07-0.23) 1.04E-11 2.56E-10 0.0 0.722
      Triglyceride TG(51:3) [M+NH4]1+ 7 42.55 (39.88-47.94) 0.57 (0.41-0.79) 0.001 0.009 15.3 0.313
      Triglyceride TG(53:3) [M+NH4]1+ 12 40.97 (39.32-51.85) 0.67 (0.52-0.87) 0.002 0.018 16.8 0.279
      Urate/histidine 322 52.04 (30.11-67.40) 0.07 (0.05-0.09) 2.07E-65 9.67E-63 0.0 1.000
      X-08402 64 34.02 (29.82-67.40) 0.11 (0.06-0.20) 5.67E-13 1.62E-11 0.0 0.927
      X-08402/cholesterol 84 42.63 (29.72-67.40) 0.02 (0.01-0.04) 5.75E-34 4.71E-32 0.0 1.000
      X-11247 38 46.83 (31.27-50.93) 0.38 (0.29-0.50) 2.38E-12 6.34E-11 0.0 1.000
      X-17178 102 43.31 (31.02-63.87) 0.80 (0.77-0.83) 3.2E-33 2.44E-31 0.0 0.903
      Cholesteryl ester (20:4) 447 33.75 (30.12-65.23) 0.84 (0.80-0.89) 6.48E-11 1.47E-09 0.0 1.000
      2-aminooctanoate 117 49.35 (30.49-67.40) 0.82 (0.76-0.89) 5.69E-07 9.28E-06 0.0 1.000
      3-hydroxyisovalerate 134 26.23 (19.55-35.74) 0.18 (0.11-0.29) 1.31E-12 3.57E-11 40.2 1.88E-06
      Table 1. Genetically metabolites positively associated with hepatocellular carcinoma risk

      OR, odds ratio; CI, confidence interval; FDR, false discovery rate; VLDL, very low density lipoprotein; IDL, intermediate-density lipoprotein; HDL, high-density lipoprotein; LDL, low-density lipoprotein.

      Table 2. Genetically metabolites negatively associated with hepatocellular carcinoma risk

      OR, odds ratio; CI, confidence interval; FDR, false discovery rate; PC, phosphatidylcholine.


      JLC : Journal of Liver Cancer
      TOP