Skip to main content
Erschienen in: BMC Medicine 1/2015

Open Access 01.12.2015 | Research article

Metabolomic profiles of hepatocellular carcinoma in a European prospective cohort

verfasst von: Anne Fages, Talita Duarte-Salles, Magdalena Stepien, Pietro Ferrari, Veronika Fedirko, Clément Pontoizeau, Antonia Trichopoulou, Krasimira Aleksandrova, Anne Tjønneland, Anja Olsen, Françoise Clavel-Chapelon, Marie-Christine Boutron-Ruault, Gianluca Severi, Rudolf Kaaks, Tilman Kuhn, Anna Floegel, Heiner Boeing, Pagona Lagiou, Christina Bamia, Dimitrios Trichopoulos, Domenico Palli, Valeria Pala, Salvatore Panico, Rosario Tumino, Paolo Vineis, H. Bas Bueno-de-Mesquita, Petra H. Peeters, Elisabete Weiderpass, Antonio Agudo, Esther Molina-Montes, José María Huerta, Eva Ardanaz, Miren Dorronsoro, Klas Sjöberg, Bodil Ohlsson, Kay-Tee Khaw, Nick Wareham, Ruth C. Travis, Julie A. Schmidt, Amanda Cross, Marc Gunter, Elio Riboli, Augustin Scalbert, Isabelle Romieu, Benedicte Elena-Herrmann, Mazda Jenab

Erschienen in: BMC Medicine | Ausgabe 1/2015

Abstract

Background

Hepatocellular carcinoma (HCC), the most prevalent form of liver cancer, is difficult to diagnose and has limited treatment options with a low survival rate. Aside from a few key risk factors, such as hepatitis, high alcohol consumption, smoking, obesity, and diabetes, there is incomplete etiologic understanding of the disease and little progress in identification of early risk biomarkers.

Methods

To address these aspects, an untargeted nuclear magnetic resonance metabolomic approach was applied to pre-diagnostic serum samples obtained from first incident, primary HCC cases (n = 114) and matched controls (n = 222) identified from amongst the participants of a large European prospective cohort.

Results

A metabolic pattern associated with HCC risk comprised of perturbations in fatty acid oxidation and amino acid, lipid, and carbohydrate metabolism was observed. Sixteen metabolites of either endogenous or exogenous origin were found to be significantly associated with HCC risk. The influence of hepatitis infection and potential liver damage was assessed, and further analyses were made to distinguish patterns of early or later diagnosis.

Conclusion

Our results show clear metabolic alterations from early stages of HCC development with application for better etiologic understanding, prevention, and early detection of this increasingly common cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12916-015-0462-9) contains supplementary material, which is available to authorized users.
Dimitrios Trichopoulos deceased.
Benedicte Elena-Herrmann and Mazda Jenab contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

The authors’ responsibilities were as follows: ER is the overall PI of the EPIC study which is jointly coordinated from ICL (ER) and IARC (IR); MJ and BE-H conceptualized, designed, obtained funding for, and implemented/managed the present research; AF, CP, and BE-H performed the laboratory analyses; AF and PF conducted the statistical analyses; AF, MJ, VF, TDS, MS, and BE-H contributed to the writing of the manuscript and data interpretation. Contributing authors from each collaborating centre provided the original data and biological samples, information on the respective populations, advice on study design/analysis, and interpretation of the results. All authors contributed comments on the draft manuscript and provided an approval of the final version of the manuscript for publication.
Abkürzungen
AAA
Aromatic amino acids
AFP
α-fetoprotein
ALT
Alanine aminotransferase
AP
Alkaline phosphatase
AST
Aspartate aminotransferase
AUC
Area under the curve
BCAA
Branched-chain amino-acids
CI
Confidence intervals
CLR
Conditional logistic regression
CMIAs
Chemiluminescent microparticle immunoassays
CPMG
Carr-Purcell-Meiboom-Gill
CV
Cross-validated
EPIC
European Prospective Investigation into Cancer and Nutrition
GGT
Gamma-glutamyltransferase
HBV
Chronic hepatitis B
HCC
Hepatocellular carcinoma
HCV
Chronic hepatitis C
HDL
High-density lipoproteins
LDL
Low-density lipoproteins
NMR
Nuclear magnetic resonance
NOESY
Nuclear Overhauser effect spectroscopy
O-PLS
Orthogonal partial least-square
OR
Odds ratios
ROC
Receiver operating characteristics
VLDL
Very-low-density lipoproteins

Background

Liver cancer is the sixth most commonly diagnosed cancer and the second leading cause of cancer death worldwide [1]. Hepatocellular carcinoma (HCC), the most frequent type of liver cancer, is primarily associated with chronic hepatitis B (HBV) and C (HCV) infections and aflatoxin exposure [2], while other major risk factors include obesity, type 2 diabetes, tobacco smoking, and heavy alcohol drinking [35]. HCC is highly malignant, usually diagnosed at late stages, and often has a poor prognosis with limited treatment options [6]. The late diagnosis and consequent poor survival associated with the disease are often attributed to its lack of pathognomonic symptoms and limitations of diagnostic modalities. Improving both the understanding of HCC etiology and the early detection of the disease is an important first step towards the design of effective prevention strategies aimed at early diagnosis and reduction of HCC incidence. A valuable tool toward these goals is the analysis of bio-samples from prospective cohort studies, where healthy participants are enrolled and followed over time for the appearance of various diseases. Since HCC development implies alterations in the metabolic functions of the liver and, in a majority of cases, progresses from pre-cancerous lesions through to cirrhosis and cancer, it is conceivable that metabolic changes may be detected from the very early stages of the disease, long prior to clinical diagnosis. Thus, metabolomics may serve as a valuable tool for the identification of biomarkers for early detection of HCC.
Metabolomics is a powerful high-throughput approach that relies on state of the art analytical methods, such as nuclear magnetic resonance (NMR), to identify metabolic signatures or biomarkers associated with homeostasis perturbations [7]. Metabolomic strategies play an increasingly important role in clinical and observational studies, in the hope that they will offer new perspectives not only in understanding the processes of disease development, but also for identification of diagnostic/prognostic markers and targeted healthcare [8]. Indeed, several recent studies have leveraged metabolite profiling to provide new insights into pathological processes pertaining to cancer, heart disease, or diabetes mellitus [915]. Although a number of metabolomic-based approaches have been applied to HCC, they have either been largely based on traditional case–control designs, high risk patient groups (e.g. hepatitis infection, cirrhosis, or other chronic liver diseases), non-Western populations where traditional HCC risk factors predominate, or on tumor tissues [1631]. However, there is currently very little information derived from prospective settings where biological samples have been collected prior to disease diagnosis [3234].
In this study, we investigated whether metabolic differences could be detected between HCC cases and matched controls derived from a prospective cohort study using serum samples collected prior to diagnosis. A NMR-based metabolomic approach was applied to a case–control study nested within a large, multi-center prospective cohort.

Methods

Study design

The present study is based on a case–control study nested within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort, a multicenter prospective study designed to investigate the association between diet, lifestyle, and environmental factors and the incidence of various types of cancer and other chronic diseases. The rationale, detailed study design, and methods have been previously detailed [35]. Briefly, diet and lifestyle data were collected at recruitment from approximately 520,000 men and women aged 35–85 years enrolled between 1992 and 2000 in 23 centers from 10 Western European countries (Denmark, France, Germany, Greece, Italy, Norway, Spain, Sweden, the Netherlands, and the United Kingdom) [35]. The study subjects were recruited from the general population, except for France (women who were members of a health insurance scheme for state school employees), Naples and Norway (women only), Utrecht and Florence (women attending breast cancer screening), and subsamples of the Oxford “Health Conscious” sub-cohort (vegetarians) and the Italian and Spanish cohorts (mainly members of blood donor associations).

Ethics

The EPIC cohort in general, and this study in particular, have received approval from the Ethics Committee of the International Agency for Research on Cancer as well as the ethics review boards of individual EPIC centers. EPIC participants provided written consent for the use of their blood samples and all data.

Blood sample collection

Blood samples were collected using standardized methods at recruitment from most participants and are stored at IARC (Lyon, France) in liquid nitrogen at –196 °C for all countries except Denmark (−150 °C, nitrogen vapor) and Sweden (−80 °C, freezers) where samples are stored locally [35].

Cancer and vital status assessment

Vital status during follow-up (98.5 % complete) was assessed by record linkage with regional and/or national mortality registries in all countries except Germany and Greece, where follow-up was actively reported by study subjects or their next-of-kin. Cancer incidence was determined through record linkage with population-based regional cancer registries (Denmark, Italy, the Netherlands, Norway, Spain, Sweden, and the United Kingdom) or via a combination of methods, including the use of health insurance records, contacts with cancer and pathology registries, and active follow-up through study subjects and their next-of-kin (France, Germany, Greece). For the present study, the dates of follow-up for cancer incidence and vital status are complete up to end of 2006.

The HCC nested case–control study

Ascertainment of cases

HCC cases were defined as tumor in the liver (C22.0) according to the 10th Revision of the International Statistical Classification of Diseases, Injury and Causes of Death. For each HCC case identified, the histology, methods used to diagnose the cancer, and α-fetoprotein (AFP) levels were reviewed to exclude metastatic cases or other types of primary liver cancers.

The nested case–control study

The design of the nested case–control study has been previously described in detail [36]. Briefly, 125 HCC cases with available blood samples at baseline were identified between participants’ recruitment and 2006. For each case, two controls were selected by incidence density sampling from all cohort members alive and free of cancer (except non-melanoma skin cancer), and matched by age at blood collection (±1 year), sex, study center, date (±2 months) and time of the day at blood collection (±3 h), and fasting status at blood collection (<3/3–6/>6 h). Women were additionally matched by menopausal status (pre-/peri-/postmenopausal) and hormone replacement therapy use at time of blood collection (yes/no). Participants with insufficient remaining blood sample for NMR analyses were excluded (Ncases = 11). For six cases, only one eligible control was available for each case. Therefore, the final sample size for the present analysis included 114 HCC cases and 222 matched controls.

Serum sample analysis

Laboratory assays: HBV/HCV infection, biomarkers of liver function and AFP

HBV and HCV seropositivity were detected in serum samples using the ARCHITECT HBsAg and anti-HCV chemiluminescent microparticle immunoassays (CMIAs; Abbott Diagnostics, France): HBsAg-positive when ≥0.05 IU/mL and HCV-positive when the ratio of sample relative light units to cutoff relative light units was ≥1 in two measurements [36]. Biochemical markers of hepatic injury, including albumin, total bilirubin, alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyltransferase (GGT), and liver-specific alkaline phosphatase (AP) were measured on the ARCHITECT c Systems™ (Abbott Diagnostics) using standard protocols. The normal ranges were: albumin, 35–50 g/L; total bilirubin, 3.4–20.5 mmol/L; ALT, <55 U/L; AST, 5–34 U/L; GGT, 12–64 U/L (men) and 9–36 U/L (women); and AP, 40–150 U/L. A liver function score was calculated from concentrations of albumin, total bilirubin, ALT, AST, GGT, and AP, each contributing 1 point when outside of the normal range [37]. The liver score was categorized as no liver damage (liver score 0), probable liver damage (liver score 1–2), and likely liver damage (liver score ≥3). Additionally, the concentration of serum AFP, which is currently a pre-diagnostic biomarker for HCC, was measured in blood using the ARCHITECT AFP kit. The laboratory analyses were performed at the Centre de Biologie République, Lyon, France [36].

NMR metabolomic data acquisition

Serum samples (200 μL) were processed according to standard procedures for NMR metabolomic measurement [38]. One-dimensional 1H Carr-Purcell-Meiboom-Gill (CPMG) and Nuclear Overhauser effect spectroscopy (NOESY) NMR spectra were recorded for each serum sample on a Bruker Avance III spectrometer operating at 800.15 MHz 1H NMR frequency. Additional two-dimensional NMR spectra were recorded on a set of representative samples (one control and one case) to achieve assignment of the NMR signals observed in the 1H one-dimensional fingerprints to metabolites. The measured chemical shifts were compared to reference shifts of pure compounds using the HMDB [39], MMCB [40], and ChenomX NMR Suite (Chenomx Inc., Edmonton, Canada) databases. Figure 1 shows the mean CPMG spectrum with metabolite assignments. The detailed list of the 44 annotated metabolites is provided in Additional file 1: Table S1. NMR signals arising from lipids enabled the quantification of unsaturated lipids in the serum (signal at 5.28 ppm, resonance of -CH = CH- from unsaturated lipids) as well as terminal lipids methyls corresponding to several classes of lipoproteins: very-low-density lipoproteins (VLDL; δ 0.86 ppm), low-density lipoproteins (LDL; δ 0.84 ppm), and high-density lipoproteins (HDL; δ 0.82 ppm). After processing and calibration, each 1D NMR spectrum was reduced into bins of 0.001 ppm width over a chemical shift range of 0.5–9 ppm using the AMIX software (Bruker GmbH, Rheinstetten, Germany), giving a total number of 8,500 NMR variables.
All NMR analyses were performed blindly with respect to case/control status. Further details on sample preparation, NMR data acquisition, and spectra processing are available in Additional file 1.

Statistical analysis

Orthogonal partial least-square (O-PLS)

O-PLS [41] analyses were conducted in order to build predictive sample classification models based on whole CPMG or NOESY NMR spectra to discriminate between HCC cases and controls, by relating the 8,500 NMR variables to case/control status. Results were visualized on score plots corresponding to sample projection onto the predictive axis and the first orthogonal component of the model. The metabolic signature discriminating HCC cases from controls was visualized by the corresponding loading plot. The optimal number of orthogonal components for building O-PLS models was selected using a 7-fold cross validation procedure. The associated R2 and Q2 parameters were calculated as a measure of the “goodness of fit and prediction”, i.e. the explained and predicted variances, respectively. The robustness of O-PLS models was further validated using permutations (1000 times) under the null hypothesis; for each permutated case/control labels, R2 and Q2 values were obtained and compared to the original ones, their decrease indicating the good quality of the model [42].

Metabolite paired difference analysis

The statistical recoupling of variables [43] procedure was first applied to reduce the 8,500 NMR variables into 285 intelligent buckets, or clusters of NMR variables, that correspond to reconstructions of peak entities. ANOVA models were then carried out on each of the 285 clusters of variables by modelling the case–control set by means of a random effect variable to account for the matching design of the study in ANOVA mixed-effect models. To correct for multiple testing, q values were determined using the Benjamini-Hochberg procedure [44] to control the false discovery rate with a threshold of 0.05. In this way, 96 clusters of NMR variables were found to be significantly associated with HCC outcome. Significant clusters of variables corresponding to different peaks of the same metabolite (based on the metabolite identification reported above) were combined into a single variable by summing up the bins intensities taking into consideration the number of homolog protons in the signal resonance. This procedure resulted in a list of 23 combined clusters of variables, 16 of which corresponded to distinct metabolite or lipid classes and were retained for further analyses, while five corresponded to other signals from mixed classes of lipids and two corresponded to the superimposition of signals from different metabolites.

Conditional logistic regression (CLR)

CLR models were used to quantify the associations between the 16 metabolites selected as described above and HCC risk by computing odds ratios (OR) and 95 % confidence intervals (95 % CIs). The metabolites were modeled as continuous variables with the OR corresponding to one standard deviation increase in metabolic intensity. CLR models were run conditioned on the matching factors (referred to as crude), and after adjustment for potential confounding variables (referred to as multivariable), i.e. body mass index (continuous), smoking status (current smokers, non-smokers, former smokers, unknown), lifetime alcohol drinking pattern (never drinkers, former drinkers, drinkers only at recruitment, lifetime drinkers), level of alcohol consumption at recruitment (g/d; continuous), serum-clot contact time (≤1 d or >1 d; a value that corresponds to the time between blood collection and blood centrifugation [45]), physical activity (inactive, moderately inactive, moderately active, active, missing), educational status (primary school, secondary school, professional school, longer education, unknown; as a proxy variable for socioeconomic status), and waist circumference (cm). The multivariable models for serum ethanol concentration were not adjusted for level of alcohol consumption at recruitment. For all metabolites, an additional CLR model with further adjustment for liver function score was also run.

Receiver operating characteristics (ROC)

ROC curves and corresponding area under the curve (AUC) were generated for several models including the AFP concentration, the liver function score, the multivariate metabolic profile using both the score values from the O-PLS classification model (referred as O-PLS score), and the cross-validated predicted-Y values (referred as O-PLS CV status) as well as a combination between the O-PLS CV status and AFP or the liver score. Combinations of the variables were obtained by summing up the O-PLS CV status with either AFP or the liver score after normalization of each variable to one unit variance. The specificity, sensitivity, and accuracy were obtained from the optimal cut-off point that corresponded to the minimal distance to the ideal point.

Subgroup analyses

Analyses stratified by hepatitis infection status (37 HCC cases Hep+, 77 HCC cases Hep), by liver function score (34 HCC cases with no liver damage, 80 HCC cases with probable to certain liver damage), by years between blood collection and cancer diagnosis with a cut-off at 2 years (22 HCC cases diagnosed <2 years, 92 HCC cases diagnosed ≥2 years from blood collection) were also conducted. In the grouping of cases diagnosed <2 years, the small sample size prevented model stability upon multivariable adjustment. Thus, only crude CLR models were run for this subgroup.
The analyses were performed using SIMCA-P 12 (Umetrics, Umeå, Sweden), MATLAB (The MathWorks Inc., Natick, MA) routines developed in-house, and R software [46] using the packages ‘splines’ and ‘survival’.

Results

Baseline characteristics of the study participants are summarized in Table 1. The median follow-up time between blood collection and HCC diagnosis (lag time) was 4.8 years. Serum blood samples of HCC cases were more likely to test positive for HBV or HCV infections (32.5 % vs. 3.2 % in the controls), and to have altered liver function as indicated by high liver function score (36.8 % vs. 14.4 % for probable liver damage and 33.3 % vs. 0.9 % for likely liver damage for cases vs. controls, respectively).
Table 1
Characteristics of study participants in the EPIC study. Values refer to either median (minimum – maximum range) or number (percentage)
 
HCC cases
Matched controls
Characteristics
(n = 114)a
(n = 222)
Gender
 Men
79 (69.3 %)
153 (68.9 %)
 Women
35 (30.7 %)
69 (31.1 %)
Age at blood collection (years)
60.6 (46.1–77)
60.2 (45.7–77)
Age at diagnosis (years)
65.3 (47.6–86.4)
 
Lag time (years)b
4.8 (0.01–13.3)
Body mass index (kg/m2)
27.7 (19.5–43.4)
26.5 (17.2–40.2)
Waist circumference (cm)
95 (68.5–140)
92.2 (64–130)
Smoking status
 Never
30 (26.3 %)
94 (42.3 %)
 Former
39 (34.2 %)
86 (38.7 %)
 Current smoker
44 (38.6 %)
41 (18.5 %)
Unknown
1 (0.9 %)
1 (0.5 %)
Serum-clot contact timec
 ≤1 day
84 (73.7 %)
169 (76.1 %)
 >1 day
30 (26.3 %)
53 (23.9 %)
Educational status
 Primary school/None
61 (53.5 %)
106 (47.7 %)
 Secondary school
4 (3.5 %)
23 (10.4 %)
 Technical/professional school
30 (26.3 %)
45 (20.3 %)
 Longer education
18 (15.8 %)
44 (19.8 %)
 Unknown
1 (0.9 %)
4 (1.8 %)
Physical activity
 Inactive
10 (8.8 %)
29 (13.1 %)
 Moderately inactive
36 (31.6 %)
71 (32 %)
 Moderately active
57 (50 %)
100 (45 %)
 Active
11 (9.6 %)
22 (9.9 %)
Lifetime alcohol drinking pattern
 Never drinkers
10 (8.8 %)
17 (7.7 %)
 Former drinkers
17 (14.9 %)
3 (1.4 %)
 Drinkers only at recruitment
17 (14.9 %)
43 (19.4 %)
 Lifetime drinkers
70 (61.4 %)
159 (71.6 %)
Alcohol intake at recruitment (g/day)
7.5 (0–141.3)
8.8 (0–108.6)
HBV/HCV status
 No
77 (67.5 %)
215 (96.8 %)
 Yes
37 (32.5 %)
7 (3.2 %)
Liver function scored
 No
34 (29.8 %)
188 (84.7 %)
 Probable
42 (36.8 %)
32 (14.4 %)
 Likely
38 (33.3 %)
2 (0.9 %)
AFP (ng/mL)
5.3 (0–18780)
3.25 (0–20.8)
aDistribution of HCC cancer cases across EPIC countries was 29 for Denmark, 20 for Germany, 11 for Greece, 19 for Italy, 4 for the Netherlands, 7 for Spain, 16 for Sweden, and 8 for the United Kingdom
bFollow-up time between blood collection and HCC diagnosis
cSerum-clot contact time refers to the time between blood collection and blood centrifugation for serum isolation
dA categorical liver function score describing liver damage was constructed based on values of albumin (<35 g/L), total bilirubin (>20.5 μmol/L), AST (>34 U/L), ALT (>55 U/L), GGT (men >64 U/L, women >36 U/L), and AP (>150 U/L), each contributing 1 point when outside of the normal range
The O-PLS analysis presented in Fig. 2a shows a metabolic profile discriminating between HCC cases and the matched controls (R2 = 35 %, Q2 = 21 %). The metabolic signature (Fig. 2b) associated with HCC occurrence presented (1) higher levels in the aromatic amino acids (AAA) tyrosine and phenylalanine, glutamate, acetate, citrate, glucose, propylene glycol, and ethanol; (2) lower levels in unsaturated lipids and VLDL, N-acetyl glycoproteins, choline, glutamine, acetone, mannose and the branched-chain amino-acids (BCAA) valine, leucine, and isoleucine levels, compared to the control group. The corresponding P values, q values, and fold changes of the metabolites are presented in Table 2. The ROC analyses (Fig. 2c) of the metabolic signature (O-PLS score) and of the cross-validated data (O-PLS CV status) presented an AUC of 85 % and 74 %, respectively (Table 3). The ROC parameters obtained from AFP compared to the combination of O-PLS CV status with AFP were increased after combining the variables (AUC 73 % vs. 75 %, specificity 65.3 % vs. 80.6 %, sensitivity 71.9 % vs. 75.4 %, accuracy 67.5 % vs. 78.9 %). Multivariable adjusted CLR models showed that AAA (per 1-SD), tyrosine (OR = 2.46; 95 % CI, 1.65–3.6), phenylalanine (OR = 2.07; 95 % CI, 1.40–3.06), glutamate (OR = 2.44; 95 % CI, 1.54–3.87), citrate (OR = 1.76; 95 % CI, 1.22–2.54), glucose (OR = 1.67; 95 % CI, 1.19–2.35), and propylene glycol (OR = 2.20; 95 % CI, 1.06–4.60) were associated with a statistically significant higher HCC risk. In contrast, BCAA, leucine (OR = 0.60; 95 % CI, 0.43–0.85), isoleucine (OR = 0.72; 95 % CI, 0.53–0.98), choline (OR = 0.45; 95 % CI, 0.31–0.65), N-acetyl glycoproteins (OR = 0.46; 95 % CI, 0.32–0.67), unsaturated lipids (OR = 0.36; 95 % CI, 0.21–0.63), and VLDL (OR = 0.52; 95 % CI, 0.36–0.74) were inversely associated with HCC risk (Table 4).
Table 2
P values, q values, and fold differences of metabolites observed to be statistically significantly different between HCC cases (n = 114) and matched controls (n = 222)
 
HCC cases vs. matched controls
Metabolites
P valuea
q valueb
Fold difference
Lipids = CH-CH2-CH=
9 × 10-11
3 × 10-08
0.84
N-acetyl glycoproteins
3 × 10-10
4 × 10-08
0.90
Unspecificc,d
4 × 10-10
4 × 10-08
0.82
Glutamate
4 × 10-09
3 × 10-07
1.22
Tyrosine
1 × 10-08
7 × 10-07
1.24
Choline
2 × 10-08
7 × 10-07
0.82
Lipids O-CH2
1 × 10-07
3 × 10-06
0.84
Phenylalanine
2 × 10-07
5 × 10-06
1.23
Unspecificc,e
4 × 10-07
9 × 10-06
0.84
Leucine
2 × 10-06
3 × 10-05
0.91
Lipids CH2-CH2-COOC
2 × 10-06
3 × 10-05
0.82
Unsaturated lipidsf
2 × 10-06
3 × 10-05
0.86
Isoleucine
2 × 10-05
2 × 10-04
0.90
Lipids CH2-CH=
2 × 10-05
2 × 10-04
0.94
CH3 bond of lipids (VLDL)g
2 × 10-05
2 × 10-04
0.87
Propylene glycol
7 × 10-05
6 × 10-04
1.45
Lipids (CH2)n
1 × 10-04
8 × 10-04
0.88
Ethanol
3 × 10-04
2 × 10-03
1.65
Glutamine
7 × 10-04
4 × 10-03
0.90
Glucose
8 × 10-04
4 × 10-03
1.10
Valine
3 × 10-03
1 × 10-02
0.93
Acetate
5 × 10-03
2 × 10-02
1.24
Citrate
1 × 10-02
4 × 10-02
1.08
aP value was obtained from ANOVA mixed-effects models
bq value corresponds to the value obtained after Benjamini-Hochberg correction for multiple testing
cMultiple assignments for the same cluster of variables due to spectral overlap
dAcetone and CH2-CH2-COOC of bound lipids
eMannose and glyceride of lipids
fUnsaturated lipids as estimated from the CH = CH signal of lipids
gPart of terminal lipids methyl signal corresponding to VLDLs
Table 3
AUC, specificity, sensitivity, and accuracy of the ROC models (in %)
 
All
Cases diagnosed <2 years
Cases diagnosed ≥2 years
 
AUC
Spe
Sen
Acc
AUC
Spe
Sen
Acc
AUC
Spe
Sen
Acc
AFPa
73
65.3
71.9
67.5
81
79
77.3
78.5
71
60.9
74
65.3
Liver function scoreb
80
84.7
70.2
79.8
79
88.4
68.2
81.5
80
83.8
70.6
79.3
O-PLS scorec
85
81.5
71.1
78.1
93
83.7
86.4
84.6
79
74.9
68.5
72.7
O-PLS CV statusd
74
68
68.4
68.1
82
100
63.6
87.7
71
68.7
67.4
68.3
O-PLS CV status + AFPe
75
80.6
75.4
78.9
82
100
63.6
87.7
73
70.9
70.6
70.8
O-PLS CV status + liver scoref
82
68
68.4
68.1
84
86
77.3
83
80
83.8
70.7
79.3
AUC, Area under the curve; Spe, Specificity; Sen, Sensitivity; Acc, Accuracy of the model
aModel based on the serum AFP concentration
bModel based on the liver function score
cModel based on the score of the O-PLS analysis
dModel based on the cross-validated (CV) case/control status of the O-PLS analysis
eCombination between the CV status of the O-PLS and the AFP variable
fCombination between the CV status of the O-PLS and the liver function score
Table 4
Odds ratios [OR (95 % confidence intervals)] of HCC risk by serum metabolites
 
Crudea
Multivariableb
Multivariable + liver function scorec
Multivariable for non-hepatitis infected casesd
Crude <2 years follow-upe
Crude ≥2 years follow-upf
Multivariable ≥2 years follow-upg
114 cases
114 cases
114 cases
77 cases
22 cases
92 cases
92 cases
N-acetyl glycoproteins
0.47 (0.36–0.62)***
0.46 (0.32–0.67)***
0.76 (0.49–1.17)
0.74 (0.49–1.14)
0.64 (0.39–1.05)
0.43 (0.31–0.60)***
0.35 (0.21–0.59)***
Glutamate
2.37 (1.64–3.41)***
2.44 (1.54–3.87)***
1.46 (0.80–2.63)
2.06 (1.18–3.61)*
4.87 (1.26–18.82)*
2.05 (1.43–2.94)***
2.13 (1.30–3.49)**
Tyrosine
2.00 (1.51–2.65)***
2.46 (1.65–3.66)***
1.58 (0.98–2.56)
1.42 (0.83–2.43)
3.90 (1.50–10.14)**
1.83 (1.36–2.47)***
2.23 (1.41–3.54)***
Choline
0.37 (0.27–0.51)***
0.45 (0.31–0.65)***
0.83 (0.52–1.32)
0.68 (0.42–1.12)
0.30 (0.13–0.66)**
0.39 (0.27–0.56)***
0.45 (0.29–0.71)***
Phenylalanine
1.83 (1.37–2.44)***
2.07 (1.40–3.06)***
1.75 (1.04–2.94)*
1.59 (0.94–2.67)
3.59 (1.29–10)*
1.62 (1.20–2.19)**
1.64 (1.07–2.54)*
Leucine
0.54 (0.41–0.71)***
0.60 (0.43–0.85)**
0.67 (0.43–1.05)
0.68 (0.43–1.09)
0.80 (0.48–1.33)
0.47 (0.34–0.66)***
0.51 (0.33–0.80)**
Unsaturated lipids
0.29 (0.18–0.46)***
0.36 (0.21–0.63)***
0.77 (0.41–1.44)
0.59 (0.29–1.23)
0.16 (0.04–0.60)**
0.33 (0.20–0.53)***
0.46 (0.24–0.84)*
Isoleucine
0.64 (0.49–0.83)***
0.72 (0.53–0.98)*
0.84 (0.56–1.26)
0.76 (0.50–1.15)
0.84 (0.50–1.42)
0.59 (0.44–0.80)***
0.66 (0.45–0.96)*
VLDL
0.58 (0.45–0.76)***
0.52 (0.36–0.74)***
0.77 (0.51–1.15)
0.74 (0.45–1.20)
0.30 (0.12–0.72)**
0.64 (0.49–0.85)**
0.58 (0.39–0.87)**
Propylene glycol
3.07 (1.58–5.97)***
2.20 (1.06–4.60)*
1.00 (0.59–1.69)
1.27 (0.73–2.21)
727.9 (1.56–340468)*
2.11 (1.24–3.58)**
1.46 (0.83–2.55)
Ethanolh
1.76 (1.13–2.74)*
1.46 (0.97–2.21)
1.02 (0.69–1.51)
1.10 (0.74–1.65)
126.1 (1.3–12227)*
1.66 (1.09–2.53)*
1.40 (0.95–2.08)
Glutamine
0.66 (0.51–0.84)**
0.75 (0.54–1.03)
0.96 (0.64–1.44)
0.56 (0.34–0.92)*
0.60 (0.30–1.15)
0.67 (0.51–0.88)**
0.74 (0.51–1.07)
Glucose
1.53 (1.18–1.99)**
1.67 (1.19–2.35)**
1.28 (0.86–1.92)
1.24 (0.80–1.92)
1.95 (0.98–3.88)
1.47 (1.10–1.96)**
1.77 (1.16–2.69)**
Valine
0.69 (0.54–0.88)**
0.82 (0.61–1.11)
0.90 (0.61–1.34)
0.78 (0.51–1.18)
0.75 (0.44–1.27)
0.68 (0.52–0.89)**
0.79 (0.55–1.13)
Acetate
1.40 (1.08–1.82)*
1.20 (0.90–1.60)
0.92 (0.58–1.46)
1.01 (0.63–1.61)
3.04 (0.45–20.69)
1.32 (1.01–1.75)*
1.12 (0.76–1.63)
Citrate
1.36 (1.06–1.76)*
1.76 (1.22–2.54)**
1.88 (1.14–3.11)*
1.15 (0.65–2.01)
2.19 (1.10–4.35)*
1.22 (0.93–1.61)
1.37 (0.91–2.06)
*P value <0.05; **P value <0.01; ***P value <0.001
aModel 1, Crude OR based on logistic regression conditioned on matching factors (sex, age, date and time of blood collection, fasting status, and menopausal status and use of hormones)
bModel 2, OR based on multivariable CLR adjusted for smoking status, ethanol at recruitment, lifetime alcohol, educational status, physical activity, body mass index, serum-clot contact time, and waist circumference
cModel 3, as in model 2 and additionally adjusted for liver function score
dModel 4, as model 2 but obtained on HCC cases free of HBV/HCV infection and their matched controls (n = 224)
eModel 5, Crude OR obtained on HCC cases diagnosed <2 years after blood collection and their matched controls
fModel 6, Crude OR obtained on HCC cases diagnosed ≥2 years after blood collection and their matched controls
gModel 7, as in model 2 but obtained on HCC cases diagnosed ≥2 years after blood collection and their matched controls
hOR of ethanol from multivariable CLR model are obtained after adjustment for variables as mentioned for model 2 expect for lifetime alcohol
The O-PLS analyses stratified by hepatitis infection status of the cases (Fig. 3a,b) presented distinct metabolic signatures from hepatitis-infected HCC cases (R2 = 45 %, Q2 = 34 %) and hepatitis-free HCC cases (R2 = 28 %, Q2 = 12 %). Hepatitis-infected HCC cases presented (1) higher levels of AAA, glucose, and citrate and (2) lower VLDL and unsaturated lipids levels, while on the other hand HCC hepatitis-free cases were characterized by (1) higher levels in ethanol and glutamate and (2) lower levels in glutamine, BCAA, and choline. In hepatitis-free HCC cases, the risk associations of glutamine (OR = 0.56; 95 % CI, 0.34–0.92) and glutamate (OR = 2.06; 95 % CI, 1.18–3.61) were significantly different from matched controls (Table 4).
Figure 3c shows O-PLS subgroup analysis of HCC cases with abnormal liver function (score ≥1). A robust model was obtained (R2 = 58 %, Q2 = 43 %) and the metabolic signature was similar to that including all samples (Fig. 2b). However, no significant model was obtained from HCC cases with a normal liver function (score = 0) only (data not shown). Table 4 shows results of multivariable CLR additionally adjusted for liver function score for which only citrate (OR = 1.88; 95 % CI, 1.14–3.11) and phenylalanine (OR = 1.75; 95 % CI, 1.04–2.94) remained significantly associated with HCC risk.
Figure 4 presents the O-PLS and ROC analyses stratified by lag time between blood collection and diagnosis. The metabolic signature of HCC cases diagnosed within 2 years after blood collection is characterized by (1) higher levels in AAA and glutamate, and (2) lower levels in unsaturated lipids and choline while in addition, the metabolic signature of HCC diagnosed later (≥2 years) presented (1) higher levels in glucose, ethanol, and propylene glycol and (2) lower levels in BCAA and N-acetyl glycoproteins. Among the cases diagnosed <2 years from recruitment, the AUC of ROC curves from the O-PLS metabolic signature and from O-PLS CV data were 93 % and 82 %, respectively (Fig. 4c).
Higher ROC parameters (Table 3) were found for O-PLS CV status compared to AFP and the liver score (O-PLS CV status vs. AFP, liver score: AUC 82 % vs. 81 %, 79 %; specificity 100 % vs. 79 %, 88.4 %; sensitivity 63.6 % vs. 77.3 %, 68.2 %; accuracy 87.7 % vs. 78.5 %, 81.5 %). However, the parameters did not improve after combining O-PLS CV status with AFP while they were slightly improved after combining O-PLS CV status with the liver score (AUC 84 %; specificity 86 %; sensitivity 77.3 %; accuracy 83 %). ROC analysis of the cases diagnosed ≥2 years from recruitment showed an AUC of 79 % for the O-PLS metabolic signature and 71 % for the O-PLS CV status. Combining the O-PLS CV status with AFP improved the ROC parameters in comparison to AFP alone or O-PLS CV alone (O-PLS CV status + AFP vs. AFP, O-PLS CV: AUC: 73 % vs. 71 %, 71 %; specificity: 70.9 % vs. 60.9 %, 68.7 %; sensitivity: 70.6 % vs. 74 %, 67.4 %; accuracy: 70.8 % vs. 65.3 %, 68.3 %). However, the best model was obtained from the liver score (AUC 80 %, specificity 83.8 %, sensitivity 70.6 %, accuracy 79.3 %).
Findings for subgroup CLR analyses of individual metabolites by lag time of diagnosis from recruitment show a significant HCC risk association for citrate in cases diagnosed <2 years from recruitment (OR = 2.19; 95 % CI, 1.10–4.35), while for cases diagnosed ≥2 years from recruitment significant HCC risk associations were observed for glucose (OR = 1.47; 95 % CI, 1.10–1.96), acetate (OR = 1.32; 95 % CI, 1.01–1.75), N-acetyl glycoproteins (OR = 0.43; 95 % CI, 0.31–0.60), BCAA (valine OR = 0.68; 95 % CI, 0.52–0.89); leucine (OR = 0.47; 95 % CI, 0.34–0.66); isoleucine (OR = 0.59; 95 % CI, 0.44–0.80), and glutamine (OR = 0.67; 95 % CI, 0.51–0.88) (Table 4).

Discussion

This study is, to the best of our knowledge, the first NMR metabolomic analysis based on subjects from a prospective cohort study on Western European populations for epidemiology of liver cancer. We have identified a number of metabolites that differed between HCC cases and corresponding matched controls. As concerns the specificity of these associations, we note that an analogous study was conducted in parallel on extrahepatic/intrahepatic bile duct carcinomas without providing any significant results (data not shown). We also note that the impact of long-term storage of EPIC samples as well as other potential sources of systematic variations of the metabolic profiles has been thoroughly detailed earlier [45].
O-PLS analysis showed a clear discrimination between cases and controls with somewhat different metabolomic profiles with respect to the length of time from blood collection to diagnosis, hepatitis infection status, and liver function. Importantly, this study showed that consideration of metabolomic profiles can improve HCC diagnosis beyond that provided by AFP and liver enzyme levels, which are currently the most common HCC biomarkers often applied in clinical practice.
The liver is central for the metabolism of carbohydrates, fats and proteins, and also plays key roles in detoxification and hormone production. Thus, a degree of metabolic dysregulation would be expected with liver diseases, particularly HCC. For this reason, the application of metabolomic technologies may be able to provide some insight into the etiology and mechanisms of HCC and, possibly, the identification of early diagnostic biomarkers or biomarker patterns characteristic of cancer at this anatomical site.
To date, three NMR or NMR/mass spectrometry, serum-based metabolomic studies have been conducted looking specifically at HCC [20, 23, 24]. All three case–control studies were based on sera collected from HCC cases post-diagnosis. The comparison group in one of the studies was hepatitis-infected subjects [20], while that of the others were cirrhotic patients [23, 24]. The studies identified potential (1) impairment of the tricarboxylic acid cycle, increased lipid catabolism, and elevation of essential amino acids [20], and (2) defects on ammonium detoxification and increased fatty acid beta-oxidation [24] in HCC. The fundamental design differences with the present study are that the latter is based on prospectively identified HCC cases, such that metabolomic profiles are likely indicative of pre-diagnostic changes, and that the matched control subjects were cancer-free cohort participants. The key metabolic alterations observed are related to changes in amino acid, polyunsaturated lipid, acetate, and citrate metabolism, among the 16 individual metabolites highlighted here. Because our study is nested within the prospective EPIC cohort, which has detailed information on dietary and lifestyle factors and measured anthropometry, we were able to make statistical adjustments for many important confounding variables such as smoking status, alcohol consumption and habits, physical activity, educational attainment (as a proxy marker for socioeconomic status), body mass index, and waist circumference.
Of particular note is our observation of a 0.82-fold reduction in choline in HCC cases (Table 2), meaning a significant inverse HCC risk association for this compound (Table 4; OR = 0.45; 95 % CI, 0.31–0.65). In animal studies, choline deficiency has been shown to cause liver damage, oxidative stress, and spontaneous liver cancer [4749]. In human studies, HCC has been associated with a down regulation in choline metabolism [25].
Also interesting is our identification of circulating ethanol as a strong HCC risk factor, alcohol being a major lifestyle risk factor for this disease. We also observed a shift, in terms of fold difference between HCC cases and their matched controls, from glutamine to glutamate, indicating a possible defect in ammonium detoxification [50], as also observed in the study by Nahon et al. [24]. It is of interest that Nahon et al. [24] observed this shift comparing HCC cases to cirrhotic controls, while our findings indicate that this important change may actually be present for some time prior to diagnosis. In the study by Gao et al. [20], higher levels of AAA were associated with liver cirrhosis and HCC, together with lower levels of BCAA, choline, and unsaturated lipids. The same changes were observed suggesting an important alteration of amino acid and lipid metabolism in the progression to HCC.
An interesting observation in the present study was a strong, significant positive HCC risk association for the exogenous metabolite propylene glycol. Identification of propylene glycol in human serum is not uncommon [51], and it is thought to derive largely from pharmaceutical use since it is widely used as a solvent in many intravenous, oral, and topical pharmaceutical preparations (as well as in other general products including cosmetics, food, and toothpastes). The liver of an adult with normal liver and kidney functions will metabolize propylene glycol into lactate, acetate, and pyruvate within several hours [52]. Therefore, high levels of propylene glycol could be reflective of medication use, possibly in participants with liver damage or due to its simple accumulation resulting from impaired liver function. Despite the prospective nature of our study, it may be speculated that HCC cases may have encountered some symptoms, which may have prompted medical surveillance and/or alteration of dietary/lifestyle habits (e.g. reduced alcohol intake or smoking cessation). Yet, such changes would likely bias risk estimates towards the null or be unrelated to the disease outcome.
In addition to its prospective design, availability of detailed pre-diagnostic lifestyle/dietary data, and anthropometric measures, additional strengths of our study include the ability to consider liver function parameters based on a score developed from clinically relevant liver enzyme concentrations. The assumption is that decreased liver function is associated with a greater degree of liver damage. From our findings, it is apparent that the metabolic pattern associated with HCC may be reflective of liver dysfunction, as suggested by the stratified analysis on the liver function score. These results support the fact that HCC largely arises from a background of increasingly severe liver damage. Indeed, the process ending with HCC is considered to be gradual, involving infection by hepatitis viruses or the development of fatty liver diseases or cirrhosis [53]. Each part of the process may be characterized by alterations in metabolic factors, which may be detectable by metabolomic approaches [5458]. Due to this gradual process, we note that longer follow-up time would be required in order to thoroughly assess, prior to any liver damage, the specificity of the identified HCC risk associations. Our study was composed of a large number of HCC cases that were not infected with either hepatitis B or C. Thus, we attempted to determine whether metabolomic differences could be observed in the absence of these predominant HCC risk factors. Although exclusion of hepatitis-positive cases attenuated some of our findings and resulted in loss of significance for specific metabolites, strong associations were observed for glutamate and glutamine. This is indicative of a potential defect in ammonium detoxification in non-hepatitis HCC. This observation deserves further in-depth investigation.
In our study, we were also interested in comparing metabolic changes preceding cancer diagnosis by several years. Thus, we conducted stratified analysis by lag time between blood collection and diagnosis, which showed specific metabolic changes according to follow-up time. However, a key limitation of the present study is the lack of any clinical data, assessment of any medication usage, or subgroup analyses based on pathways of HCC development. The metabolite changes associated with the later cases are more likely to be informative on the etiology and/or risk exposure (e.g. dietary components, environmental, lifestyle, and pollutants), while metabolic changes in cases diagnosed <2 years after recruitment likely reflect a direct influence of the tumor.

Conclusion

For the first time, a metabolic pattern based on serum samples was identified to be associated with HCC risk within a large prospective study. Several metabolites associated with either an increased or decreased HCC risk have been highlighted. The majority of associations remained significant after controlling for potential confounders and consideration of correction for multiple testing. The results suggest that metabolic patterns can provide meaningful etiologic insight into HCC development and can potentially be used to detect this cancer in its early stages, even several years prior to clinical diagnosis.

Acknowledgments

Guarantor of the article: Bénédicte Elena-Herrmann, PhD and Mazda Jenab, PhD.

Funding

This work was supported by the French National Cancer Institute (L’Institut National du Cancer; INCA; grant number 2009-139; PI: M. Jenab). AF received financial support (BDI fellowship) from the Centre National de la Recherche Scientifique (CNRS) and Bruker Biospin. The coordination of EPIC is financially supported by the European Commission (DG-SANCO) and the International Agency for Research on Cancer. The national cohorts are supported by Danish Cancer Society (Denmark); Ligue Contre le Cancer, Institut Gustave Roussy, Mutuelle Générale de l’Education Nationale, and Institut National de la Santé et de la Recherche Médicale (INSERM) (France); Deutsche Krebshilfe, Deutsches Krebsforschungszentrum (DKFZ), and Federal Ministry of Education and Research (Germany); Hellenic Health Foundation (Greece); Italian Association for Research on Cancer (AIRC), National Research Council, Associazione Italiana per la Ricerca sul Cancro-AIRC-Italy, and AIRE-ONLUS Ragusa, AVIS Ragusa, Sicilian Government (Italy); Dutch Ministry of Public Health, Welfare and Sports (VWS), Netherlands Cancer Registry (NKR), LK Research Funds, Dutch Prevention Funds, Dutch ZON (Zorg Onderzoek Nederland), World Cancer Research Fund (WCRF), and Statistics Netherlands (the Netherlands); European Research Council (ERC; grant number ERC-2009-AdG 232997) and Nordforsk, and Nordic Center of Excellence Programme on Food, Nutrition and Health (Norway); Health Research Fund (FIS), Regional Governments of Andalucía, Asturias, Basque Country, Murcia (No. 6236) and Navarra, and ISCIII RETIC (RD06/0020) (Spain); Swedish Cancer Society, Swedish Scientific Council, and Regional Government of Skåne and Västerbotten (Sweden); Cancer Research UK, Medical Research Council, Stroke Association, British Heart Foundation, Department of Health, Food Standards Agency, and Wellcome Trust (UK). The funders had no role in the study design, data collection, analysis, and interpretation presented in this article. They were neither involved in the writing of the manuscript, nor in the decision to submit it for publication.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

The authors’ responsibilities were as follows: ER is the overall PI of the EPIC study which is jointly coordinated from ICL (ER) and IARC (IR); MJ and BE-H conceptualized, designed, obtained funding for, and implemented/managed the present research; AF, CP, and BE-H performed the laboratory analyses; AF and PF conducted the statistical analyses; AF, MJ, VF, TDS, MS, and BE-H contributed to the writing of the manuscript and data interpretation. Contributing authors from each collaborating centre provided the original data and biological samples, information on the respective populations, advice on study design/analysis, and interpretation of the results. All authors contributed comments on the draft manuscript and provided an approval of the final version of the manuscript for publication.
Literatur
1.
Zurück zum Zitat Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide 2013. http://globocan.iarc.fr, accessed on 30/01/2015. Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide 2013. http://​globocan.​iarc.​fr, accessed on 30/01/2015.
3.
Zurück zum Zitat Chen Y, Wang X, Wang J, Yan Z, Luo J. Excess body weight and the risk of primary liver cancer: an updated meta-analysis of prospective studies. Eur J Cancer. 2012;48:2137–45.CrossRefPubMed Chen Y, Wang X, Wang J, Yan Z, Luo J. Excess body weight and the risk of primary liver cancer: an updated meta-analysis of prospective studies. Eur J Cancer. 2012;48:2137–45.CrossRefPubMed
4.
Zurück zum Zitat Chuang SC, La Vecchia C, Boffetta P. Liver cancer: descriptive epidemiology and risk factors other than HBV and HCV infection. Cancer Lett. 2009;286:9–14.CrossRefPubMed Chuang SC, La Vecchia C, Boffetta P. Liver cancer: descriptive epidemiology and risk factors other than HBV and HCV infection. Cancer Lett. 2009;286:9–14.CrossRefPubMed
5.
Zurück zum Zitat Wang P, Kang D, Cao W, Wang Y, Liu Z. Diabetes mellitus and risk of hepatocellular carcinoma: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2012;28:109–22.CrossRefPubMed Wang P, Kang D, Cao W, Wang Y, Liu Z. Diabetes mellitus and risk of hepatocellular carcinoma: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2012;28:109–22.CrossRefPubMed
6.
7.
Zurück zum Zitat Nicholson JK, Lindon JC, Holmes E. ‘Metabonomics’: understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica. 1999;29:1181–9.CrossRefPubMed Nicholson JK, Lindon JC, Holmes E. ‘Metabonomics’: understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica. 1999;29:1181–9.CrossRefPubMed
8.
Zurück zum Zitat Nicholson JK, Holmes E, Kinross JM, Darzi AW, Takats Z, Lindon JC. Metabolic phenotyping in clinical and surgical environments. Nature. 2012;491:384–92.CrossRefPubMed Nicholson JK, Holmes E, Kinross JM, Darzi AW, Takats Z, Lindon JC. Metabolic phenotyping in clinical and surgical environments. Nature. 2012;491:384–92.CrossRefPubMed
9.
Zurück zum Zitat Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan D, Joost HG, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62:639–48.CrossRefPubMedPubMedCentral Floegel A, Stefan N, Yu Z, Muhlenbruch K, Drogan D, Joost HG, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62:639–48.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Griffin JL, Atherton H, Shockor J, Atzori L. Metabolomics as a tool for cardiac research. Nat Rev Cardiol. 2011;8:630–43.CrossRefPubMed Griffin JL, Atherton H, Shockor J, Atzori L. Metabolomics as a tool for cardiac research. Nat Rev Cardiol. 2011;8:630–43.CrossRefPubMed
11.
Zurück zum Zitat Griffin JL, Shockcor JP. Metabolic profiles of cancer cells. Nat Rev Cancer. 2004;4:551–61.CrossRefPubMed Griffin JL, Shockcor JP. Metabolic profiles of cancer cells. Nat Rev Cancer. 2004;4:551–61.CrossRefPubMed
12.
Zurück zum Zitat Mayers JR, Wu C, Clish CB, Kraft P, Torrence ME, Fiske BP, et al. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med. 2014;20:1193–8.CrossRefPubMedPubMedCentral Mayers JR, Wu C, Clish CB, Kraft P, Torrence ME, Fiske BP, et al. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med. 2014;20:1193–8.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Shah SH, Sun JL, Stevens RD, Bain JR, Muehlbauer MJ, Pieper KS, et al. Baseline metabolomic profiles predict cardiovascular events in patients at risk for coronary artery disease. Am Heart J. 2012;163:844–50.CrossRefPubMed Shah SH, Sun JL, Stevens RD, Bain JR, Muehlbauer MJ, Pieper KS, et al. Baseline metabolomic profiles predict cardiovascular events in patients at risk for coronary artery disease. Am Heart J. 2012;163:844–50.CrossRefPubMed
14.
15.
Zurück zum Zitat Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–53.CrossRefPubMedPubMedCentral Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–53.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Beyoglu D, Imbeaud S, Maurhofer O, Bioulac-Sage P, Zucman-Rossi J, Dufour JF, et al. Tissue metabolomics of hepatocellular carcinoma: tumor energy metabolism and the role of transcriptomic classification. Hepatology. 2013;58:229–38.CrossRefPubMedPubMedCentral Beyoglu D, Imbeaud S, Maurhofer O, Bioulac-Sage P, Zucman-Rossi J, Dufour JF, et al. Tissue metabolomics of hepatocellular carcinoma: tumor energy metabolism and the role of transcriptomic classification. Hepatology. 2013;58:229–38.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Bowers J, Hughes E, Skill N, Maluccio M, Raftery D. Detection of hepatocellular carcinoma in hepatitis C patients: biomarker discovery by LC-MS. J Chromatogr B. 2014;966:154–62.CrossRef Bowers J, Hughes E, Skill N, Maluccio M, Raftery D. Detection of hepatocellular carcinoma in hepatitis C patients: biomarker discovery by LC-MS. J Chromatogr B. 2014;966:154–62.CrossRef
18.
Zurück zum Zitat Chen J, Wang WZ, Lv S, Yin PY, Zhao XJ, Lu X, et al. Metabonomics study of liver cancer based on ultra performance liquid chromatography coupled to mass spectrometry with HILIC and RPLC separations. Anal Chim Acta. 2009;650:3–9.CrossRefPubMed Chen J, Wang WZ, Lv S, Yin PY, Zhao XJ, Lu X, et al. Metabonomics study of liver cancer based on ultra performance liquid chromatography coupled to mass spectrometry with HILIC and RPLC separations. Anal Chim Acta. 2009;650:3–9.CrossRefPubMed
19.
Zurück zum Zitat da Costa AN, Pontoizeau C, Plymoth A, Santos-Silva D, Mendy M, Sangrajrang S, et al. A multi-marker approach for early detection of HBV-related hepatocellular carcinoma in areas of high incidence. Eur J Cancer. 2012;48:S169–70.CrossRef da Costa AN, Pontoizeau C, Plymoth A, Santos-Silva D, Mendy M, Sangrajrang S, et al. A multi-marker approach for early detection of HBV-related hepatocellular carcinoma in areas of high incidence. Eur J Cancer. 2012;48:S169–70.CrossRef
20.
Zurück zum Zitat Gao HC, Lu Q, Liu X, Cong H, Zhao LC, Wang HM, et al. Application of H-1 NMR-based metabonomics in the study of metabolic profiling of human hepatocellular carcinoma and liver cirrhosis. Cancer Sci. 2009;100:782–5.CrossRefPubMed Gao HC, Lu Q, Liu X, Cong H, Zhao LC, Wang HM, et al. Application of H-1 NMR-based metabonomics in the study of metabolic profiling of human hepatocellular carcinoma and liver cirrhosis. Cancer Sci. 2009;100:782–5.CrossRefPubMed
21.
Zurück zum Zitat Huang Q, Tan YX, Yin PY, Ye GZ, Gao P, Lu X, et al. Metabolic characterization of hepatocellular carcinoma using nontargeted tissue metabolomics. Cancer Res. 2013;73:4992–5002.CrossRefPubMed Huang Q, Tan YX, Yin PY, Ye GZ, Gao P, Lu X, et al. Metabolic characterization of hepatocellular carcinoma using nontargeted tissue metabolomics. Cancer Res. 2013;73:4992–5002.CrossRefPubMed
22.
Zurück zum Zitat Liu SY, Zhang RL, Kang H, Fan ZJ, Du Z. Human liver tissue metabolic profiling research on hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol. 2013;19:3423–32.CrossRefPubMedPubMedCentral Liu SY, Zhang RL, Kang H, Fan ZJ, Du Z. Human liver tissue metabolic profiling research on hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol. 2013;19:3423–32.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Liu Y, Hong Z, Tan G, Dong X, Yang G, Zhao L, et al. NMR and LC/MS-based global metabolomics to identify serum biomarkers differentiating hepatocellular carcinoma from liver cirrhosis. Int J Cancer. 2014;135:658–68.CrossRefPubMed Liu Y, Hong Z, Tan G, Dong X, Yang G, Zhao L, et al. NMR and LC/MS-based global metabolomics to identify serum biomarkers differentiating hepatocellular carcinoma from liver cirrhosis. Int J Cancer. 2014;135:658–68.CrossRefPubMed
24.
Zurück zum Zitat Nahon P, Amathieu R, Triba MN, Bouchemal N, Nault JC, Ziol M, et al. Identification of serum proton NMR metabolomic fingerprints associated with hepatocellular carcinoma in patients with alcoholic cirrhosis. Clin Cancer Res. 2012;18:6714–22.CrossRefPubMed Nahon P, Amathieu R, Triba MN, Bouchemal N, Nault JC, Ziol M, et al. Identification of serum proton NMR metabolomic fingerprints associated with hepatocellular carcinoma in patients with alcoholic cirrhosis. Clin Cancer Res. 2012;18:6714–22.CrossRefPubMed
25.
Zurück zum Zitat Patterson AD, Maurhofer O, Beyoglu D, Lanz C, Krausz KW, Pabst T, et al. Aberrant lipid metabolism in hepatocellular carcinoma revealed by plasma metabolomics and lipid profiling. Cancer Res. 2011;71:6590–600.CrossRefPubMedPubMedCentral Patterson AD, Maurhofer O, Beyoglu D, Lanz C, Krausz KW, Pabst T, et al. Aberrant lipid metabolism in hepatocellular carcinoma revealed by plasma metabolomics and lipid profiling. Cancer Res. 2011;71:6590–600.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Ressom HW, Xiao JF, Tuli L, Varghese RS, Zhou B, Tsai TH, et al. Utilization of metabolomics to identify serum biomarkers for hepatocellular carcinoma in patients with liver cirrhosis. Anal Chim Acta. 2012;743:90–100.CrossRefPubMed Ressom HW, Xiao JF, Tuli L, Varghese RS, Zhou B, Tsai TH, et al. Utilization of metabolomics to identify serum biomarkers for hepatocellular carcinoma in patients with liver cirrhosis. Anal Chim Acta. 2012;743:90–100.CrossRefPubMed
27.
Zurück zum Zitat Soga T, Sugimoto M, Honma M, Mori M, Igarashi K, Kashikura K, et al. Serum metabolomics reveals gamma-glutamyl dipeptides as biomarkers for discrimination among different forms of liver disease. J Hepatology. 2011;55:896–905.CrossRef Soga T, Sugimoto M, Honma M, Mori M, Igarashi K, Kashikura K, et al. Serum metabolomics reveals gamma-glutamyl dipeptides as biomarkers for discrimination among different forms of liver disease. J Hepatology. 2011;55:896–905.CrossRef
28.
Zurück zum Zitat Tan YX, Yin PY, Tang L, Xing WB, Huang Q, Cao D, et al. Metabolomics study of stepwise hepatocarcinogenesis from the model rats to patients: potential biomarkers effective for small hepatocellular carcinoma diagnosis. Mol Cell Proteomics. 2012;11:M111.010694.CrossRefPubMed Tan YX, Yin PY, Tang L, Xing WB, Huang Q, Cao D, et al. Metabolomics study of stepwise hepatocarcinogenesis from the model rats to patients: potential biomarkers effective for small hepatocellular carcinoma diagnosis. Mol Cell Proteomics. 2012;11:M111.010694.CrossRefPubMed
29.
Zurück zum Zitat Wu H, Xue RY, Dong L, Liu TT, Deng CH, Zeng HZ, et al. Metabolomic profiling of human urine in hepatocellular carcinoma patients using gas chromatography/mass spectrometry. Anal Chim Acta. 2009;648:98–104.CrossRefPubMed Wu H, Xue RY, Dong L, Liu TT, Deng CH, Zeng HZ, et al. Metabolomic profiling of human urine in hepatocellular carcinoma patients using gas chromatography/mass spectrometry. Anal Chim Acta. 2009;648:98–104.CrossRefPubMed
30.
Zurück zum Zitat Xiao JF, Varghese RS, Zhou B, Ranjbar MRN, Zhao Y, Tsai TH, et al. LC-MS based serum metabolomics for identification of hepatocellular carcinoma biomarkers in Egyptian cohort. J Proteome Res. 2012;11:5914–23.PubMedPubMedCentral Xiao JF, Varghese RS, Zhou B, Ranjbar MRN, Zhao Y, Tsai TH, et al. LC-MS based serum metabolomics for identification of hepatocellular carcinoma biomarkers in Egyptian cohort. J Proteome Res. 2012;11:5914–23.PubMedPubMedCentral
31.
Zurück zum Zitat Zhou LN, Wang QC, Yin PY, Xing WB, Wu ZM, Chen SL, et al. Serum metabolomics reveals the deregulation of fatty acids metabolism in hepatocellular carcinoma and chronic liver diseases. Anal Bioanal Chem. 2012;403:203–13.CrossRefPubMed Zhou LN, Wang QC, Yin PY, Xing WB, Wu ZM, Chen SL, et al. Serum metabolomics reveals the deregulation of fatty acids metabolism in hepatocellular carcinoma and chronic liver diseases. Anal Bioanal Chem. 2012;403:203–13.CrossRefPubMed
32.
Zurück zum Zitat Fedirko V, Duarte-Salles T, Bamia C, Trichopoulou A, Aleksandrova K, Trichopoulos D, et al. Prediagnostic circulating vitamin D levels and risk of hepatocellular carcinoma in European populations: a nested case-control study. Hepatology. 2014;60:1222–30.CrossRefPubMed Fedirko V, Duarte-Salles T, Bamia C, Trichopoulou A, Aleksandrova K, Trichopoulos D, et al. Prediagnostic circulating vitamin D levels and risk of hepatocellular carcinoma in European populations: a nested case-control study. Hepatology. 2014;60:1222–30.CrossRefPubMed
33.
Zurück zum Zitat Lai GY, Weinstein SJ, Albanes D, Taylor PR, Virtamo J, McGlynn KA, et al. Association of serum alpha-tocopherol, beta-carotene, and retinol with liver cancer incidence and chronic liver disease mortality. Br J Cancer. 2014;111:2163–71.CrossRefPubMedPubMedCentral Lai GY, Weinstein SJ, Albanes D, Taylor PR, Virtamo J, McGlynn KA, et al. Association of serum alpha-tocopherol, beta-carotene, and retinol with liver cancer incidence and chronic liver disease mortality. Br J Cancer. 2014;111:2163–71.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Lukanova A, Becker S, Husing A, Schock H, Fedirko V, Trepo E, et al. Prediagnostic plasma testosterone, sex hormone-binding globulin, IGF-I and hepatocellular carcinoma: etiological factors or risk markers? Int J Cancer. 2013;134:164–73.CrossRefPubMed Lukanova A, Becker S, Husing A, Schock H, Fedirko V, Trepo E, et al. Prediagnostic plasma testosterone, sex hormone-binding globulin, IGF-I and hepatocellular carcinoma: etiological factors or risk markers? Int J Cancer. 2013;134:164–73.CrossRefPubMed
35.
Zurück zum Zitat Riboli E, Hunt KJ, Slimani N, Ferrari P, Norat T, Fahey M, et al. European prospective investigation into cancer and nutrition (EPIC): study populations and data collection. Public Health Nutr. 2002;5:1113–24.CrossRefPubMed Riboli E, Hunt KJ, Slimani N, Ferrari P, Norat T, Fahey M, et al. European prospective investigation into cancer and nutrition (EPIC): study populations and data collection. Public Health Nutr. 2002;5:1113–24.CrossRefPubMed
36.
Zurück zum Zitat Trichopoulos D, Bamia C, Lagiou P, Fedirko V, Trepo E, Jenab M, et al. Hepatocellular carcinoma risk factors and disease burden in a European cohort: a nested case-control study. J Natl Cancer Inst. 2011;103:1686–95.CrossRefPubMedPubMedCentral Trichopoulos D, Bamia C, Lagiou P, Fedirko V, Trepo E, Jenab M, et al. Hepatocellular carcinoma risk factors and disease burden in a European cohort: a nested case-control study. J Natl Cancer Inst. 2011;103:1686–95.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Fedirko V, Trichopolou A, Bamia C, Duarte-Salles T, Trepo E, Aleksandrova K, et al. Consumption of fish and meats and risk of hepatocellular carcinoma: the European Prospective Investigation into Cancer and Nutrition (EPIC). Ann Oncol. 2013;24:2166–73.CrossRefPubMedPubMedCentral Fedirko V, Trichopolou A, Bamia C, Duarte-Salles T, Trepo E, Aleksandrova K, et al. Consumption of fish and meats and risk of hepatocellular carcinoma: the European Prospective Investigation into Cancer and Nutrition (EPIC). Ann Oncol. 2013;24:2166–73.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Beckonert O, Keun HC, Ebbels TMD, Bundy JG, Holmes E, Lindon JC, et al. Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts. Nat Protoc. 2007;2:2692–703.CrossRefPubMed Beckonert O, Keun HC, Ebbels TMD, Bundy JG, Holmes E, Lindon JC, et al. Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts. Nat Protoc. 2007;2:2692–703.CrossRefPubMed
39.
Zurück zum Zitat Wishart DS, Knox C, Guo AC, Eisner R, Young N, Gautam B, et al. HMDB: a knowledgebase for the human metabolome. Nucleic Acids Res. 2009;37:D603–10.CrossRefPubMed Wishart DS, Knox C, Guo AC, Eisner R, Young N, Gautam B, et al. HMDB: a knowledgebase for the human metabolome. Nucleic Acids Res. 2009;37:D603–10.CrossRefPubMed
40.
Zurück zum Zitat Cui Q, Lewis IA, Hegeman AD, Anderson ME, Li J, Schulte CF, et al. Metabolite identification via the Madison Metabolomics Consortium Database. Nat Biotechnol. 2008;26:162–4.CrossRefPubMed Cui Q, Lewis IA, Hegeman AD, Anderson ME, Li J, Schulte CF, et al. Metabolite identification via the Madison Metabolomics Consortium Database. Nat Biotechnol. 2008;26:162–4.CrossRefPubMed
41.
Zurück zum Zitat Trygg J, Wold S. Orthogonal projections to latent structures (O-PLS). J Chemometrics. 2002;16:119–28.CrossRef Trygg J, Wold S. Orthogonal projections to latent structures (O-PLS). J Chemometrics. 2002;16:119–28.CrossRef
42.
Zurück zum Zitat Good PI. Permutation tests: a practical guide to resampling methods for testing hypotheses. New York: Spinger-Verlag; 2000.CrossRef Good PI. Permutation tests: a practical guide to resampling methods for testing hypotheses. New York: Spinger-Verlag; 2000.CrossRef
43.
Zurück zum Zitat Blaise BJ, Shintu L, Elena B, Emsley L, Dumas ME, Toulhoat P. Statistical recoupling prior to significance testing in nuclear magnetic resonance based metabonomics. Anal Chem. 2009;81:6242–51.CrossRefPubMed Blaise BJ, Shintu L, Elena B, Emsley L, Dumas ME, Toulhoat P. Statistical recoupling prior to significance testing in nuclear magnetic resonance based metabonomics. Anal Chem. 2009;81:6242–51.CrossRefPubMed
44.
Zurück zum Zitat Benjamini Y, Hochberg Y. Controlling the false discovery rate – a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300. Benjamini Y, Hochberg Y. Controlling the false discovery rate – a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300.
45.
Zurück zum Zitat Fages A, Ferrari P, Monni S, Dossus L, Floegel A, Mode N, et al. Investigating sources of variability in metabolomic data in the EPIC study: the principal component partial R-square (PC-PR2) method. Metabolomics. 2014;10:1074–83.CrossRef Fages A, Ferrari P, Monni S, Dossus L, Floegel A, Mode N, et al. Investigating sources of variability in metabolomic data in the EPIC study: the principal component partial R-square (PC-PR2) method. Metabolomics. 2014;10:1074–83.CrossRef
47.
Zurück zum Zitat Ghoshal AK, Farber E. Choline deficiency, lipotrope deficiency and the development of liver disease including liver cancer: a new perspective. Lab Invest. 1993;68:255–60.PubMed Ghoshal AK, Farber E. Choline deficiency, lipotrope deficiency and the development of liver disease including liver cancer: a new perspective. Lab Invest. 1993;68:255–60.PubMed
48.
Zurück zum Zitat Haussinger D. Regulation of hepatic ammonia metabolism: the intercellular glutamine cycle. Adv Enzyme Regul. 1986;25:159–80.CrossRefPubMed Haussinger D. Regulation of hepatic ammonia metabolism: the intercellular glutamine cycle. Adv Enzyme Regul. 1986;25:159–80.CrossRefPubMed
49.
Zurück zum Zitat Pogribny IP, James SJ, Beland FA. Molecular alterations in hepatocarcinogenesis induced by dietary methyl deficiency. Mol Nutr Food Res. 2012;56:116–25.CrossRefPubMed Pogribny IP, James SJ, Beland FA. Molecular alterations in hepatocarcinogenesis induced by dietary methyl deficiency. Mol Nutr Food Res. 2012;56:116–25.CrossRefPubMed
50.
Zurück zum Zitat Watford M. Glutamine and glutamate metabolism across the liver sinusoid. J Nutr. 2000;130:983S–7.PubMed Watford M. Glutamine and glutamate metabolism across the liver sinusoid. J Nutr. 2000;130:983S–7.PubMed
52.
Zurück zum Zitat Zar T, Graeber C, Perazella MA. Recognition, treatment, and prevention of propylene glycol toxicity. Semin Dial. 2007;20:217–9.CrossRefPubMed Zar T, Graeber C, Perazella MA. Recognition, treatment, and prevention of propylene glycol toxicity. Semin Dial. 2007;20:217–9.CrossRefPubMed
53.
Zurück zum Zitat Beyofülu D, Idle JR. The metabolomic window into hepatobiliary disease. J Hepatol. 2013;59:842–58.CrossRef Beyofülu D, Idle JR. The metabolomic window into hepatobiliary disease. J Hepatol. 2013;59:842–58.CrossRef
54.
Zurück zum Zitat Barr J, Caballeria J, Martinez-Arranz I, Dominguez-Diez A, Alonso C, Muntane J, et al. Obesity-dependent metabolic signatures associated with nonalcoholic fatty liver disease progression. J Proteome Res. 2012;11:2521–32.CrossRefPubMedPubMedCentral Barr J, Caballeria J, Martinez-Arranz I, Dominguez-Diez A, Alonso C, Muntane J, et al. Obesity-dependent metabolic signatures associated with nonalcoholic fatty liver disease progression. J Proteome Res. 2012;11:2521–32.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Barr J, Vazquez-Chantada M, Alonso C, Perez-Cormenzana M, Mayo R, Galan A, et al. Liquid chromatography-mass spectrometry-based parallel metabolic profiling of human and mouse model serum reveals putative biomarkers associated with the progression of nonalcoholic fatty liver disease. J Proteome Res. 2010;9:4501–12.CrossRefPubMedPubMedCentral Barr J, Vazquez-Chantada M, Alonso C, Perez-Cormenzana M, Mayo R, Galan A, et al. Liquid chromatography-mass spectrometry-based parallel metabolic profiling of human and mouse model serum reveals putative biomarkers associated with the progression of nonalcoholic fatty liver disease. J Proteome Res. 2010;9:4501–12.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Tanaka N, Matsubara T, Krausz KW, Patterson AD, Gonzalez FJ. Disruption of phospholipid and bile acid homeostasis in mice with nonalcoholic steatohepatitis. Hepatology. 2012;56:118–29.CrossRefPubMed Tanaka N, Matsubara T, Krausz KW, Patterson AD, Gonzalez FJ. Disruption of phospholipid and bile acid homeostasis in mice with nonalcoholic steatohepatitis. Hepatology. 2012;56:118–29.CrossRefPubMed
57.
Zurück zum Zitat Qi SW, Tu ZG, Peng WJ, Wang LX, Ou-Yang X, Cai AJ, et al. (1)H NMR-based serum metabolic profiling in compensated and decompensated cirrhosis. World J Gastroenterol. 2012;18:285–90.CrossRefPubMedPubMedCentral Qi SW, Tu ZG, Peng WJ, Wang LX, Ou-Yang X, Cai AJ, et al. (1)H NMR-based serum metabolic profiling in compensated and decompensated cirrhosis. World J Gastroenterol. 2012;18:285–90.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Lin X, Zhang Y, Ye G, Li X, Yin P, Ruan Q, et al. Classification and differential metabolite discovery of liver diseases based on plasma metabolic profiling and support vector machines. J Sep Sci. 2011;34:3029–36.CrossRefPubMed Lin X, Zhang Y, Ye G, Li X, Yin P, Ruan Q, et al. Classification and differential metabolite discovery of liver diseases based on plasma metabolic profiling and support vector machines. J Sep Sci. 2011;34:3029–36.CrossRefPubMed
Metadaten
Titel
Metabolomic profiles of hepatocellular carcinoma in a European prospective cohort
verfasst von
Anne Fages
Talita Duarte-Salles
Magdalena Stepien
Pietro Ferrari
Veronika Fedirko
Clément Pontoizeau
Antonia Trichopoulou
Krasimira Aleksandrova
Anne Tjønneland
Anja Olsen
Françoise Clavel-Chapelon
Marie-Christine Boutron-Ruault
Gianluca Severi
Rudolf Kaaks
Tilman Kuhn
Anna Floegel
Heiner Boeing
Pagona Lagiou
Christina Bamia
Dimitrios Trichopoulos
Domenico Palli
Valeria Pala
Salvatore Panico
Rosario Tumino
Paolo Vineis
H. Bas Bueno-de-Mesquita
Petra H. Peeters
Elisabete Weiderpass
Antonio Agudo
Esther Molina-Montes
José María Huerta
Eva Ardanaz
Miren Dorronsoro
Klas Sjöberg
Bodil Ohlsson
Kay-Tee Khaw
Nick Wareham
Ruth C. Travis
Julie A. Schmidt
Amanda Cross
Marc Gunter
Elio Riboli
Augustin Scalbert
Isabelle Romieu
Benedicte Elena-Herrmann
Mazda Jenab
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2015
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-015-0462-9

Weitere Artikel der Ausgabe 1/2015

BMC Medicine 1/2015 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.