Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Research

Identification of characteristic metabolic panels for different stages of prostate cancer by 1H NMR-based metabolomics analysis

verfasst von: Xi Zhang, Binbin Xia, Hong Zheng, Jie Ning, Yinjie Zhu, Xiaoguang Shao, Binrui Liu, Baijun Dong, Hongchang Gao

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

Abstract

Background

Prostate cancer (PCa) is the second most prevalent cancer in males worldwide, yet detecting PCa and its metastases remains a major challenging task in clinical research setups. The present study aimed to characterize the metabolic changes underlying the PCa progression and investigate the efficacy of related metabolic panels for an accurate PCa assessment.

Methods

In the present study, 75 PCa subjects, 62 PCa patients with bone metastasis (PCaB), and 50 benign prostatic hyperplasia (BPH) patients were enrolled, and we performed a cross-sectional metabolomics analysis of serum samples collected from these subjects using a 1H nuclear magnetic resonance (NMR)-based metabolomics approach.

Results

Multivariate analysis revealed that BPH, PCa, and PCaB groups showed distinct metabolic divisions, while univariate statistics integrated with variable importance in the projection (VIP) scores identified a differential metabolite series, which included energy, amino acid, and ketone body metabolism. Herein, we identified a series of characteristic serum metabolic changes, including decreased trends of 3-HB and acetone as well as elevated trends of alanine in PCa patients compared with BPH subjects, while increased levels of 3-HB and acetone as well as decreased levels of alanine in PCaB patients compared with PCa. Additionally, our results also revealed the metabolic panels of discriminant metabolites coupled with the clinical parameters (age and body mass index) for discrimination between PCa and BPH, PCaB and BPH, PCaB and PCa achieved the AUC values of 0.828, 0.917, and 0.872, respectively.

Conclusions

Overall, our study gave successful discrimination of BPH, PCa and PCaB, and we characterized the potential metabolic alterations involved in the PCa progression and its metastases, including 3-HB, acetone and alanine. The defined biomarker panels could be employed to aid in the diagnosis and classification of PCa in clinical practice.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-022-03478-5.
Xi Zhang, Binbin Xia and Hong Zheng contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
PCa
Prostate cancer
PCaB
PCa patients with bone metastasis
BPH
Benign prostatic hyperplasia
NMR
Nuclear magnetic resonance
BMI
Body mass index
PSA
Prostate-specific antigen
DRE
Digital rectal examination
TRUS
Trans-rectal ultrasound
3-HB
3-Hydroxybutyrate

Background

Prostate cancer (PCa) is the second most frequent cancer among men globally, which poses a major detriment to men’s health [1]. There were an estimated 1.4 million new cases of prostate cancer, and almost 375,304 cancer deaths occurred in 2020 [2]. Several significant efforts have been focused on the development of sensitive and accurate diagnostic tools for PCa since it is more likely to be cured if it is diagnosed early [3]. Currently, PCa is majorly screened by the prostate-specific antigen (PSA) blood test or a digital rectal examination (DRE) combined with a subsequent ultrasound-guided prostate biopsy (PBs) that confirms the cancerous growth presence [4]. Several limitations still exist in the traditional diagnostic methods like, higher procedural costs, longer examination time, low sensitivity and specificity leading to false positive findings, which sometimes may lead to overdiagnosis and consequent overtreatment [5, 6]. Therefore, there is an urgent need to discover novel biomarkers for improving the diagnosis, prognosis and treatment of PCa.
As dysregulated cellular metabolism is a vital hallmark of cancer [7], employing metabolomics analysis in cancer samples could provide critical insights for monitoring the cancer progression [8, 9]. Recently, multiple studies have demonstrated the potential of metabolomics analysis in PCa research. For example, Lima et al. [10] analyzed metabolomic profiling of volatile metabolites in urine samples from PCa patients and healthy controls using a metabolomics approach and found that urinary signatures of volatile organic compounds and volatile carbonyl compounds allowed for accurate discrimination between PCa and control groups. Based on the liquid chromatography-mass spectrometry (LC–MS) and chromatography-mass spectrometry (GC–MS), Huang et al. [11] suggested that serum N-oleoyl taurine and sterol metabolites levels were linked to a decreased PCa survival rate. Additionally, a serum metabolomics analysis integrated with GC–MS and magnetic resonance spectroscopy (MRS) separated benign prostatic hyperplasia (BPH) from PCa subjects, two common conditions that gave rise to increased PSA levels, and identified acylcarnitines, glycerophospholipids, and arginine that could be used as potential diagnostic markers for separating BPH from PCa [12]. Hence, this concept can be put forth that metabolomics analysis has a strong potential as a precise diagnostic tool for exploring metabolic biomarker’s expression patterns and elucidating the potential PCa metabolic mechanisms [13].
Nuclear magnetic resonance (NMR) spectroscopy is a promising noninvasive technique for metabolic profiling analysis due to innate advantages like simple sample preparation, non-destructive analysis, and high reproducibility [14]. In our previous study, 1H-NMR based metabolomics approach was successfully applied to characterize the significant metabolic differences in tissue and biofluid samples in five different PCa stages [15] as well as prostate tissue samples in hormone-sensitive and castration-resistant PCa cases [16]. In the previous study, we identified a series of metabolic disturbances, including energy, amino acid, choline, fatty acid, and uridine metabolism in five different PCa stages [15], and amino acid, choline metabolism, and Warburg effect from hormone-sensitive to castration-resistant PCa [16]. However, little information is available about tumor progression and metastasis. The present study characterized the metabolic profiling in serum samples from PCa, PCa patients with bone metastasis (PCaB), and BPH subjects via a 1H NMR-based metabolomics approach. The aim of this study was (1) to identify metabolic alterations among PCa, PCaB, and BPH, (2) to investigate key metabolic pathways involved in the PCa and PCaB progression, (3) to elucidate the potential biomarker panels for differentiating among BPH, PCa and PCaB subjects.

Methods, materials and participants

Study Participants

This is a retrospective cross-sectional analysis of 187 participants included in Renji Hospital of Shanghai Jiaotong University from April 2014 to October 2018, including 75 BPH, 62 PCa, and 50 PCaB patients. All participants underwent PSA screening, DRE, trans-rectal ultrasound (TRUS) and sometimes biopsy according to the PCa National Comprehensive Cancer Network (NCCN) guidelines [17]. All subjects were enrolled based on clinical criteria for different diagnoses of PCa and BPH by an urologist. The PSA values of all subjects were higher than 4 ng/mL. PCa patients were defined by the following criteria: (1) nodules at DRE; (2) hyperechoic or hypoechoic lesion at TRUS; (3) biopsy-proven prostate cancer (Gleason score for the biopsy > 6). Additionally, PCaB patients were defined with distant bone metastases by X-rays and bone scan analyses. BPH patients were diagnosed after a negative DRE, TRUS or biopsy evaluation to rule out PCa. The study protocol was approved by the Ethics Committee of Shanghai Jiao Tong University School of Medicine (IRB number: Renji [2013] 26) along with the informed written consents obtained from all the participants. The detailed pathological and clinical subject parameters are listed in Table 1.
Table 1
Participants’ pathological and clinical characteristics
Parameters
BPHb
PCac
PCaBd
p valuea
PCa
vs
BPH
PCaB
vs
BPH
PCaB
vs
PCa
N
75
62
50
e
Age (year)
65.15 ± 7.46
68.53 ± 7.58
73.82 ± 8.17
0.009
 < 0.001
 < 0.001
BMI (kg/m2)
24.10 ± 2.56
24.62 ± 2.56
23.38 ± 2.90
0.249
0.129
0.014
PSA (ng/mL)
12.64 ± 5.27
18.51 ± 15.92
401.16 ± 124.46
0.058
0.024
0.027
Gleason score
3 + 3
17
3
   
 
3 + 4
11
8
   
 
3 + 5
0
2
   
 
4 + 3
22
10
   
 
4 + 4
8
18
   
 
4 + 5
2
6
   
 
5 + 3
1
0
   
 
5 + 4
1
2
   
 
5 + 5
0
1
   
Data were presented as Mean ± SD
aThe p values were calculated by using unpaired Student’s t-test with Bonferroni adjustment, the difference was considered to be statistically significant when p < 0.05
bBPH, patient with benign prostatic hyperplasia
cPCa, prostate cancer
dPCaB, prostate cancer with bone metastasis
eNo data.

Sample collection and preparation

Once accepted and enrolled into our study, body mass index (BMI) of subjects were recorded and blood samples were collected. All the participants were overnight-fasted and the fasting blood samples were collected on the next morning following a standard operating procedure. Then, the blood samples were thawed for 15 min under ambient temperature, and centrifuged for 10 min at 1024 g and 4 °C to obtain serum samples. Serum supernatants were collected and stored at –80 °C for further analysis. Before undertaking the metabolomic analysis, serum samples were thawed at 4 °C and vortexed for ten seconds, followed by the dilution of the serum sample (200 µL) with phosphate buffer (250 µL, 0.2 mM Na2HPO4/NaH2PO4, pH = 7.4) as well as deuterium oxide (D2O, 50 µL). Thereafter, the mixed serum sample was vortexed for ten seconds and centrifuged at 10,000 g for 15 min at 4 °C followed by the supernatant transfer (500 µL) into a 5-mm NMR tube for further metabolomics analysis.

1H NMR-based metabolomics analysis

1H NMR-based metabolomics analysis was performed on a Bruker AVANCE III 600 MHz NMR spectrometer (Bruker BioSpin, Rheinstetten, Germany) equipped with a five mm Triple Resonance Probe (TXI) probe to obtain the information about small molecule metabolites in serum samples. The 1H NMR spectra of serum samples were acquired by using a Carr-Purcell-Meiboom-Gill (CPMG) pulse sequence with a fixed receiver-gain value at 37 °C for negating the comprehensive effects of lipid or protein macromolecules. The main NMR acquisition parameters, as considered in our previous study [18], were determined as follows: data points = 256 K; relaxation delay = 4 s; spectral width = 12,335.5 Hz; acquisition time = 2.66 s per scan. All CPMG spectra were transferred to TopSpin 3.0 software (Bruker BioSpin, Rheinstetten, Germany) for automatic phase and baseline corrections while the chemical shifts of the serum spectra were directed toward the anomeric signal of α-glucose at 5.23 ppm. The icoshift algorithm based on spectral interval’s calibrations was applied to align NMR spectra in MATLAB (R2012a, The Mathworks Inc., Natick, MA, USA) [19]. Henceforth, the spectral region from 0.0 to 9.0 ppm, excluding the water peak from 4.70 to 5.00 ppm, was subdivided and integrated into binned datasets with sizes of 0.01 ppm and 0.0015 ppm for multivariable and quantitative analyses, respectively. Accordingly, the metabolites were assigned by using Chenomx NMR suite 7.7.2 software (Chenomx Inc., Alberta, Canada) and the Human Metabolome Database (HMDB) [20]. The concentrations of serum metabolites were calculated according to their peak area by reference to the total area of each NMR spectrum and expressed as relative units (r.u.).

Multivariate analysis

This study conducted multivariate data analysis by using SIMCA 12.0 software (Umetrics AB, Umea, Sweden) that utilized partial least squares discriminant analysis (PLS-DA) to obtain a metabolic pattern discrimination overview among BPH, PCa and PCaB subjects. Furthermore, the orthogonal partial least squares discriminant analysis (OPLS-DA) model, another supervised method, was used to construct a group-based model for optimizing classification by removing unrelated variables of the class. The important metabolites for classification were identified by corresponding variable importance in the projection (VIP) scores, whereas metabolites with a VIP value > 1 were selected for further analysis. The OPLS-DA model’s quality was evaluated by cross-validated analysis of variance (CV-ANOVA), in which higher values of R2 and Q2 were considered as a goodness of fit and goodness of prediction parameters, respectively. The value of R2 and Q2 close to 1.0 indicates a good performance of OPLS-DA model. The principal component analysis (PCA) model was utilized to examine metabolic changes among different regions of age and BMI for detecting the influence of potential covariates on serum metabolome.

Statistical analysis

Statistical analyses were performed using SPSS 22.0 software (IBM Corp, Armonk, NY). The unpaired Student’s t-test with Bonferroni adjustment was applied to measure the significance of each metabolite, age, BMI and PSA levels. The p values across all metabolites within each comparison were also adjusted to account for multiple testing by a false discovery rate (FDR) method. A FDR value of < 0.05 was considered statistically significant. Our study selected metabolites with VIP > 1 and FDR value < 0.05 and considered them as the significant differential metabolites that could discriminate within the groups for an accurate disease assessment. Metabolic pathways were manually drawn by CorelDRAW Graphics Suite (Corel Inc., Ottawa, Canada) based on KEGG pathways (www.​genome.​jp/​kegg/​) and Small Molecule Pathway Database (SMPDB, www.​smpdb.​ca/​).

Receiver-operating characteristic (ROC) analysis

Receiver operating characteristic (ROC) curves analysis was used to evaluate the diagnostic potential of variable-identified metabolites among BPH, PCa, and PCaB groups along with the generation of the area under the ROC curve (AUC) value by MedCalc software (MedCalc Software, Ostend, Belgium). The significance level was defined at a p value < 0.05, and the AUC value < 1 indicated excellent diagnostic accuracy.

Results

Participants’ clinical characteristics

A total of 187 subjects, including 75 BPH patients, 62 PCa patients, and 50 PCaB patients, were included, and their clinical characteristics are presented in Table 1. In the cohort, the mean age of patients was significantly greater in PCaB, followed by PCa and BPH subjects. Although BMI of PCaB patients was distinctly decreased compared to PCa, no significant alteration was observed in PCa and PCaB patients compared to BPH subjects. Furthermore, PCaB patients had distinct higher PSA levels than PCa and BPH groups; however, no difference was observed in PSA levels between BPH and PCa cases. Hence, the traditional detection based on PSA levels is inapplicable for separating BPH from PCa.

Metabolic segregations among BPH, PCa, and PCaB patients

This study comprehensively analyzed the metabolic profiles of serum samples obtained from 75 BPH, 62 PCa, and 50 PCaB patients by a 1H-NMR based metabolomics approach. Figure 1A illustrates a typical 1H-NMR spectrum of BPH serum sample containing 25 metabolites that were mainly involved in energy metabolism (citrate, creatine, creatinine, α-glucose, β-glucose, lactate, and pyruvate), amino acid metabolism (alanine, glutamine, glycine, histidine, isoleucine, leucine, lysine, phenylalanine, threonine, tyrosine, and valine), ketone body metabolism (3-hydroxybutyrate (3-HB), acetone and acetoacetate), lipid metabolism (LDL/VLDL and glycerol) and fatty acid metabolism (acetate and formate).
The PLS-DA, based on the serum metabolome characteristics, was carried out for determining the metabolic pattern changes among BPH, PCa, and PCaB groups. As illustrated in Fig. 1B, the PLS-DA score plot revealed a clear metabolic pattern demarcation between BPH, PCa, and PCaB subjects. Subsequently, a supervised OPLS-DA model was utilized for characterizing the discrimination between the two groups as well as the VIP score, which confirmed the differential metabolites. As can be seen from Fig. 1C–E, the discriminant capabilities of the PLS-DA models were observed in the following groups: between BPH and PCa (R2 = 0.574, Q2 = 0.47) (Fig. 1C), between BPH and PCaB (R2 = 0.585, Q2 = 0.522) (Fig. 1D) as well as between PCa and PCaB (R2 = 0.594, Q2 = 0.475) (Fig. 1E). The BPH and PCa segregation as displayed by the VIP score of OPLS-DA was mainly attributed to metabolites like 3-HB, alanine, acetone, glutamine, tyrosine, histidine, and formate (Fig. 1C). In contrast, significant metabolites like leucine, isoleucine, valine, acetoacetate, pyruvate, creatine, phenylalanine, histidine, and formate played a remarkable role in metabolic differentiation between BPH and PCaB patients (Fig. 1D). Additionally, LDL/VLDL, leucine, isoleucine, valine, 3-HB, alanine, acetone, acetoacetate, pyruvate, citrate, and creatine were also associated with PCa and PCaB dissociation in the serum metabolome (Fig. 1E).
Given that age and BMI were significantly different among BPH, PCa, and PCaB groups, we utilized a principal component analysis (PCA) model to examine the metabolic changes among different regions of age and BMI for detecting the influence of potential covariates on serum metabolome. As shown in Additional file 1: Fig. S1, there is no distinct separation of serum metabolome detected among different regions of age (see Additional file 1: Fig. S1A) and BMI (see Additional file 1: Fig. S1B), indicating that serum metabolome of BPH, PCa and PCaB pateints had a graded relationship with disease status that was not caused by age, BMI. Hence, we set aside the effect of different age and BMI on serum metabolome in the subsequent analysis.

Quantification of differential metabolites among BPH, PCa, and PCaB

Statistical analysis of detected metabolites was obtained for exploring the characteristic metabolic changes among BPH, PCa, and PCaB groups in Table 2. When compared with BPH, PCa had significantly lower levels of 3-HB, acetone, glutamine and histidine as well as higher serum levels of alanine and tyrosine, whereas PCaB had significantly increased levels of formate, phenylalanine and pyruvate along with a remarkable decrease in acetoacetate, creatine, glutamine, histidine, isoleucine, leucine and valine when compared to BPH cases. Furthermore, in comparison with PCa, PCaB subjects had higher levels of 3-HB, acetone, citrate, phenylalanine and pyruvate in serum metabolome coupled with lower alanine, creatine, isoleucine, LDL/VLDL, leucine and valine levels. Overall, these results suggested that metabolic changes during the progression and metastasis of PCa are quite different.
Table 2
Metabolic changes in serum samples from BPH, PCa, and PCaB
Metabolites
(r.u.)
BPHc
PCad
PCaBe
PCa vs BPH
PCaB vs BPH
PCaB vs PCa
p valuea
FDRb
p value
FDR
p value
FDR
3-HBf
9.31 ± 5.69
6.18 ± 3.51
8.86 ± 7.11
0.000
0.002
0.701
0.818
0.013
0.022
Acetate
2.11 ± 0.61
1.99 ± 0.63
1.92 ± 0.99
0.250
0.397
0.219
0.348
0.682
0.853
Acetoacetate
2.36 ± 1.19
2.14 ± 0.89
1.94 ± 0.92
0.210
0.397
0.025
0.036
0.245
0.452
Acetone
5.77 ± 4.36
3.21 ± 2.24
5.21 ± 5.21
0.000
0.001
0.528
0.684
0.010
0.042
Alanine
16.80 ± 2.71
18.57 ± 2.52
17.37 ± 3.45
0.000
0.002
0.307
0.413
0.028
0.047
Creatine
2.62 ± 0.62
2.63 ± 0.55
2.36 ± 0.70
0.122
0.330
0.013
0.046
0.026
0.031
Creatinine
1.03 ± 0.32
1.02 ± 0.29
1.00 ± 0.35
0.970
0.970
0.608
0.761
0.632
0.819
Citrate
2.44 ± 0.56
2.34 ± 0.43
2.56 ± 0.61
0.232
0.397
0.137
0.266
0.020
0.031
Formate
0.09 ± 0.04
0.11 ± 0.04
0.12 ± 0.04
0.124
0.330
0.009
0.045
0.204
0.398
Glutamine
18.97 ± 3.20
17.84 ± 2.68
17.70 ± 4.18
0.026
0.047
0.012
0.047
0.829
0.893
Glycine
8.29 ± 1.87
8.01 ± 1.54
8.41 ± 2.48
0.346
0.526
0.771
0.856
0.315
0.550
Gycerol
9.60 ± 1.77
9.50 ± 2.58
10.18 ± 2.75
0.057
0.810
0.178
0.226
0.142
0.193
Histidine
1.20 ± 0.21
1.13 ± 0.18
1.09 ± 0.21
0.022
0.037
0.001
0.035
0.962
0.985
Isoleucine
2.73 ± 0.47
2.72 ± 0.39
2.55 ± 0.43
0.921
0.970
0.012
0.043
0.028
0.035
Lactate
100.63 ± 36.14
92.09 ± 29.81
90.63 ± 33.26
0.040
0.157
0.215
0.348
0.561
0.818
LDL/VLDL
56.61 ± 21.04
61.83 ± 20.69
52.34 ± 23.60
0.147
0.344
0.289
0.404
0.023
0.031
Leucine
13.53 ± 2.02
13.31 ± 1.64
12.63 ± 2.03
0.468
0.655
0.003
0.035
0.005
0.038
Lysine
2.46 ± 0.33
2.54 ± 0.46
2.37 ± 0.45
0.249
0.397
0.235
0.357
0.051
0.137
Phenylalanine
0.68 ± 0.26
0.72 ± 0.31
0.84 ± 0.35
0.078
0.249
0.002
0.028
0.026
0.049
Pyruvate
1.65 ± 0.44
1.65 ± 0.38
1.93 ± 0.56
0.969
0.970
0.002
0.028
0.002
0.034
Threonine
1.76 ± 0.31
1.73 ± 0.40
1.70 ± 0.33
0.623
0.838
0.254
0.370
0.613
0.819
Tyrosine
0.94 ± 0.19
1.02 ± 0.15
1.01 ± 0.33
0.029
0.041
0.180
0.315
0.833
0.893
Valine
9.37 ± 1.47
9.35 ± 1.32
8.65 ± 1.57
0.872
0.970
0.003
0.036
0.034
0.039
α-Glucose
14.19 ± 3.07
14.89 ± 2.89
15.31 ± 3.46
0.177
0.386
0.049
0.167
0.475
0.722
β-Glucose
12.81 ± 3.10
12.98 ± 3.29
12.97 ± 3.45
0.752
0.970
0.782
0.856
0.985
0.985
Data were presented as Mean ± SD
aAdjusted by Bonferroni correction across multiple comparisons within each metabolite
bAdjusted by false discovery rate (FDR) method across the metabolites within each comparison
cBPH, patient with benign prostatic hyperplasia
dPCa, prostate cancer
ePCaB, prostate cancer with bone metastasis
f3-HB, 3-hydroxybutyrate. The colored p value represents significantly decreased (green) or increased (red) from BPH to PCa, BPH to PCaB or PCa to PCaB

Diagnostic performance of potential metabolic biomarkers

ROC curves analysis, including the AUC value was conducted for evaluating the efficient utilization of the identified significant metabolites for BPH, PCa and PCaB patients screening, based on the metabolites with VIP value > 1 and FDR value < 0.05. Figure 2 illustrates the diagnostic performance of potential metabolic biomarker panels (AUC value > 0.6, p value < 0.05) as they account for discrimination between BPH and PCa groups due to the exhibition of good response by the five metabolite levels that included 3-HB (AUC = 0.678, sensitivity = 43.55%, specificity = 84%), alanine (AUC = 0.708, sensitivity = 58.06%, specificity = 81.33%), acetone (AUC = 0.687, sensitivity = 74.19%, specificity = 60%), tyrosine (AUC = 0.648, sensitivity = 87.10%, specificity = 38.67%) and histidine (AUC = 0.602, sensitivity = 70.97%, specificity = 46.67%). Moreover, the ROC analysis revealed that these five metabolites in combination had a 0.815 AUC value, along with 75.81% and 72% sensitivity and specificity, respectively. While an integration of these five metabolites with age and BMI might achieved 0.828 AUC value coupled with a sensitivity and specificity of 56.45% and 94.67%, respectively.
The ROC analysis results based on the BPH and PCaB differentiating values (AUC value > 0.6 and p value < 0.05) are shown in Fig. 3 which identified eight total of 8 metabolites, that included leucine (AUC = 0.607, sensitivity = 60%, specificity = 62.57%), isoleucine (AUC = 0.614, sensitivity = 54.55%, specificity = 68%), valine (AUC = 0.631, sensitivity = 61.82%, specificity = 61.33%), acetoacetate (AUC = 0.625, sensitivity = 38.18%, specificity = 85.33%), pyruvate (AUC = 0.644, sensitivity = 60%, specificity = 68%), phenylalanine (AUC = 0.675, sensitivity = 62.5%, specificity = 70.83%), histidine (AUC = 0.683, sensitivity = 72.73%, specificity = 58.67%) and formate (AUC = 0.651, sensitivity = 56.36%, specificity = 72%). Moreover, the combination of all these metabolites resulted in a 0.794 AUC value, and the sensitivity and specificity were 65.45% and 85.33%, respectively. Notably, the amalgamation of these eight metabolites with age and BMI was extremely capable of differentiating BPH from PCaB cases, with an AUC value of 0.917, 89.36% sensitivity and 90.67% specificity.
Similarly, the ROC analysis identified top eight metabolites for extricating PCaB diagnosis from PCa as shown in Fig. 4, while their combination exhibited a better classification (AUC = 0.828, sensitivity = 78.18%, specificity = 74.19%) than a singular metabolite: LDL/VLDL (AUC = 0.642, sensitivity = 54.55%, specificity = 74.19%), isoleucine (AUC = 0.622, sensitivity = 43.64%, specificity = 85.48%), valine (AUC = 0.626, sensitivity = 49.09%, specificity = 75.81%), 3-HB (AUC = 0.604, sensitivity = 80%, specificity = 41.94%), alanine (AUC = 0.614, sensitivity = 56.36%, specificity = 70.97%), pyruvate (AUC = 0.659, sensitivity = 89.09%, specificity = 38.71%), citrate (AUC = 0.613, sensitivity = 32.73%, specificity = 98.39%) and creatine (AUC = 0.622, sensitivity = 34.55%, specificity = 88.71%). Additionally, the merging of eight metabolites with age and BMI displayed the best predictability with an AUC value, sensitivity and specificity of 0.872, 87.23% and 74.19%, respectively.
In conclusion, metabolic panels of discriminant metabolites and the amalgamation of metabolites and clinical parameters displayed good diagnostic capability for differentiating among BPH, PCa and PCaB subjects.

Metabolic pathway analysis of differential metabolites among BPH, PCa, and PCaB

Metabolic pathway analysis was carried out based on the differential metabolites for exploring pathway-based metabolic features, as shown in Fig. 5. Our results suggested that a series of metabolic pathways like energy, amino acid, and ketone body metabolism were implicated in metabolic discrimination among BPH, PCa, and PCaB subjects.

Discussion

Early diagnosis plays a crucial role in the successful treatment for PCa [21]; however, the non-selective use of traditional screening tools for PCa generally leads to overdiagnosis and overtreatment that does not give expected results [22]. Several studies have reported that metabolic disturbances have been associated with increased PCa incidence [23]. Nowadays, metabolomics has emerged as an immensely powerful screening tool in PCa biomarker development due to its inherent advantages, such as noninvasive procedures, high reproducibility, and lower costs [24]. This study examined the metabolic profiles of BPH, PCa, and PCaB serum samples obtained by 1H NMR-based metabolomics approach and revealed that serum metabolome analysis exhibited clear discriminations among the three subject groups: BPH, PCa, and PCaB. After conducting univariate statistical and VIP analyses, the most important metabolites were selected, and the diagnostic capacity of these metabolites was evaluated by ROC analysis, while the potential biomarker panels were identified in serum samples for segregating BPH, PCa, and PCaB subjects. Accordingly, the present study identified valuable metabolic panels based on serum metabolome, which exhibited superior performance in discriminating the three groups. A combination of variable metabolites from MRS and MS analysis [12] showed a good classification capacity for discriminating BPH from PCa, with 73.7% of sensitivity, 69% specificity, and 54.1% and 76.3% sensitivity and specificity, respectively. In the present study, significant metabolites in combination for discriminating BPH from PCa patients, had a 0.815 AUC value, along with 75.81% and 72% sensitivity and specificity, respectively. According to Zhang et al. [25], the prognostic value of serum sialic acid levels in predicting PCa and PCaB achieved an AUC value of 0.57, the sensitivity of 60% and the specificity of 58.6%. By combining variable metabolites found in the present study, PCa and PCaB patients were separated with a AUC value of 0.828, a sensitivity and specificity of 78.18% and 74.19%, respectively. Especially, the metabolic biomarker panels in the cluster of differential metabolites and clinical characteristics, showing the AUC values > 0.8, suggesting a relatively good clinical value for diagnosing PCa and its metastases. Based on these, our study imply that serum metabolome based on 1H NMR-based metabolomics approach has a great potential as a diagnostic supplemental tool for PCa and its metastases. In our study, it was observed that among all identified potential metabolic biomarkers for different PCa stages, a series of metabolic pathways were mainly involved, including energy, amino acid, and ketone body metabolism. Of note, a series of characteristic metabolic changes were identified, including decreased trends of 3-HB and acetone as well as elevated trends of alanine in PCa patients compared with BPH subjects, while increased levels of 3-HB and acetone as well as decreased levels of alanine in PCaB patients compared with PCa.
Energy metabolism, which expedites the uptake and incorporation of glucose into the biomass needed to produce new cells, is critical for maintaining the abnormal growth and intensive proliferation of cancer cells [26]. Furthermore, the glycolytic breakdown of glucose, the main substrate for energy supply, results in pyruvate production that consequently gets oxidized through the tricarboxylic acid (TCA) cycle for ATP production or converted into lactate by anaerobic glycolysis [27]. According to the Warburg effect [28], most cancer cells primarily convert glucose to lactate through anaerobic glycolysis for meeting the energy requirements for cellular cell proliferation. However, our study revealed that the pyruvate concentration in serum samples was significantly enhanced in PCaB cases compared to PCa and BPH, which might further imply that PCaB may have a reverse Warburg effect in serum metabolome that still needs validation. Citrate is not only an important substrate for de novo lipid synthesis but also serves as a key TCA cycle intermediate for ATP production [29, 30]. In the present study, the citrate level of PCaB serum samples was higher than the PCa samples, which is in line with our previous study [15], that increased TCA cycle intermediates might play a pivotal role in malignant tumor cell growth and proliferation. Additionally, the concentration of creatine, an important regulator of energy metabolism, was remarkably decreased in the PCaB serum levels when compared to PCa and BPH subjects. Given high energy demand of tumor cells [31], a decrease in serum creatine may attributed to providing fuels to meet energy production. Overall, our results implies that a distinct energy metabolism occurred in tumor progression and metastasis.
Out of the many energy sources, amino acids are precisely involved in biosynthesis as well as are important reserves for supporting the survival and proliferation of cancer cells [32]. A recent study investigated the emerging roles of amino acids in epigenetic regulation and initiating immune responses that were related to tumorigenesis and metastatic pathways [33]. Our study noted that a majority of amino acids were increased in the serum samples from BPH to PCa subjects but gradually decreased when the disease progressed to PCaB. It was suggested that leucine, isoleucine, and valine are members of the branched-chain amino acids (BCAAs) that can be catabolized to TCA cycle intermediates for energy production [34], which was also supported by Teahan et al. [35] that BCAAs are potential biomarkers for PCa aggression by using NMR-based metabolomics. Hence, it was evident that increased levels of serum BCAAs are utilized as an energy source for tumor proliferation and development. In contrast, it was also observed that the serum BCAAs levels were significantly decreasing in PCaB when compared with PCa or BPH patients, thereby suggesting that downregulated BCAAs might be closely related to bone metastasis in PCa progression. Recent evidence indicated that histidine catabolism was associated with PCa progression [15] and was consistent with our findings that the histidine level was markedly decreased in the serum levels of both PCa and PCaB patients as compared to BPH patients. As proposed by Lapek et al. in 2015 [36] that histidine phosphorylation might be greatly associated with metastatic PCa. Hence, this concept could be put forth that the downregulation of PCa and PCaB histidine levels, when compared to BPH patients, might be attributed to the upregulated histidine phosphorylation in PCa and the resultant metastasis development. Several previous studies have indicated that alanine level was significantly higher in serum and biopsy tissues when progressing from BPH to PCa [37, 38]. In contrast, a decreased alanine level was associated with advanced cancer stage and poor cancer-specific survival [39]. Accordingly, our study denoted that due to alanine’s differential nature, it was observed that tumor proliferation was consistent with increased protein synthesis; as a result, the alanine serum level was distinctly elevated from BPH to PCa but decreased from PCa to PCaB subjects. Correspondingly, tyrosine and phenylalanine were two additional metabolites that exhibited increased trends in the serum of PCa and PCaB patients. Several studies have demonstrated that dysregulated tyrosine and phenylalanine metabolism is closely related to PCa progression [40, 41]. Another observation in our study was the presence of higher levels of tyrosine and phenylalanine in PCa and PCaB patients’ serum when compared with BPH patients, respectively. It was substantiated by Gomez-Cebrian et al. [42] that phenylalanine hydroxylase (PAH) is the enzyme that metabolizes excess phenylalanine into tyrosine; hence, it is also reported to have a direct association with protein acetylation and energy production [41, 43]. Moreover, our study revealed that a decreased PAH expression might be directly proportional to the enhanced levels of both phenylalanine and tyrosine in PCa and its metastatic progression.
Ketone bodies (3-HB, acetoacetate, and acetone) are alternative mitochondrial energy reserves that can be converted into acetyl-CoA and reutilized as energy substrates[44, 45]. Several studies have stated that ketone body metabolism is critical for tumor biomass expansion [46], as well as the fact that ketogenesis plays an important role in PCa progression [47]. A study by Martinez et al. [48] generated a series of cells and fibroblasts overexpressing the enzyme initiating ketone body production and suggested that the production and reutilization of ketone bodies promote tumor progression and metastasis. A prominent ketone body, 3-HB, has been proved to be specifically associated with metastatic prostate cancer [49]. The current study displayed significant reductions in 3-HB and acetone in PCa cases relative to BPH, but significant elevations were duly observed in PCaB subjects when compared to PCa cases. Collectively, the characteristic changes in 3-HB and acetone levels might be applicable for the potential detection of the PCa progressive stages. However, it was observed that the acetoacetate concentration was significantly decreased in PCaB as compared to BPH patients, which may be due to the uptake of ketone bodies from the tumor tissues in PCa proliferation and development.
Formate is a member of short-chain fatty acids and can be remodeled back for re-synthesizing serine via a one-carbon metabolism pathway [50]. Additionally, formate is an extremely vital component of one-carbon metabolism for tumor proliferation and growth [51, 52]. A study by Meiser et al. [53] revealed that mice with oxidative cancers have higher circulating formate levels than the healthy controls, thereby proposed that elevated formate overflow is a hallmark of oxidative cancers. Our study results also observed an increased formate level in the PCaB patients' serum when compared to BPH, which was also confirmed by our previous study that depicted a higher formate level in the metastatic PCa tissue when compared to other PCa stages [15]. Thus, our results might imply that upregulated formate metabolism mainly occurred in the tumor tissue and serum samples of the tumor in its metastatic stage.
LDL/VLDL is an essential member of lipoproteins, fundamental to the reverse cholesterol transport pathway and lipid homeostasis [54, 55]. Recent studies have demonstrated that lipoprotein might be considered as a risk factor for PCa [56, 57]. However, several literary insights display contradictory findings regarding the association between lipid metabolism and PCa progression and development. A study by Bull et al. [58] suggested there is a weaker association between higher lipoprotein levels and aggressive PCa risk, which was also substantiated by a meta-analysis of 14 large prospective studies that indicated an absence of association between the blood lipoprotein levels and overall PCa or high-grade PCa risk cases [59]. Our results revealed that PCaB cases had lower circulating LDL/VLDL levels than PCa, thus, suggesting that PCaB may have a disruptive lipid metabolism relative to PCa. Moreover, the lipid metabolism inconsistency along with several metabolic aberrations in PCa development could contribute to the initiation of several more studies focused on the extremely significant role of lipid metabolism in PCa progression.
In this study, BPH patients were enrolled as a control group, which is a typical clinical setting. The healthy control donors usually had lower PSA levels and may have a small chance of holding undiagnosed PCa [12]. However, BPH patients with significantly higher levels of PSA are very common among elderly and middle-aged men, which frequently leads to the lack of specificity in PSA measurements. Besides, understanding the metabolic profiles among BPH, PCa and PCaB offers vital information about metabolic biomarkers for PCa progression and its metastases since PSA measurements are not optimal. Moreover, the appropriate use of metabolic biomarkers can provide a more comprehensive evidence for the early screening and diagnosis to predict the corresponding PCa progression and make individualized treatment options. Thus, we believe this is a highly reasonable and relevant clinical setting for diagnosing and classifying of PCa. However, there are several potential limitations in this study: (1) As the majority of BPH patients did not undergo biopsy, there was no relationship analysis between differential metabolic changes and the subjects pathology in this study; but the pathologic analysis of subjects is recommended for elucidating the link of metabolic biomarkers in different PCa pathologies. (2) Although the potential metabolic biomarkers were identified in serum samples, this finding still needs validation by using other biofluids and matching tissues. (3) As our study was a prospective, single-center study with a smaller sample size, large clinical multi-center cohorts would be required for verifying the clinical potential of our findings. (4) Since 1H NMR-based metabolomics analysis is generally limited to small metabolites at higher concentrations, multi-omics analysis integrating several useful genes, proteins, and metabolome are recommended for elucidating potential mechanisms underlying the PCa progression and its metastases.

Conclusion

To conclude, 1H NMR-based metabolomics analysis of serum metabolic profiles in BPH, PCa, and PCaB patients can be successfully utilized to establish the significant metabolic signatures related to PCa progression and its metastases. The identified metabolic panels in our study might provide crucial insights into the diagnosis and classification of PCa.

Acknowledgements

We thank the Department of Urology at Renji Hospital for collecting plasma samples and all patients involved in this study.

Declarations

All procedures carried out in this study were in accordance with the ethical standards of the institutional and national responsible committee on human experimentation and the Helsinki Declaration of 1964 and its later amendments or equivalents. This study was approved by the Ethics Committee of Shanghai Jiao Tong University School of Medicine (IRB number: Renji [2013] 26). Informed consent was obtained from all individual patients included in the study.
Not applicable.

Competing interests

The authors have declared that no competing interest exists.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
2.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef
3.
Zurück zum Zitat Olleik G, Kassouf W, Aprikian A, Hu J, Vanhuyse M, Cury F, et al. Evaluation of new tests and interventions for prostate cancer management: a systematic review. J Natl Compr Canc Netw. 2018;16(11):1340–51.PubMedCrossRef Olleik G, Kassouf W, Aprikian A, Hu J, Vanhuyse M, Cury F, et al. Evaluation of new tests and interventions for prostate cancer management: a systematic review. J Natl Compr Canc Netw. 2018;16(11):1340–51.PubMedCrossRef
4.
Zurück zum Zitat McNevin CS, Baird AM, McDermott R, Finn SP. Diagnostic strategies for treatment selection in advanced prostate cancer. Diagnostics (Basel). 2021;11(2): e345.CrossRef McNevin CS, Baird AM, McDermott R, Finn SP. Diagnostic strategies for treatment selection in advanced prostate cancer. Diagnostics (Basel). 2021;11(2): e345.CrossRef
5.
Zurück zum Zitat Kearns JT, Holt SK, Wright JL, Lin DW, Lange PH, Gore JL. PSA screening, prostate biopsy, and treatment of prostate cancer in the years surrounding the USPSTF recommendation against prostate cancer screening. Cancer. 2018;124(13):2733–9.PubMedCrossRef Kearns JT, Holt SK, Wright JL, Lin DW, Lange PH, Gore JL. PSA screening, prostate biopsy, and treatment of prostate cancer in the years surrounding the USPSTF recommendation against prostate cancer screening. Cancer. 2018;124(13):2733–9.PubMedCrossRef
6.
Zurück zum Zitat Catalona WJ, Richie JP, Ahmann FR, Hudson MA, Scardino PT, Flanigan RC, et al. Comparison of digital rectal examination and serum prostate specific antigen in the early detection of prostate cancer: results of a multicenter clinical trial of 6630 men. J Urol. 2017;197(2S):S200–7.PubMedCrossRef Catalona WJ, Richie JP, Ahmann FR, Hudson MA, Scardino PT, Flanigan RC, et al. Comparison of digital rectal examination and serum prostate specific antigen in the early detection of prostate cancer: results of a multicenter clinical trial of 6630 men. J Urol. 2017;197(2S):S200–7.PubMedCrossRef
7.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
8.
9.
Zurück zum Zitat Liang L, Sun F, Wang H, Hu Z. Metabolomics, metabolic flux analysis and cancer pharmacology. Pharmacol Ther. 2021;224: e107827.CrossRef Liang L, Sun F, Wang H, Hu Z. Metabolomics, metabolic flux analysis and cancer pharmacology. Pharmacol Ther. 2021;224: e107827.CrossRef
10.
Zurück zum Zitat Lima AR, Pinto J, Azevedo AI, Barros-Silva D, Jeronimo C, Henrique R, et al. Identification of a biomarker panel for improvement of prostate cancer diagnosis by volatile metabolic profiling of urine. Br J Cancer. 2019;121(10):857–68.PubMedPubMedCentralCrossRef Lima AR, Pinto J, Azevedo AI, Barros-Silva D, Jeronimo C, Henrique R, et al. Identification of a biomarker panel for improvement of prostate cancer diagnosis by volatile metabolic profiling of urine. Br J Cancer. 2019;121(10):857–68.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Huang J, Weinstein SJ, Moore SC, Derkach A, Hua X, Mondul AM, et al. Pre-diagnostic serum metabolomic profiling of prostate cancer survival. J Gerontol A Biol Sci Med Sci. 2019;74(6):853–9.PubMedCrossRef Huang J, Weinstein SJ, Moore SC, Derkach A, Hua X, Mondul AM, et al. Pre-diagnostic serum metabolomic profiling of prostate cancer survival. J Gerontol A Biol Sci Med Sci. 2019;74(6):853–9.PubMedCrossRef
12.
Zurück zum Zitat Giskeodegard GF, Hansen AF, Bertilsson H, Gonzalez SV, Kristiansen KA, Bruheim P, et al. Metabolic markers in blood can separate prostate cancer from benign prostatic hyperplasia. Br J Cancer. 2015;113(12):1712–9.PubMedPubMedCentralCrossRef Giskeodegard GF, Hansen AF, Bertilsson H, Gonzalez SV, Kristiansen KA, Bruheim P, et al. Metabolic markers in blood can separate prostate cancer from benign prostatic hyperplasia. Br J Cancer. 2015;113(12):1712–9.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Gomez-Cebrian N, Rojas-Benedicto A, Albors-Vaquer A, Lopez-Guerrero JA, Pineda-Lucena A, Puchades-Carrasco L. Metabolomics contributions to the discovery of prostate cancer biomarkers. Metabolites. 2019;9(3): e48.PubMedCrossRef Gomez-Cebrian N, Rojas-Benedicto A, Albors-Vaquer A, Lopez-Guerrero JA, Pineda-Lucena A, Puchades-Carrasco L. Metabolomics contributions to the discovery of prostate cancer biomarkers. Metabolites. 2019;9(3): e48.PubMedCrossRef
14.
Zurück zum Zitat Markley JL, Bruschweiler R, Edison AS, Eghbalnia HR, Powers R, Raftery D, et al. The future of NMR-based metabolomics. Curr Opin Biotechnol. 2017;43:34–40.PubMedCrossRef Markley JL, Bruschweiler R, Edison AS, Eghbalnia HR, Powers R, Raftery D, et al. The future of NMR-based metabolomics. Curr Opin Biotechnol. 2017;43:34–40.PubMedCrossRef
15.
Zurück zum Zitat Zheng H, Dong B, Ning J, Shao X, Zhao L, Jiang Q, et al. NMR-based metabolomics analysis identifies discriminatory metabolic disturbances in tissue and biofluid samples for progressive prostate cancer. Clin Chim Acta. 2020;501:241–51.PubMedCrossRef Zheng H, Dong B, Ning J, Shao X, Zhao L, Jiang Q, et al. NMR-based metabolomics analysis identifies discriminatory metabolic disturbances in tissue and biofluid samples for progressive prostate cancer. Clin Chim Acta. 2020;501:241–51.PubMedCrossRef
16.
Zurück zum Zitat Zheng H, Zhu Y, Shao X, Cai A, Dong B, Xue W, et al. Distinct metabolic signatures of hormone-sensitive and castration-resistant prostate cancer revealed by a (1)H NMR-based metabolomics of biopsy tissue. J Proteome Res. 2020;19(9):3741–9.PubMedCrossRef Zheng H, Zhu Y, Shao X, Cai A, Dong B, Xue W, et al. Distinct metabolic signatures of hormone-sensitive and castration-resistant prostate cancer revealed by a (1)H NMR-based metabolomics of biopsy tissue. J Proteome Res. 2020;19(9):3741–9.PubMedCrossRef
17.
Zurück zum Zitat Carroll PH, Mohler JL. NCCN guidelines updates: prostate cancer and prostate cancer early detection. J Natl Compr Canc Netw. 2018;16(5S):620–3.PubMedCrossRef Carroll PH, Mohler JL. NCCN guidelines updates: prostate cancer and prostate cancer early detection. J Natl Compr Canc Netw. 2018;16(5S):620–3.PubMedCrossRef
18.
Zurück zum Zitat Zheng H, Cai A, Zhou Q, Xu P, Zhao L, Li C, et al. Optimal preprocessing of serum and urine metabolomic data fusion for staging prostate cancer through design of experiment. Anal Chim Acta. 2017;991:68–75.PubMedCrossRef Zheng H, Cai A, Zhou Q, Xu P, Zhao L, Li C, et al. Optimal preprocessing of serum and urine metabolomic data fusion for staging prostate cancer through design of experiment. Anal Chim Acta. 2017;991:68–75.PubMedCrossRef
19.
Zurück zum Zitat Savorani F, Tomasi G, Engelsen SB. icoshift: A versatile tool for the rapid alignment of 1D NMR spectra. J Magn Reson. 2010;202(2):190–202.PubMedCrossRef Savorani F, Tomasi G, Engelsen SB. icoshift: A versatile tool for the rapid alignment of 1D NMR spectra. J Magn Reson. 2010;202(2):190–202.PubMedCrossRef
20.
Zurück zum Zitat Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vazquez-Fresno R, et al. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res. 2018;46(D1):D608–17.PubMedCrossRef Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vazquez-Fresno R, et al. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res. 2018;46(D1):D608–17.PubMedCrossRef
21.
Zurück zum Zitat Johansson JE, Andren O, Andersson SO, Dickman PW, Holmberg L, Magnuson A, et al. Natural history of early, localized prostate cancer. JAMA. 2004;291(22):2713–9.PubMedCrossRef Johansson JE, Andren O, Andersson SO, Dickman PW, Holmberg L, Magnuson A, et al. Natural history of early, localized prostate cancer. JAMA. 2004;291(22):2713–9.PubMedCrossRef
22.
Zurück zum Zitat Welch HG, Albertsen PC. Prostate cancer diagnosis and treatment after the introduction of prostate-specific antigen screening: 1986–2005. J Natl Cancer Inst. 2009;101(19):1325–9.PubMedPubMedCentralCrossRef Welch HG, Albertsen PC. Prostate cancer diagnosis and treatment after the introduction of prostate-specific antigen screening: 1986–2005. J Natl Cancer Inst. 2009;101(19):1325–9.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Kelly RS, Vander Heiden MG, Giovannucci E, Mucci LA. Metabolomic biomarkers of prostate cancer: prediction, diagnosis, progression, prognosis, and recurrence. Cancer Epidemiol Biomarkers Prev. 2016;25(6):887–906.PubMedPubMedCentralCrossRef Kelly RS, Vander Heiden MG, Giovannucci E, Mucci LA. Metabolomic biomarkers of prostate cancer: prediction, diagnosis, progression, prognosis, and recurrence. Cancer Epidemiol Biomarkers Prev. 2016;25(6):887–906.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Kdadra M, Hockner S, Leung H, Kremer W, Schiffer E. Metabolomics biomarkers of prostate cancer: a systematic review. Diagnostics (Basel). 2019;9(1): e21.CrossRef Kdadra M, Hockner S, Leung H, Kremer W, Schiffer E. Metabolomics biomarkers of prostate cancer: a systematic review. Diagnostics (Basel). 2019;9(1): e21.CrossRef
25.
Zurück zum Zitat Zhang C, Yan L, Song H, Ma Z, Chen D, Yang F, et al. Elevated serum sialic acid levels predict prostate cancer as well as bone metastases. J Cancer. 2019;10(2):449–57.PubMedPubMedCentralCrossRef Zhang C, Yan L, Song H, Ma Z, Chen D, Yang F, et al. Elevated serum sialic acid levels predict prostate cancer as well as bone metastases. J Cancer. 2019;10(2):449–57.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Schwartz L, Supuran CT, Alfarouk KO. The Warburg effect and the hallmarks of cancer. Anticancer Agents Med Chem. 2017;17(2):164–70.PubMedCrossRef Schwartz L, Supuran CT, Alfarouk KO. The Warburg effect and the hallmarks of cancer. Anticancer Agents Med Chem. 2017;17(2):164–70.PubMedCrossRef
29.
Zurück zum Zitat Mycielska ME, Milenkovic VM, Wetzel CH, Rummele P, Geissler EK. Extracellular citrate in health and disease. Curr Mol Med. 2015;15(10):884–91.PubMedCrossRef Mycielska ME, Milenkovic VM, Wetzel CH, Rummele P, Geissler EK. Extracellular citrate in health and disease. Curr Mol Med. 2015;15(10):884–91.PubMedCrossRef
30.
Zurück zum Zitat Mycielska ME, Patel A, Rizaner N, Mazurek MP, Keun H, Patel A, et al. Citrate transport and metabolism in mammalian cells: prostate epithelial cells and prostate cancer. BioEssays. 2009;31(1):10–20.PubMedCrossRef Mycielska ME, Patel A, Rizaner N, Mazurek MP, Keun H, Patel A, et al. Citrate transport and metabolism in mammalian cells: prostate epithelial cells and prostate cancer. BioEssays. 2009;31(1):10–20.PubMedCrossRef
31.
Zurück zum Zitat Rodriguez-Enriquez S, Marin-Hernandez A, Gallardo-Perez JC, Pacheco-Velazquez SC, Belmont-Diaz JA, Robledo-Cadena DX, et al. Transcriptional regulation of energy metabolism in cancer cells. Cells. 2019;8(10): e1225.PubMedCrossRef Rodriguez-Enriquez S, Marin-Hernandez A, Gallardo-Perez JC, Pacheco-Velazquez SC, Belmont-Diaz JA, Robledo-Cadena DX, et al. Transcriptional regulation of energy metabolism in cancer cells. Cells. 2019;8(10): e1225.PubMedCrossRef
32.
Zurück zum Zitat Wei Z, Liu X, Cheng C, Yu W, Yi P. Metabolism of amino acids in cancer. Front Cell Dev Biol. 2020;8: e603837.CrossRef Wei Z, Liu X, Cheng C, Yu W, Yi P. Metabolism of amino acids in cancer. Front Cell Dev Biol. 2020;8: e603837.CrossRef
34.
Zurück zum Zitat Green CR, Wallace M, Divakaruni AS, Phillips SA, Murphy AN, Ciaraldi TP, et al. Branched-chain amino acid catabolism fuels adipocyte differentiation and lipogenesis. Nat Chem Biol. 2016;12(1):15–21.PubMedCrossRef Green CR, Wallace M, Divakaruni AS, Phillips SA, Murphy AN, Ciaraldi TP, et al. Branched-chain amino acid catabolism fuels adipocyte differentiation and lipogenesis. Nat Chem Biol. 2016;12(1):15–21.PubMedCrossRef
35.
Zurück zum Zitat Teahan O, Bevan CL, Waxman J, Keun HC. Metabolic signatures of malignant progression in prostate epithelial cells. Int J Biochem Cell Biol. 2011;43(7):1002–9.PubMedCrossRef Teahan O, Bevan CL, Waxman J, Keun HC. Metabolic signatures of malignant progression in prostate epithelial cells. Int J Biochem Cell Biol. 2011;43(7):1002–9.PubMedCrossRef
36.
Zurück zum Zitat Lapek JD Jr, Tombline G, Kellersberger KA, Friedman MR, Friedman AE. Evidence of histidine and aspartic acid phosphorylation in human prostate cancer cells. Naunyn-Schmiedeberg’s Arch Pharmacol. 2015;388(2):161–73.CrossRef Lapek JD Jr, Tombline G, Kellersberger KA, Friedman MR, Friedman AE. Evidence of histidine and aspartic acid phosphorylation in human prostate cancer cells. Naunyn-Schmiedeberg’s Arch Pharmacol. 2015;388(2):161–73.CrossRef
37.
Zurück zum Zitat Kumar D, Gupta A, Mandhani A, Sankhwar SN. NMR spectroscopy of filtered serum of prostate cancer: a new frontier in metabolomics. Prostate. 2016;76(12):1106–19.PubMedCrossRef Kumar D, Gupta A, Mandhani A, Sankhwar SN. NMR spectroscopy of filtered serum of prostate cancer: a new frontier in metabolomics. Prostate. 2016;76(12):1106–19.PubMedCrossRef
38.
Zurück zum Zitat Tessem MB, Swanson MG, Keshari KR, Albers MJ, Joun D, Tabatabai ZL, et al. Evaluation of lactate and alanine as metabolic biomarkers of prostate cancer using 1H HR-MAS spectroscopy of biopsy tissues. Magn Reson Med. 2008;60(3):510–6.PubMedPubMedCentralCrossRef Tessem MB, Swanson MG, Keshari KR, Albers MJ, Joun D, Tabatabai ZL, et al. Evaluation of lactate and alanine as metabolic biomarkers of prostate cancer using 1H HR-MAS spectroscopy of biopsy tissues. Magn Reson Med. 2008;60(3):510–6.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Sirnio P, Vayrynen JP, Klintrup K, Makela J, Karhu T, Herzig KH, et al. Alterations in serum amino-acid profile in the progression of colorectal cancer: associations with systemic inflammation, tumour stage and patient survival. Br J Cancer. 2019;120(2):238–46.PubMedCrossRef Sirnio P, Vayrynen JP, Klintrup K, Makela J, Karhu T, Herzig KH, et al. Alterations in serum amino-acid profile in the progression of colorectal cancer: associations with systemic inflammation, tumour stage and patient survival. Br J Cancer. 2019;120(2):238–46.PubMedCrossRef
40.
Zurück zum Zitat Corsetti S, Rabl T, McGloin D, Nabi G. Raman spectroscopy for accurately characterizing biomolecular changes in androgen-independent prostate cancer cells. J Biophotonics. 2018;11(3): e201700166.CrossRef Corsetti S, Rabl T, McGloin D, Nabi G. Raman spectroscopy for accurately characterizing biomolecular changes in androgen-independent prostate cancer cells. J Biophotonics. 2018;11(3): e201700166.CrossRef
41.
Zurück zum Zitat Akbari Z, Dijojin RT, Zamani Z, Hosseini RH, Arjmand M. Aromatic amino acids play a harmonizing role in prostate cancer: a metabolomics-based cross-sectional study. Int J Reprod Biomed. 2021;19(8):741–50.PubMedPubMedCentral Akbari Z, Dijojin RT, Zamani Z, Hosseini RH, Arjmand M. Aromatic amino acids play a harmonizing role in prostate cancer: a metabolomics-based cross-sectional study. Int J Reprod Biomed. 2021;19(8):741–50.PubMedPubMedCentral
42.
Zurück zum Zitat Gomez-Cebrian N, Garcia-Flores M, Rubio-Briones J, Lopez-Guerrero JA, Pineda-Lucena A, Puchades-Carrasco L. Targeted metabolomics analyses reveal specific metabolic alterations in high-grade prostate cancer patients. J Proteome Res. 2020;19(10):4082–92.PubMedCrossRef Gomez-Cebrian N, Garcia-Flores M, Rubio-Briones J, Lopez-Guerrero JA, Pineda-Lucena A, Puchades-Carrasco L. Targeted metabolomics analyses reveal specific metabolic alterations in high-grade prostate cancer patients. J Proteome Res. 2020;19(10):4082–92.PubMedCrossRef
43.
Zurück zum Zitat Khan A, Choi SA, Na J, Pamungkas AD, Jung KJ, Jee SH, et al. Noninvasive serum metabolomic profiling reveals elevated Kynurenine pathway’s metabolites in humans with prostate cancer. J Proteome Res. 2019;18(4):1532–41.PubMedCrossRef Khan A, Choi SA, Na J, Pamungkas AD, Jung KJ, Jee SH, et al. Noninvasive serum metabolomic profiling reveals elevated Kynurenine pathway’s metabolites in humans with prostate cancer. J Proteome Res. 2019;18(4):1532–41.PubMedCrossRef
44.
Zurück zum Zitat Moller N. Ketone body, 3-hydroxybutyrate: minor metabolite - major medical manifestations. J Clin Endocrinol Metab. 2020;105(9):dgaa370.PubMedCrossRef Moller N. Ketone body, 3-hydroxybutyrate: minor metabolite - major medical manifestations. J Clin Endocrinol Metab. 2020;105(9):dgaa370.PubMedCrossRef
45.
Zurück zum Zitat Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262–84.PubMedPubMedCentralCrossRef Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262–84.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Yoshii Y, Furukawa T, Saga T, Fujibayashi Y. Acetate/acetyl-CoA metabolism associated with cancer fatty acid synthesis: overview and application. Cancer Lett. 2015;356(2 Pt A):211–6.PubMedCrossRef Yoshii Y, Furukawa T, Saga T, Fujibayashi Y. Acetate/acetyl-CoA metabolism associated with cancer fatty acid synthesis: overview and application. Cancer Lett. 2015;356(2 Pt A):211–6.PubMedCrossRef
47.
Zurück zum Zitat Saraon P, Trudel D, Kron K, Dmitromanolakis A, Trachtenberg J, Bapat B, et al. Evaluation and prognostic significance of ACAT1 as a marker of prostate cancer progression. Prostate. 2014;74(4):372–80.PubMedCrossRef Saraon P, Trudel D, Kron K, Dmitromanolakis A, Trachtenberg J, Bapat B, et al. Evaluation and prognostic significance of ACAT1 as a marker of prostate cancer progression. Prostate. 2014;74(4):372–80.PubMedCrossRef
48.
Zurück zum Zitat Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, Howell A, Sotgia F, Lisanti MP. Ketone body utilization drives tumor growth and metastasis. Cell Cycle. 2012;11(21):3964–71.PubMedPubMedCentralCrossRef Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, Howell A, Sotgia F, Lisanti MP. Ketone body utilization drives tumor growth and metastasis. Cell Cycle. 2012;11(21):3964–71.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Huang J, Mondul AM, Weinstein SJ, Derkach A, Moore SC, Sampson JN, et al. Prospective serum metabolomic profiling of lethal prostate cancer. Int J Cancer. 2019;145(12):3231–43.PubMedPubMedCentralCrossRef Huang J, Mondul AM, Weinstein SJ, Derkach A, Moore SC, Sampson JN, et al. Prospective serum metabolomic profiling of lethal prostate cancer. Int J Cancer. 2019;145(12):3231–43.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Ducker GS, Chen L, Morscher RJ, Ghergurovich JM, Esposito M, Teng X, et al. Reversal of cytosolic one-carbon flux compensates for loss of the mitochondrial folate pathway. Cell Metab. 2016;24(4):640–1.PubMedCrossRef Ducker GS, Chen L, Morscher RJ, Ghergurovich JM, Esposito M, Teng X, et al. Reversal of cytosolic one-carbon flux compensates for loss of the mitochondrial folate pathway. Cell Metab. 2016;24(4):640–1.PubMedCrossRef
51.
Zurück zum Zitat Maddocks OD, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E, et al. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature. 2013;493(7433):542–6.PubMedCrossRef Maddocks OD, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E, et al. Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature. 2013;493(7433):542–6.PubMedCrossRef
52.
Zurück zum Zitat Meiser J, Tumanov S, Maddocks O, Labuschagne CF, Athineos D, Van Den Broek N, et al. Serine one-carbon catabolism with formate overflow. Sci Adv. 2016;2(10): e1601273.PubMedPubMedCentralCrossRef Meiser J, Tumanov S, Maddocks O, Labuschagne CF, Athineos D, Van Den Broek N, et al. Serine one-carbon catabolism with formate overflow. Sci Adv. 2016;2(10): e1601273.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Meiser J, Schuster A, Pietzke M, Vande Voorde J, Athineos D, Oizel K, et al. Increased formate overflow is a hallmark of oxidative cancer. Nat Commun. 2018;9(1): e1368.CrossRef Meiser J, Schuster A, Pietzke M, Vande Voorde J, Athineos D, Oizel K, et al. Increased formate overflow is a hallmark of oxidative cancer. Nat Commun. 2018;9(1): e1368.CrossRef
54.
Zurück zum Zitat Grosman H, Fabre B, Mesch V, Lopez MA, Schreier L, Mazza O, et al. Lipoproteins, sex hormones and inflammatory markers in association with prostate cancer. Aging Male. 2010;13(2):87–92.PubMedCrossRef Grosman H, Fabre B, Mesch V, Lopez MA, Schreier L, Mazza O, et al. Lipoproteins, sex hormones and inflammatory markers in association with prostate cancer. Aging Male. 2010;13(2):87–92.PubMedCrossRef
56.
Zurück zum Zitat Moses KA, Abd TT, Goodman M, Hsiao W, Hall JA, Marshall FF, et al. Increased low density lipoprotein and increased likelihood of positive prostate biopsy in black americans. J Urol. 2009;182(5):2219–25.PubMedCrossRef Moses KA, Abd TT, Goodman M, Hsiao W, Hall JA, Marshall FF, et al. Increased low density lipoprotein and increased likelihood of positive prostate biopsy in black americans. J Urol. 2009;182(5):2219–25.PubMedCrossRef
57.
Zurück zum Zitat Borgquist S, Butt T, Almgren P, Shiffman D, Stocks T, Orho-Melander M, et al. Apolipoproteins, lipids and risk of cancer. Int J Cancer. 2016;138(11):2648–56.PubMedCrossRef Borgquist S, Butt T, Almgren P, Shiffman D, Stocks T, Orho-Melander M, et al. Apolipoproteins, lipids and risk of cancer. Int J Cancer. 2016;138(11):2648–56.PubMedCrossRef
58.
Zurück zum Zitat Bull CJ, Bonilla C, Holly JM, Perks CM, Davies N, Haycock P, et al. Blood lipids and prostate cancer: a Mendelian randomization analysis. Cancer Med. 2016;5(6):1125–36.PubMedPubMedCentralCrossRef Bull CJ, Bonilla C, Holly JM, Perks CM, Davies N, Haycock P, et al. Blood lipids and prostate cancer: a Mendelian randomization analysis. Cancer Med. 2016;5(6):1125–36.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Liu YP, Zhang YX, Li PF, Cheng C, Zhao YS, Li DP, et al. Cholesterol levels in blood and the risk of prostate cancer: a meta-analysis of 14 prospective studies. Cancer Epidemiol Biomarkers Prev. 2015;24(7):1086–93.CrossRef Liu YP, Zhang YX, Li PF, Cheng C, Zhao YS, Li DP, et al. Cholesterol levels in blood and the risk of prostate cancer: a meta-analysis of 14 prospective studies. Cancer Epidemiol Biomarkers Prev. 2015;24(7):1086–93.CrossRef
Metadaten
Titel
Identification of characteristic metabolic panels for different stages of prostate cancer by 1H NMR-based metabolomics analysis
verfasst von
Xi Zhang
Binbin Xia
Hong Zheng
Jie Ning
Yinjie Zhu
Xiaoguang Shao
Binrui Liu
Baijun Dong
Hongchang Gao
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03478-5

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

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