Next Article in Journal
Exploring the Impact of Nutrition on Non-Alcoholic Fatty Liver Disease Management: Unveiling the Roles of Various Foods, Food Components, and Compounds
Previous Article in Journal
Stingless Bee (Heterotrigona Itama) Honey and Its Phenolic-Rich Extract Ameliorate Oxidant–Antioxidant Balance via KEAP1-NRF2 Signalling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Metabolomic Analysis of Vitamin E Supplement Use in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial

by
Jungeun Lim
1,
Hyokyoung G. Hong
1,
Stephanie J. Weinstein
1,
Mary C. Playdon
2,
Amanda J. Cross
3,4,
Rachael Stolzenberg-Solomon
1,
Neal D. Freedman
1,
Jiaqi Huang
1 and
Demetrius Albanes
1,*
1
Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
2
University of Utah and Cancer Control and Population Sciences Program, Department of Nutrition and Integrative Physiology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
3
Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2AZ, UK
4
Cancer Screening & Prevention Research Group, Department of Surgery & Cancer, Imperial College London, London SW7 2AZ, UK
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(13), 2836; https://doi.org/10.3390/nu15132836
Submission received: 26 May 2023 / Revised: 13 June 2023 / Accepted: 15 June 2023 / Published: 22 June 2023
(This article belongs to the Section Nutrition and Public Health)

Abstract

:
The effects of vitamin E supplementation on cancer and other chronic diseases are not clear. We compared the serum metabolomic profile of differing vitamin E dosages in order to re-examine the previously observed changes in a novel C22 lactone sulfate compound, androgenic steroids, and other metabolites. A total of 3409 women and men previously selected for metabolomics studies in the PLCO Cancer Screening Trial were included in this investigation. Serum metabolites were profiled using ultrahigh-performance liquid and gas chromatography/tandem mass spectrometry. Seventy known metabolites including C22 lactone sulfate and androgens were significantly associated with vitamin E supplementation. In the sex-stratified analysis, 10 cofactors and vitamins (e.g., alpha-CEHC sulfate and alpha-CEHC glucuronide), two carbohydrates (glyceric and oxalic acids), and one lipid (glycocholenate sulfate) were significantly associated with vitamin E dose in both males and females (FDR-adjusted p-value < 0.01). However, the inverse association between C22 lactone sulfate and daily vitamin E supplementation was evident in females only, as were two androgenic steroids, 5-androstenediol and androsterone glucuronide. Our study provides evidence of distinct steroid hormone pathway responses based on vitamin E dosages. Further studies are needed to gain biological insights into vitamin E biochemical effects relevant to cancer and other chronic diseases.

1. Introduction

Alpha-tocopherol is a common dietary and supplemental vitamin E compound that is preferentially taken up by the liver and distributed throughout the body, serving as a major lipid antioxidant [1]. Epidemiological studies and laboratory experiments indicate inverse associations between vitamin E intake from dietary sources and/or supplements and cancer risk [2,3]. Along with increased interest in research on cancer treatment, it has also been reported that vitamin E is involved in the amelioration of side-effects from chemotherapy and radiation therapy [4,5]. Alpha-tocopherol may prevent cancer by inhibiting cell proliferation and angiogenesis, inducing apoptosis and enhancing immune function [6,7,8]. In a randomized controlled trial (RCT), a 50 IU daily dose of vitamin E (as alpha-tocopheryl acetate) resulted in significant 32% and 40% reductions in prostate cancer incidence and mortality, respectively [9]. However, subsequent RCTs using higher vitamin E dosages of 200 IU or 400 IU daily showed contradictory (17% increased) or insignificant effects on prostate cancer incidence [10,11].
To gain biological insights into vitamin E biochemical effects relevant to cancer, including the divergent prostate cancer findings in those RCTs, a metabolomic analysis was conducted in another RCT of high-dose vitamin E, which showed the expected increases in serum vitamin E-related metabolites including alpha-carboxyethyl hydrochroman (CEHC) sulfate and alpha-tocopherol in response to the 400 IU/day vitamin E, as well as significant reductions in a novel C22 lactone sulfate and androgen metabolites [12]. A strong correlation was also observed between the changes in androgenic steroid metabolites and the vitamin E supplement-associated reduction in C22 lactone sulfate in the 400 IU/day RCT only. As for the other cancer sites, in vitro studies and animal models of breast cancer supplemented with tocopherol or tocotrienol vitamers showed antitumor and chemo-preventive activity [13], and inverse associations have been observed between vitamin E consumption and risk of bladder and esophageal cancer [14,15].
In the present cross-sectional investigation, we compared the serum metabolomic profile of low and high-dose vitamin E supplementation in order to re-examine the previously observed changes in a novel C22 lactone sulfate compound, androgenic steroids, and other metabolites.

2. Materials and Methods

2.1. Study Population

Data for this study were derived from an RCT, the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. The design of this trial was described previously [16]. In brief, participants aged 55 to 74 years were recruited from 10 study centers across the US between September 1993 and June 2001. Participants provided written informed consent, and the study was approved by the institutional review boards of the US National Cancer Institute and the ten PLCO screening centers.
The present investigation includes participants in the PLCO screening arm who were previously selected for seven metabolomic nested case–control studies of cancer risk. These included two studies of prostate (525 cases and 541 controls) [17] (and unpublished), and one study each of colorectum (254 cases and 253 controls) [18], breast (594 cases and 593 controls) [19], pancreas (97 cases and 102 controls) [20], esophagus (127 cases and 131 controls; unpublished), and glioma (158 cases and 161 controls; unpublished). After excluding 127 participants missing data for daily vitamin E supplement intake, a total of 3409 participants including 1837 males and 1572 females with serum metabolomic data were included in the present analysis (Figure 1). All vitamin supplement and metabolomic data were from baseline (i.e., before cancers were diagnosed).

2.2. Serum and DATA Collection

At enrollment, participants completed questionnaires regarding behavioral and lifestyle information, including use of supplemental vitamin E and multivitamins, and smoking status (i.e., never, current, and former), and blood (of variable fasting states) was collected and processed to serum. Daily vitamin E supplement dosages were calculated from single-vitamin and multivitamin supplementation questions. Regarding the use of vitamin E capsules/tablets, people responded to the question ‘what dose per day did/do you usually take? (i.e., 100 IU, 200 IU, 400 IU, 800 IU, 1000 + IU, do not know)’. Participants were asked to answer the following question for multivitamin use: How many pills did/do you usually take? (i.e., <2/week, 2–4/week, 5–6/week, 1/day, and 2+/day). Dosage values for multivitamins were derived from the Third National Health and Nutrition Examination Survey (NHANES III) database (e.g., 30 IU).

2.3. Metabolomic Analysis

Serum metabolites were assayed at Metabolon, Inc. (Durham, NC, USA) using a high-resolution, accurate mass platform of ultrahigh-performance liquid chromatography/mass spectrometry and gas chromatograph/mass spectrometry (GC–MS). The details of the analytical platform that integrated the chemical analysis were described in [21,22,23]. A total of 2356 metabolites were detected. After excluding those with >10% missing values, 1713 compounds (including 1155 known metabolites) remained for analysis. On the basis of the existing literature, metabolites were categorized across eight mutually exclusive chemical classes: amino acids, carbohydrates, cofactors and vitamins, energy metabolites, lipids, nucleotides, peptides, and xenobiotics [24].

2.4. Statistical Analysis

According to the daily vitamin E supplementation dosages and multivitamin use, we created three daily dosage categories: 0 IU, 4.3–<400 IU, and ≥400 IU. ANCOVA analysis was used to identify metabolites showing significant differences among daily vitamin E supplementation levels, adjusting for potential confounders such as age (continuous), sex (male or female), body weight (continuous), smoking status (never, current, and former), and multivitamin use (yes or no). Including a variable for case status (case, control) in the model did not materially alter the vitamin E association with any metabolite (i.e., <10% change).
Metabolites associated with daily supplemental vitamin E intake were determined for all participants combined and separately for males and females. Sensitivity analyses were conducted to additionally adjust for alcohol and dietary consumption (i.e., whole grains, fruits, eggs, dairy products, fish, and meats). Considering the higher vitamin E dose of therapeutic multivitamins (e.g., 60 IU), sensitivity analyses were also conducted that excluded therapeutic multivitamin use. Models were also stratified by age (63 years), body mass index (BMI, 27.2 kg/m2 for males and 26.6 kg/m2 for women), body weight (86.2 kg for males and 70.3 kg for females) based on median values, and smoking status (never, current, and former).
To account for multiple comparisons, the false discovery rate (FDR) using the Benjamini and Hochberg approach [25] was used to help to avoid type I errors [26]. All analyses were performed using SAS statistical software version 9.4 (SAS Institute, Cary, NC, USA). All reported p-values were two-sided. The null hypothesis of no difference was rejected if p-values were <0.05.

3. Results

Baseline characteristics of the study participants according to daily supplemental vitamin E dosage are presented in Table 1. The study population included 1837 (53.9%) males and 1572 (46.1%) females. Of these, 835 males and 919 females reported taking a single vitamin E supplement or multivitamin, with the median supplemental vitamin E intake being 200 IU and 400 IU, respectively.
Among the 1713 compounds measured, 70 known metabolites, including alpha-tocopherol and its metabolites, C22 lactone sulfate, and androgen metabolites, were significantly associated with daily supplemental vitamin E intake and showed some dose–response gradients (Table 2). Metabolites in tocopherol metabolism such as alpha-CEHC, alpha-CEHC-glucuronide, and alpha-CEHC sulfate were significantly positively associated with supplemental vitamin E after multiple comparisons correction (FDR-adjusted p-value <0.001 for each metabolite). C22 lactone sulfate was significantly lower with increasing vitamin E dosage (betas of −0.04 and −0.17 and p-values 0.500 and <0.001 for the <400 and >400 IU categories, respectively) (Table 2). Results were consistent when we additionally adjusted for dietary and alcohol consumption. Nine sex steroids, including seven androgenic steroids, were inversely and significantly associated with vitamin E supplement use (FDR-adjusted p-value <0.01 for each metabolite; Table 2).
In the sex-stratified analysis, 13 metabolites including 10 cofactors and vitamins such as alpha-CEHC sulfate and alpha-CEHC glucuronide, two carbohydrates (glyceric acid and oxalic acid), and glycocholenate sulfate were significantly related to vitamin E supplementation in both males and females (FDR-adjusted p-value < 0.01) (Table 3). An inverse association between C22 lactone sulfate and dose of supplemental vitamin E was apparent only in females (FDR-adjusted p-value = 0.046, p for interaction = 0.0218). This finding was unchanged when we additionally adjusted for dietary and alcohol consumption. 5-Androstenediol and androsterone glucuronide were also inversely and significantly associated with vitamin E intake only in females (p for interaction = 0.0214 and 0.0008, respectively). In the body weight-stratified analysis, C22 lactone sulfate (FDR-adjusted p-value = 0.139) and two androgen metabolites, androsterone glucuronide (FDR-adjusted p-value = 0.006) and 5-androstenediol (FDR-adjusted p-value = 0.064), were significantly related to vitamin E supplementation only in females >70.3 kg (the median) (data not shown). There were no apparent interactions of the vitamin E-metabolite associations with age, BMI, or smoking status. Our findings were not materially altered when we excluded therapeutic multivitamin users.

4. Discussion

In this first metabolomic analysis of vitamin E supplementation in both males and females, along with the expected increases in alpha-tocopherol and its metabolites, a novel C22 lactone sulfate compound and androgenic steroid metabolites were significantly lower in vitamin E supplement users, with some indication of a dose–response relationship. Interestingly, the findings were stronger in females than in males, whereas a previous RCT-based study showed a strong signal for C22 lactone sulfate and androgens in males [10].
Previous metabolomic studies of different cancers having a focus on vitamin E are limited. Regarding the biological insights into vitamin E biochemical effects (including sex steroids) relevant to cancers, the delta-tocopherol vitamers significant in our study showed antitumor and adjuvant chemopreventive potential in breast cancer cells [13]. Although prospective studies in healthy women have consistently shown a strong relationship between high serum androgen levels and increased risk of developing ER-positive breast cancers [27,28], the link between androgen receptor (AR) positivity and improved outcomes in ER-positive tumors has revealed the beneficial effect of androgens [29]. The inhibition of glioma cell proliferation induced by tocopherols was also observed in experimental studies [30], but inconsistent associations were found for colon cancer risk and vitamin E supplements [31] even though a meta-analysis of case–control studies showed lower concentrations of serum vitamin E in patients with colorectal cancer compared with healthy controls [32]. Regarding esophageal cancer risk, an in vivo study suggested vitamin E may suppress N-nitrosomethylbenzylamine-induced carcinogenesis in the rat esophagus by blocking activation of nuclear factor-kappa B (NF-κB) and abnormal arachidonic acid metabolism [33]. Little is known regarding vitamin E-related metabolites and pancreatic cancer risk.
With regard to prostate cancer risk, an early large prevention trial of male Finnish smokers showed significant 32% and 40% reductions in prostate cancer incidence and mortality, respectively, in response to a modest daily dose of 50 IU dl-alpha-tocopheryl acetate. Subsequent ‘confirmator’” trials using substantially higher vitamin E dosages of 200 and 400 IU/day yielded contradictory outcomes (e.g., 17% increased incidence) [11] and no effects on prostate cancer [10]. Although the present study provided little evidence for distinct steroid hormone pathway changes based on varying vitamin E dosages in males which could have direct relevance to prostate cancer risk, changes in androgenic steroid metabolites were strongly correlated with the high-dose vitamin E supplement-associated change in C22 lactone sulfate in the Vitamin E Atherosclerosis Prevention Study (VEAPS) trial [12], with similar, albeit more modest, associations in females in the present study. Further investigation is needed to more fully understand the interrelationships among C22 lactone sulfate, androgenic hormones, and vitamin E supplementation, including possible sex differences.
The molecular structure of C22 lactone sulfate suggests possible involvement in the lanosterol synthase pathway [10]. One genome-wide association study (GWAS) showed that C22 lactone sulfate variation was associated with cytochrome P450 (CYP) 3A5 on chromosome 7 which encodes a member of the CYP superfamily of enzymes involved in drug metabolism (including ibuprofen) and synthesis of cholesterol, steroids, and other lipids related to the lipid-lowering efficacy of simvastatin [34,35,36]. Metabolism of vitamin E involves CYP enzymes, with CYP3A4 and CYP4F2 suggested to be involved in tocopherol degradation and drug metabolism, the latter being substrate-induced through activation of the pregnane X receptor (PXR) [37]. Tocopherols and tocotrienols induce the expression of a PXR-driven reporter gene and endogenous CYP3A4 and CYP3A5. With respect to prostate cancer, higher PXR expression in cancerous versus normal tissues has been observed, and PXR activation is associated with increased tumor progression and resistance to the chemotherapeutic drugs [38,39,40]. C22 lactone sulfate has also been associated with genetic variation of the protein-coding SLCO1B1 which is involved in hepatic uptake of statins [41]. Since prospective and registry-based studies support a lower risk of advanced and fatal prostate cancer in statin users relative to nonusers, as well as better outcomes among prostate cancer patients, C22 lactone sulfate may be related to the statin–prostate cancer association [42]. Functional studies of this compound are needed to elucidate the precise biochemical actions and pathways involved.
It is interesting to note that significant inverse associations between androgenic steroids and vitamin E intake were observed only in females in our study. The results were unchanged after additional adjustment for use of hormone replacement therapy. Experiments have shown that alpha-tocopheryl succinate can suppress androgen receptor (AR) expression by means of transcriptional and posttranscriptional modulation [43]. Results from NHANES III showed an inverse association between serum alpha-tocopherol and circulating testosterone, estradiol, and sex hormone-binding globulin, but only in males who smoked [44]. The effect of sex on the relationship between androgenic steroid metabolites and vitamin E should be re-examined within other populations.
This is the first metabolomic analysis of vitamin E supplementation in males and females. We utilized an untargeted metabolomic platform exhibiting high laboratory validity and reproducibility. More than 1700 metabolites reflecting a broad array of biochemicals and biological pathways were identified. Application of a rigorous FDR correction for multiple comparisons revealed several individual metabolites related to higher vitamin E dosage, including C22 lactone sulfate and androgens. Although use of self-reported supplementation data afforded us a less stringent test of the vitamin E/metabolite hypothesis as compared with the previous controlled trial analysis in males [12], dose–response associations between alpha-tocopherol and its metabolites and increasing vitamin E dosages support the validity of our exposure measure; that is, elevated alpha-CEHC and alpha-tocopherol, and decreased delta-tocopherol reflected the increase in serum vitamin E from higher supplement dosage. Including participants who had been previously selected for nested case–control studies of metabolomics in the PLCO trial is a potential limitation; however, supplement use data and metabolite measurements were based on baseline reports and serum up to two decades prior to cancer diagnoses. Furthermore, our primary findings were similar when we restricted the analysis to the control participants. Despite the large number of measured compounds, other unmeasured metabolites or biochemical pathways related to high-dose vitamin E supplementation may not have been identified using the assay platform utilized. It is also possible that some findings of xenobiotic drugs (e.g., 2-methoxyacetaminophen glucuronide) were confounded by other drug intake such as acetaminophen, and other diet biomarkers may have affected the findings although we adjusted for potential confounding factors.
In conclusion, vitamin E supplementation was associated with significantly lower serum C22 lactone sulfate and androgenic steroid metabolites in PLCO. The associations were evident primarily in females, however. Although our study provides some evidence of distinct steroid hormone pathway responses based on vitamin E dosages that could have direct relevance to previous trial findings for prostate cancer, they require re-examination in other populations of males and females, including especially in vitamin E clinical trial settings, where possible. Since experimental evidence does support inhibitory effects of lactone-containing metabolites and lactone-based derivatives on cancer cell line growth [45,46,47], re-examination in other populations along with further elucidation of the interrelationships and biochemical pathways among C22 lactone sulfate and vitamin E would be helpful to gain greater biological insights into the biochemical effects of vitamin E relevant to cancer.

Author Contributions

Study design, J.L. and D.A.; statistical analysis and writing—original draft, J.L.; Methodology, H.G.H., J.L., and D.A.; writing—review & editing, S.J.W., H.G.H., M.C.P., A.J.C., R.S.-S., N.D.F., J.H., and D.A.; study oversight and primary responsibility for the final content, D.A. All authors have read and agreed to the published version of the manuscript.

Funding

The PLCO Trial and cohort were supported by the Intramural Research Program of the Division of Cancer Epidemiology and Genetics, and contracts from the Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Department of Health and Human Services.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of the United States NCI.

Informed Consent Statement

Written informed consent has been obtained from participants included in the PLCO study.

Data Availability Statement

The data used in this study can be obtained from the PLCO website (https://cdas.cancer.gov/learn/plco/instructions/ (accessed on 16 June 2023)).

Acknowledgments

The authors thank the PLCO participants, the NCI study management team, and staff at Information Management Services, Inc., Westat, Inc., Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., and American Type Culture Collection.

Conflicts of Interest

The authors declare no conflict of interest. The design of the present study and analysis, interpretation of the data, writing of the manuscript, and decision to submit the manuscript for publication rest solely with the individual study investigators.

References

  1. Traber, M.G.; Sies, H. Vitamin E in humans: Demand and delivery. Annu. Rev. Nutr. 1996, 16, 321–347. [Google Scholar] [CrossRef] [PubMed]
  2. Huang, J.; Weinstein, S.J.; Yu, K.; Mannisto, S.; Albanes, D. A Prospective Study of Serum Vitamin E and 28-Year Risk of Lung Cancer. J. Natl. Cancer Inst. 2020, 112, 191–199. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, H.; Yang, X.; Liu, A.; Wang, G.; Bosland, M.C.; Yang, C.S. delta-Tocopherol inhibits the development of prostate adenocarcinoma in prostate specific Pten-/- mice. Carcinogenesis 2018, 39, 158–169. [Google Scholar] [CrossRef] [PubMed]
  4. Zhou, J.; Rao, L.; Yu, G.; Cook, T.R.; Chen, X.; Huang, F. Supramolecular cancer nanotheranostics. Chem. Soc. Rev. 2021, 50, 2839–2891. [Google Scholar] [CrossRef] [PubMed]
  5. Jain, A.; Tiwari, A.; Verma, A.; Jain, S.K. Vitamins for Cancer Prevention and Treatment: An Insight. Curr. Mol. Med. 2017, 17, 321–340. [Google Scholar] [CrossRef] [PubMed]
  6. Shklar, G.; Schwartz, J.L. Vitamin E inhibits experimental carcinogenesis and tumour angiogenesis. Eur. J. Cancer B Oral Oncol. 1996, 32B, 114–119. [Google Scholar] [CrossRef]
  7. Sigounas, G.; Anagnostou, A.; Steiner, M. dl-alpha-tocopherol induces apoptosis in erythroleukemia, prostate, and breast cancer cells. Nutr. Cancer 1997, 28, 30–35. [Google Scholar] [CrossRef]
  8. Meydani, S.N.; Beharka, A.A. Recent developments in vitamin E and immune response. Nutr. Rev. 1998, 56, S49–S58. [Google Scholar] [CrossRef]
  9. Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. New Engl. J. Med. 1994, 330, 1029–1035. [Google Scholar] [CrossRef]
  10. Gaziano, J.M.; Glynn, R.J.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Sesso, H.D.; Buring, J.E. Vitamins E and C in the prevention of prostate and total cancer in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2009, 301, 52–62. [Google Scholar] [CrossRef] [Green Version]
  11. Klein, E.A.; Thompson, I.M., Jr.; Tangen, C.M.; Crowley, J.J.; Lucia, M.S.; Goodman, P.J.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; et al. Vitamin E and the Risk of Prostate Cancer: Updated Results of The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556. [Google Scholar] [CrossRef] [Green Version]
  12. Huang, J.; Hodis, H.N.; Weinstein, S.J.; Mack, W.J.; Sampson, J.N.; Mondul, A.M.; Albanes, D. Serum Metabolomic Response to Low- and High-Dose Vitamin E Supplementation in Two Randomized Controlled Trials. Cancer Epidemiol. Biomark. Prev. 2020, 29, 1329–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. de Sousa Coelho, M.; Pereira, I.C.; de Oliveira, K.G.F.; Oliveira, I.K.F.; Dos Santos Rizzo, M.; de Oliveira, V.A.; Carneiro da Silva, F.C.; Torres-Leal, F.L.; de Castro E Sousa, J.M. Chemopreventive and anti-tumor potential of vitamin E in preclinical breast cancer studies: A systematic review. Clin. Nutr. ESPEN 2023, 53, 60–73. [Google Scholar] [CrossRef] [PubMed]
  14. Lin, J.H.; Chen, S.J.; Liu, H.; Yan, Y.; Zheng, J.H. Vitamin E consumption and the risk of bladder cancer. Int. J. Vitam. Nutr. Res. 2019, 89, 168–175. [Google Scholar] [CrossRef] [PubMed]
  15. Cui, L.; Li, L.; Tian, Y.; Xu, F.; Qiao, T. Association between Dietary Vitamin E Intake and Esophageal Cancer Risk: An Updated Meta-Analysis. Nutrients 2018, 10, 801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Prorok, P.C.; Andriole, G.L.; Bresalier, R.S.; Buys, S.S.; Chia, D.; Crawford, E.D.; Fogel, R.; Gelmann, E.P.; Gilbert, F.; Hasson, M.A.; et al. Design of the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. Control Clin. Trials 2000, 21, 273S–309S. [Google Scholar] [CrossRef]
  17. Huang, J.; Mondul, A.M.; Weinstein, S.J.; Koutros, S.; Derkach, A.; Karoly, E.; Sampson, J.N.; Moore, S.C.; Berndt, S.I.; Albanes, D. Serum metabolomic profiling of prostate cancer risk in the prostate, lung, colorectal, and ovarian cancer screening trial. Br. J. Cancer 2016, 115, 1087–1095. [Google Scholar] [CrossRef] [Green Version]
  18. Cross, A.J.; Moore, S.C.; Boca, S.; Huang, W.Y.; Xiong, X.; Stolzenberg-Solomon, R.; Sinha, R.; Sampson, J.N. A prospective study of serum metabolites and colorectal cancer risk. Cancer 2014, 120, 3049–3057. [Google Scholar] [CrossRef] [Green Version]
  19. Playdon, M.C.; Ziegler, R.G.; Sampson, J.N.; Stolzenberg-Solomon, R.; Thompson, H.J.; Irwin, M.L.; Mayne, S.T.; Hoover, R.N.; Moore, S.C. Nutritional metabolomics and breast cancer risk in a prospective study. Am. J. Clin. Nutr. 2017, 106, 637–649. [Google Scholar] [CrossRef] [Green Version]
  20. Stolzenberg-Solomon, R.; Derkach, A.; Moore, S.; Weinstein, S.J.; Albanes, D.; Sampson, J. Associations between metabolites and pancreatic cancer risk in a large prospective epidemiological study. Gut 2020, 69, 2008–2015. [Google Scholar] [CrossRef]
  21. Ford, L.; Kennedy, A.D.; Goodman, K.D.; Pappan, K.L.; Evans, A.M.; Miller, L.A.D.; Wulff, J.E.; Wiggs, B.R.; Lennon, J.J.; Elsea, S.; et al. Precision of a Clinical Metabolomics Profiling Platform for Use in the Identification of Inborn Errors of Metabolism. J. Appl. Lab. Med. 2020, 5, 342–356. [Google Scholar] [CrossRef] [PubMed]
  22. Evans, A.M.; DeHaven, C.D.; Barrett, T.; Mitchell, M.; Milgram, E. Integrated, nontargeted ultrahigh performance liquid chromatography/electrospray ionization tandem mass spectrometry platform for the identification and relative quantification of the small-molecule complement of biological systems. Anal. Chem. 2009, 81, 6656–6667. [Google Scholar] [CrossRef] [PubMed]
  23. Dehaven, C.D.; Evans, A.M.; Dai, H.; Lawton, K.A. Organization of GC/MS and LC/MS metabolomics data into chemical libraries. J. Cheminform. 2010, 2, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mondul, A.M.; Moore, S.C.; Weinstein, S.J.; Karoly, E.D.; Sampson, J.N.; Albanes, D. Metabolomic analysis of prostate cancer risk in a prospective cohort: The alpha-tocolpherol, beta-carotene cancer prevention (ATBC) study. Int. J. Cancer 2015, 137, 2124–2132. [Google Scholar] [CrossRef] [PubMed]
  25. Benjamini, Y.; Hochberg, Y. Controlling the false discovery rate: A practical and powerful approach to multiple testing. J. R. Stat. Soc. 1995, 57, 289–300. [Google Scholar] [CrossRef]
  26. Wang, N.; Ru, Y.; Yang, Z.; Sun, C.; Li, S.; Min, Y.; Zhao, X.; Lu, Y.; Hsing, A.W.; Zhu, S. Metabolomic Profiles of Plasma Retinol-Associated Dyslipidemia in Men and Women. Front. Nutr. 2021, 8, 740435. [Google Scholar] [CrossRef]
  27. Missmer, S.A.; Eliassen, A.H.; Barbieri, R.L.; Hankinson, S.E. Endogenous estrogen, androgen, and progesterone concentrations and breast cancer risk among postmenopausal women. J. Natl. Cancer Inst. 2004, 96, 1856–1865. [Google Scholar] [CrossRef] [PubMed]
  28. Cummings, S.R.; Lee, J.S.; Lui, L.Y.; Stone, K.; Ljung, B.M.; Cauleys, J.A. Sex hormones, risk factors, and risk of estrogen receptor–positive breast cancer in older women: A long-term prospective study. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1047–1051. [Google Scholar] [CrossRef] [Green Version]
  29. Elebro, K.; Borgquist, S.; Simonsson, M.; Markkula, A.; Jirström, K.; Ingvar, C.; Rose, C.; Jernström, H. Combined androgen and estrogen receptor status in breast cancer: Treatment prediction and prognosis in a population-based prospective cohort. Clin. Cancer Res. 2015, 21, 3640–3650. [Google Scholar] [CrossRef] [Green Version]
  30. Samandari, E.; Visarius, T.; Zingg, J.M.; Azzi, A. The effect of gamma-tocopherol on proliferation, integrin expression, adhesion, and migration of human glioma cells. Biochem. Biophys. Res. Commun. 2006, 342, 1329–1333. [Google Scholar] [CrossRef]
  31. Heine-Bröring, R.C.; Winkels, R.M.; Renkema, J.M.; Kragt, L.; van Orten-Luiten, A.C.; Tigchelaar, E.F.; Chan, D.S.; Norat, T.; Kampman, E. Dietary supplement use and colorectal cancer risk: A systematic review and meta-analyses of prospective cohort studies. Int. J. Cancer 2015, 136, 2388–2401. [Google Scholar] [CrossRef] [PubMed]
  32. Dong, Y.; Liu, Y.; Shu, Y.; Chen, X.; Hu, J.; Zheng, R.; Ma, D.; Yang, C.; Guan, X. Link between risk of colorectal cancer and serum vitamin E levels: A meta-analysis of case-control studies. Medicine 2017, 9, e7470. [Google Scholar] [CrossRef] [PubMed]
  33. Hui, Y.; Jia, X.; Chen, X.; Yang, C.S.; Li, N. Time-selective chemoprevention of vitamin E and selenium on esophageal carcinogenesis in rats: The possible role of nuclear factor kappaB signaling pathway. Int. J. Cancer 2012, 131, 1517–1527. [Google Scholar]
  34. Korostishevsky, M.; Steves, C.J.; Malkin, I.; Spector, T.; Williams, F.M.; Livshits, G. Genomics and metabolomics of muscular mass in a community-based sample of UK females. Eur. J. Hum. Genet. 2016, 24, 277–283. [Google Scholar] [CrossRef] [Green Version]
  35. Li, Y.P.; Zhang, L.R.; Jia, M.; Hu, X.J. CYP3AP1*3 allele is associated with lipid-lowering efficacy of simvastatin and atorvastatin in Chinese women. J. Clin. Pharmacol. 2011, 51, 181–188. [Google Scholar] [CrossRef]
  36. Baris, D.; Karagas, M.R.; Koutros, S.; Colt, J.S.; Johnson, A.; Schwenn, M.; Fischer, A.H.; Figueroa, J.D.; Berndt, S.I.; Han, S.; et al. Nonsteroidal anti-inflammatory drugs and other analgesic use and bladder cancer in northern New England. Int. J. Cancer. 2013, 132, 162–173. [Google Scholar] [CrossRef] [Green Version]
  37. Brigelius-Flohe, R. Vitamin E and drug metabolism. Biochem. Biophys. Res. Commun. 2003, 305, 737–740. [Google Scholar] [CrossRef]
  38. Chen, Y.; Tang, Y.; Wang, M.T.; Zeng, S.; Nie, D. Human pregnane X receptor and resistance to chemotherapy in prostate cancer. Cancer Res. 2007, 67, 10361–10367. [Google Scholar] [CrossRef] [Green Version]
  39. Fredericks, W.J.; Sepulveda, J.; Lai, P.; Tomaszewski, J.E.; Lin, M.F.; McGarvey, T.; Rauscher, F.J., 3rd; Malkowicz, S.B. The tumor suppressor TERE1 (UBIAD1) prenyltransferase regulates the elevated cholesterol phenotype in castration resistant prostate cancer by controlling a program of ligand dependent SXR target genes. Oncotarget 2013, 4, 1075–1092. [Google Scholar] [CrossRef] [Green Version]
  40. Pondugula, S.R.; Pavek, P.; Mani, S. Pregnane X Receptor and Cancer: Context-Specificity is Key. Nucl. Recept. Res. 2016, 3, 101198. [Google Scholar] [CrossRef] [Green Version]
  41. Carr, D.F.; O’Meara, H.; Jorgensen, A.L.; Campbell, J.; Hobbs, M.; McCann, G.; van Staa, T.; Pirmohamed, M. SLCO1B1 genetic variant associated with statin-induced myopathy: A proof-of-concept study using the clinical practice research datalink. Clin. Pharmacol. Ther. 2013, 94, 695–701. [Google Scholar] [CrossRef] [PubMed]
  42. Craig, E.L.; Stopsack, K.H.; Evergren, E.; Penn, L.Z.; Freedland, S.J.; Hamilton, R.J.; Allott, E.H. Statins and prostate cancer-hype or hope? The epidemiological perspective. Prostate Cancer Prostatic Dis. 2022, 25, 641–649. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, Y.; Ni, J.; Messing, E.M.; Chang, E.; Yang, C.R.; Yeh, S. Vitamin E succinate inhibits the function of androgen receptor and the expression of prostate-specific antigen in prostate cancer cells. Proc. Natl. Acad. Sci. USA 2002, 99, 7408–7413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mondul, A.M.; Rohrmann, S.; Menke, A.; Feinleib, M.; Nelson, W.G.; Platz, E.A.; Albanes, D. Association of serum alpha-tocopherol with sex steroid hormones and interactions with smoking: Implications for prostate cancer risk. Cancer Causes Control. 2011, 22, 827–836. [Google Scholar] [CrossRef] [Green Version]
  45. Kawasaki, B.T.; Hurt, E.M.; Kalathur, M.; Duhagon, M.A.; Milner, J.A.; Kim, Y.S.; Farrar, W.L. Effects of the sesquiterpene lactone parthenolide on prostate tumor-initiating cells: An integrated molecular profiling approach. Prostate 2009, 69, 827–837. [Google Scholar] [CrossRef] [Green Version]
  46. Kim, E.J.; Lim, S.S.; Park, S.Y.; Shin, H.K.; Kim, J.S.; Park, J.H. Apoptosis of DU145 human prostate cancer cells induced by dehydrocostus lactone isolated from the root of Saussurea lappa. Food Chem. Toxicol. 2008, 46, 3651–3658. [Google Scholar] [CrossRef]
  47. Sanderson, T.; Renaud, M.; Scholten, D.; Nijmeijer, S.; van den Berg, M.; Cowell, S.; Guns, E.; Nelson, C.; Mutarapat, T.; Ruchirawat, S. Effects of lactone derivatives on aromatase (CYP19) activity in H295R human adrenocortical and (anti)androgenicity in transfected LNCaP human prostate cancer cells. Eur. J. Pharmacol. 2008, 593, 92–98. [Google Scholar] [CrossRef]
Figure 1. Participant flowchart.
Figure 1. Participant flowchart.
Nutrients 15 02836 g001
Table 1. Participant baseline characteristics by daily supplemental vitamin E intake in the PLCO Study 1.
Table 1. Participant baseline characteristics by daily supplemental vitamin E intake in the PLCO Study 1.
All ParticipantsMenWomen
0 IU4.3–<400 IU IU≥400 IU0 IU4.3–<400 IU IU≥400 IU0 IU4.3–<400 IU IU≥400 IU
n16558728821002446389653426493
Age, years63.4 ± 5.063.1 ± 5.363.0 ± 5.263.6 ± 5.062.8 ± 5.363.0 ± 5.163.3 ± 5.163.3 ± 5.462.9 ± 5.2
Race, %
White, non-Hispanic79.886.685.772.881.677.690.591.892.1
Black, non-Hispanic16.78.99.124.213.517.05.24.22.8
Other races combined3.64.55.23.14.95.44.34.05.1
Height, cm172.1 ± 9.9171.0 ± 10.1169.9 ± 9.8177.9 ± 7.1178.2 ± 7.0178.1 ± 7.0163.2 ± 6.3163.5 ± 6.9163.5 ± 6.4
Weight, kg82.9 ± 16.779.9 ± 15.578.5 ± 16.588.7 ± 15.286.6 ± 14.088.0 ± 14.874.0 ± 15.072.8 ± 13.871.0 ± 13.7
Body mass index, kg/m227.9 ± 4.827.3 ± 4.627.1 ± 4.728.0 ± 4.527.3 ± 4.027.7 ± 4.127.8 ± 5.327.3 ± 5.226.6 ± 5.1
Tobacco smoking
status, %
Never smoker44.547.747.435.736.835.557.959.256.8
Former smoker9.76.27.410.96.58.77.85.96.3
Current smoker45.946.145.253.456.755.834.335.036.9
Multivitamin use, %095.874.3095.772.8095.875.5
Abbreviations: PLCO = Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. 1 Mean ± standard deviation (all values except as noted).
Table 2. Metabolites associated with daily supplemental vitamin E intake in the PLCO Study (FDR-adjusted p-value <0.1) 1,2.
Table 2. Metabolites associated with daily supplemental vitamin E intake in the PLCO Study (FDR-adjusted p-value <0.1) 1,2.
4.3–<400 IU IU≥400 IU
MetaboliteChemical ClassSub-PathwayF-ValueFDR-
Adjusted
p-Value
Effect Size (β)p-ValueEffect Size
(β)
p-Value
Alpha-CEHCCofactors and
vitamins
Tocopherol metabolism364.47<0.0010.62<0.0011.80<0.001
Alpha-CEHC-glucuronideCofactors and
vitamins
Tocopherol metabolism338.54<0.0010.66<0.0011.90<0.001
Alpha-TocopherolCofactors and
vitamins
Tocopherol metabolism318.33<0.0010.18<0.0010.44<0.001
Alpha-CEHC sulfateCofactors and
vitamins
Tocopherol metabolism214.10<0.0011.09<0.0012.38<0.001
Pantothenic acidCofactors and
vitamins
Pantothenate and
CoA metabolism
53.15<0.0010.17<0.0010.29<0.001
L-Threonic acidCofactors and
vitamins
Ascorbate and aldarate metabolism43.46<0.0010.110.0020.27<0.001
4-Pyridoxic acidCofactors and
vitamins
Vitamin B6 metabolism43.16<0.0010.140.0210.43<0.001
Delta-TocopherolCofactors and
vitamins
Tocopherol metabolism34.05<0.001−0.200.008−0.49<0.001
Glyceric acidCarbohydrateGlycolysis, gluconeogenesis, pyruvate metabolism30.86<0.0010.08<0.0010.13<0.001
Oxalic acidCarbohydrateGlyoxylate and
dicarboxylate metabolism
28.66<0.0010.100.0130.24<0.001
PyridoxalCofactors and
vitamins
Vitamin B6 metabolism22.79<0.0010.030.790.45<0.001
Hydroxypropanedioic acidXenobioticsFood component/plant16.12<0.0010.120.0020.18<0.001
N1-Methyl-2-pyridone-5-carboxamideCofactors and
vitamins
Nicotinate and
nicotinamide metabolism
15.21<0.0010.080.0410.18<0.001
Glycocholenate sulfateLipidBile acid metabolism15.16<0.001−0.100.001−0.15<0.001
Beta-TocopherolCofactors and
vitamins
Tocopherol metabolism14.46<0.001−0.260.020−0.49<0.001
Hydroxy-carboxy-4-methyl-5-propyl-2-furanpropionic acid (CMPF)LipidFatty acid, dicarboxylate14.36<0.001−0.120.0430.130.013
N1-Methyl-4-pyridone-3-carboxamideCofactors and
vitamins
Nicotinate and
nicotinamide metabolism
11.370.0010.130.0040.18<0.001
Isovaleric acidLipidFatty acid metabolism10.980.0010.000.8820.09<0.001
4-Androsten-3alpha,17alpha-diol monosulfate (2) or androstenediol (3alpha, 17alpha) monsulfate (2)LipidAndrogenic steroids10.340.002−0.150.065−0.180.012
C22 lactone sulfate (X_12063)Partially
characterized
Partially characterized8.990.007−0.040.500−0.17<0.001
DecanoylcarnitineLipidCarnitine metabolism8.280.0130.010.885−0.110.003
Docosahexaenoic acidLipidEssential fatty acid8.190.014−0.040.2280.060.031
Androsterone glucuronideLipidAndrogenic steroids8.080.016−0.190.008−0.26<0.001
PhosphateEnergyOxidative phosphorylation8.020.0160.06<0.0010.06<0.001
L-OctanoylcarnitineLipidCarnitine metabolism7.860.0170.010.806−0.100.004
N-Stearoyltaurine or N-Stearoyl taurineLipidEndocannabinoid7.860.0180.070.2440.010.863
Sphingomyelin (d17:1/14:0, d16:1/15:0)LipidSphingomyelins7.670.020−0.150.014−0.100.063
Eicosapentaenoic acidLipidEssential fatty acid7.550.0210.010.7330.100.002
5alpha-Androstan-3alpha,17alpha-diol disulfateLipidSterol/steroid7.500.0210.030.733−0.050.492
S-AllylcysteineXenobioticsFood component/plant7.290.026−0.190.1110.170.100
3-Carboxy-4-methyl-5-propyl-2-furanpropionic acid (CMPF)LipidFatty acid, dicarboxylate7.250.026−0.080.2100.110.054
L-Aspartic acidAmino acidAlanine and aspartate
metabolism
7.190.0270.010.663−0.050.010
Monoglyceride (18:1(9Z)/0:0/0:0)LipidMonoacylglycerol7.180.0270.030.5940.010.760
L-UrobilinCofactors and
vitamins
Hemoglobin and
porphyrin metabolism
7.050.030−0.020.799−0.240.003
L-GlutamineAmino acidGlutamate metabolism7.020.030−0.020.032−0.03<0.001
Sphingomyelin (d17:1/16:0, d18:1/15:0, d16:1/17:0) or sphingomyelin (d18:1/15:0, d16:1/17:0)LipidSphingomyelins6.900.034−0.080.001−0.09<0.001
Tauro-b-muricholic acidLipidPrimary bile acid metabolism6.820.0340.180.0640.30<0.001
Lactosylceramide (d18:1/22:0)LipidLactosylceramides (LCER)6.870.034−0.19<0.001−0.110.008
2-Methoxyacetaminophen glucuronideXenobioticsDrug6.800.0340.420.0040.220.090
9-HexadecenoylcarnitineLipidFatty acid metabolism (acyl carnitine, monounsaturated6.750.0360.040.3930.020.585
Sphingomyelin (d17:2/16:0, d18:2/15:0)LipidSphingomyelins6.780.036−0.170.004−0.120.013
Phosphatidylcholine (P-16:0/18:2)LipidPlasmalogen6.670.038−0.110.001−0.090.002
Homo-L-ArginineAmino acidUrea cycle; arginine and proline metabolism6.540.042−0.110.1050.080.147
PregnanediolLipidSterol/steroid6.470.044−0.070.126−0.140.001
5alpha-Androstan-3alpha,17beta-diol disulfateLipidSterol/steroid6.460.044−0.110.192−0.180.013
(R)C(S)S-AlliinXenobioticsFood component/plant6.270.051−0.170.1770.190.096
OxypurinolXenobioticsDrug—metabolic6.140.057−0.660.002−0.160.371
L-SerineAmino acidGlycine, serine and threonine metabolism6.130.057−0.040.010−0.040.001
O-DesmethyltramadolXenobioticsDrug—analgesics, anesthetics6.040.063−0.130.008−0.070.137
Dimethyl sulfoneXenobioticsChemical5.890.0700.060.4260.200.002
O-PhosphoethanolamineLipidPhospholipid metabolism5.880.070−0.100.2390.130.090
cis-4-DecenoateLipidMedium-chain fatty acid5.870.070−0.030.668−0.110.069
L-MethionineAmino acidCysteine, methionine, SAM, taurine metabolism5.860.0700.010.6620.000.807
3b,17a-Dihydroxy-5a-androstaneLipidAndrogenic steroids5.840.070−0.150.063−0.200.005
HydrochlorothiazideXenobioticsDrug5.680.0780.240.0550.370.001
Monoglyceride (22:6(4Z,7Z,10Z,13Z,16Z,19Z)/0:0/0:0)LipidMonoacylglycerol5.630.0820.000.9940.090.075
Sphingomyelin (d18:1/22:0)LipidSphingomyelins5.620.082−0.070.008−0.080.001
Dehydroepiandrosterone (DHEA) sulfateLipidSterol/steroid5.560.083−0.090.034−0.130.001
L-CysteineAmino acidCysteine, methionine, SAM, taurine metabolism5.560.083−0.020.4060.030.055
Etiocholanolone glucuronideLipidAndrogenic steroids5.460.089−0.170.026−0.220.001
4-Androsten-3alpha,17alpha-diol monosulfate (3) or androstenediol (3alpha, 17alpha) monsulfate (3)LipidAndrogenic steroids5.460.089−0.100.050−0.140.003
LidocaineXenobioticsDrug5.420.0910.180.0010.120.017
ErgothioneineXenobioticsFood component/plant5.380.091−0.050.2240.060.118
Gamma-GlutamylalaninePeptideGamma-glutamyl5.370.0910.030.3000.020.353
PioglitazoneXenobioticsDrug—metabolic5.370.0910.600.0010.380.018
Phenol sulfateAmino acidPhenylalanine and tyrosine metabolism5.310.0920.020.742−0.090.024
5′-MethylthioadenosineAmino acidPolyamine metabolism5.250.0960.140.0010.080.033
KetopioglitazoneXenobioticsDrug—metabolic5.230.0980.200.0010.120.020
Hydroxypioglitazone (M-IV)XenobioticsDrug—metabolic5.220.0980.120.0010.070.021
Phosphatidylcholine (18:2(9Z,12Z))LipidLysolipid5.180.099−0.070.001−0.030.055
Abbreviations: PLCO, Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial; CEHC, carboxyethylhydroxychroman. 1 Estimates and p-values were derived using ANCOVA with the 0 IU/day as the reference group. Metabolites were ordered by descending statistical significance of FDR-adjusted p-values and alphabetically if they had the same p-value. 2 Adjusted for age, sex, body weight, smoking status, and multivitamin use.
Table 3. Metabolites associated with daily supplemental vitamin E intake according to sex in the PLCO Study (FDR-adjusted p-value <0.1) 1,2.
Table 3. Metabolites associated with daily supplemental vitamin E intake according to sex in the PLCO Study (FDR-adjusted p-value <0.1) 1,2.
Men (n = 1837)Women (n = 1572)
4.3–<400 IU≥400 IU 4.3–<400 IU≥400 IU
MetaboliteFDR-
Adjusted
p-Value
Effect
Size
(β)
p-ValueEffect
Size
(β)
p-ValueFDR-
Adjusted
p-Value
Effect Size
(β)
p-ValueEffect
Size
(β)
p-Value
Alpha-CEHC sulfate<0.0010.94<0.00110−18<0.001<0.0011.26<0.0012.69<0.001
Alpha-CEHC glucuronide<0.0010.47<0.0011.50<0.001<0.0010.90<0.0012.29<0.001
Alpha-Tocopherol<0.0010.16<0.0010.43<0.001<0.0010.19<0.0010.45<0.001
Alpha-CEHC<0.0010.48<0.0011.37<0.001<0.0010.78<0.0012.15<0.001
Pantothenic acid<0.0010.25<0.0010.34<0.001<0.0010.100.0340.24<0.001
4-Pyridoxic acid<0.0010.180.0180.42<0.001<0.0010.120.2220.45<0.001
L-Threonic acid<0.0010.150.0070.29<0.001<0.0010.070.1180.24<0.001
Glyceric acid<0.0010.100.0010.15<0.001<0.0010.060.0250.11<0.001
Delta-Tocopherol<0.001−0.190.176−0.62<0.001<0.001−0.210.019−0.45<0.001
Oxalic acid<0.0010.110.0550.26<0.001<0.0010.070.1780.21<0.001
N1-Methyl-4-pyridone-3-carboxamide0.0030.110.0270.20<0.0010.0740.040.4680.150.002
Pyridoxal0.010−0.010.9160.290.002<0.0010.120.4720.61<0.001
Glycocholenate sulfate0.052−0.080.055−0.13<0.0010.009−0.130.004−0.17<0.001
C22 lactone sulfate (X_12063) 30.5950.020.814−0.080.1880.046−0.120.186−0.260.001
5-Androstenediol 30.305−0.050.253−0.110.0110.057−0.150.023−0.21<0.001
Androsterone glucuronide 30.531−0.060.343−0.110.0480.091−0.320.010−0.370.001
Abbreviations: FDR, false discovery rate; CEHC, carboxyethylhydroxychroman. 1 Beta-estimates and p-values were derived using ANCOVA with the 0 IU intake/day as the reference group. 2 Adjusted for age, sex, body weight, smoking status, and multivitamin use. 3 Metabolite significant in women (FDR < 0.1) but not in men.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lim, J.; Hong, H.G.; Weinstein, S.J.; Playdon, M.C.; Cross, A.J.; Stolzenberg-Solomon, R.; Freedman, N.D.; Huang, J.; Albanes, D. Metabolomic Analysis of Vitamin E Supplement Use in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Nutrients 2023, 15, 2836. https://doi.org/10.3390/nu15132836

AMA Style

Lim J, Hong HG, Weinstein SJ, Playdon MC, Cross AJ, Stolzenberg-Solomon R, Freedman ND, Huang J, Albanes D. Metabolomic Analysis of Vitamin E Supplement Use in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Nutrients. 2023; 15(13):2836. https://doi.org/10.3390/nu15132836

Chicago/Turabian Style

Lim, Jungeun, Hyokyoung G. Hong, Stephanie J. Weinstein, Mary C. Playdon, Amanda J. Cross, Rachael Stolzenberg-Solomon, Neal D. Freedman, Jiaqi Huang, and Demetrius Albanes. 2023. "Metabolomic Analysis of Vitamin E Supplement Use in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial" Nutrients 15, no. 13: 2836. https://doi.org/10.3390/nu15132836

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop