Search for Early Pancreatic Cancer Blood Biomarkers in Five European Prospective Population Biobanks Using Metabolomics

Jesse Fest, Lisanne S Vijfhuizen, Jelle J Goeman, Olga Veth, Anni Joensuu, Markus Perola, Satu Männistö, Eivind Ness-Jensen, Kristian Hveem, Toomas Haller, Neeme Tonisson, Kairit Mikkel, Andres Metspalu, Cornelia M van Duijn, Arfan Ikram, Bruno H Stricker, Rikje Ruiter, Casper H J van Eijck, Gert-Jan B van Ommen, Peter A C ʼt Hoen, Jesse Fest, Lisanne S Vijfhuizen, Jelle J Goeman, Olga Veth, Anni Joensuu, Markus Perola, Satu Männistö, Eivind Ness-Jensen, Kristian Hveem, Toomas Haller, Neeme Tonisson, Kairit Mikkel, Andres Metspalu, Cornelia M van Duijn, Arfan Ikram, Bruno H Stricker, Rikje Ruiter, Casper H J van Eijck, Gert-Jan B van Ommen, Peter A C ʼt Hoen

Abstract

Most patients with pancreatic cancer present with advanced disease and die within the first year after diagnosis. Predictive biomarkers that signal the presence of pancreatic cancer in an early stage are desperately needed. We aimed to identify new and validate previously found plasma metabolomic biomarkers associated with early stages of pancreatic cancer. Prediagnostic blood samples from individuals who were to receive a diagnosis of pancreatic cancer between 1 month and 17 years after sampling (N = 356) and age- and sex-matched controls (N = 887) were collected from five large population cohorts (HUNT2, HUNT3, FINRISK, Estonian Biobank, Rotterdam Study). We applied proton nuclear magnetic resonance-based metabolomics on the Nightingale platform. Logistic regression identified two interesting hits: glutamine (P = 0.011) and histidine (P = 0.012), with Westfall-Young family-wise error rate adjusted P values of 0.43 for both. Stratification in quintiles showed a 1.5-fold elevated risk for the lowest 20% of glutamine and a 2.2-fold increased risk for the lowest 20% of histidine. Stratification by time to diagnosis suggested glutamine to be involved in an earlier process (2 to 5 years before diagnosis), and histidine in a process closer to the actual onset (<2 years). Our data did not support the branched-chain amino acids identified earlier in several US cohorts as potential biomarkers for pancreatic cancer. Thus, although we identified glutamine and histidine as potential biomarkers of biological interest, our results imply that a study at this scale does not yield metabolomic biomarkers with sufficient predictive value to be clinically useful per se as prognostic biomarkers.

Conflict of interest statement

Restrictions apply to the availability of data generated or analyzed during this study to preserve patient confidentiality or because they were used under license. The corresponding author will on request detail the restrictions and any conditions under which access to some data may be provided.

Copyright © 2019 Endocrine Society.

Figures

Figure 1.
Figure 1.
Schematic overview of the sample set used for data analysis and the different data analysis approaches performed in the current study. aAny individual containing missing values in metabolomics measurements or phenotypical information were assumed to be missing at random and were removed from the data set. bAny individual containing missing values in phenotypical information were removed from the data set. PC, pancreatic cancer.
Figure 2.
Figure 2.
Concentrations (logarithmic scale) of (A–D) glutamine and (E–H) histidine in the blood circulation in controls and cases, that is, those individuals who developed pancreatic cancer within a time window after blood sampling. (A and E) Distribution of the concentrations of controls (light blue) and cases (dark blue) in the different cohorts analyzed (EGCUT, FR, HUNT2, HUNT3, RS). (B and F) Distribution of concentrations in nondiabetics (light blue) and individuals diagnosed with T2DM (dark blue). (C and G) Distribution of concentrations in controls and cases sampled within 2 y before diagnosis, between 2 and 5 y before diagnosis, and >5 y before diagnosis. (D and H) Distribution of concentrations in nonfasting individuals (light blue), individuals who had a meal between 4 and 8 h before blood draw (dark blue), and fasting individuals (green, last meal was >8 h before blood draw). Box plots reflect the distribution of the concentrations in individual samples, including the middle quartiles (25th to 75th percentile of the data points are in the boxes); the horizontal band; the median value; the lower whiskers representing the data points up to 1.5 × the interquartile range (IQR) below the 25th percentile; the upper whiskers representing the data points up to 1.5 × IQR above the 75th percentile; the data points outside these ranges plotted as individual data points.
Figure 3.
Figure 3.
Forest plots from random effects meta-analysis across different cohorts for (A) glutamine and (B) histidine. The meta-analysis was performed on the β coefficients and SD from the logistic regressions run for each cohort separately. In the logistic regression, pancreatic cancer status was modeled as a function of log-transformed and standardized metabolite concentration, sex, age, BMI, smoking status, T2DM, and fasting status. Shown are the estimated effect size, the SE on this estimate, the estimated OR and the CI on this ratio, the weight of the individual cohort on the calculation of the final estimate, the heterogeneity measure (modeling differences between cohorts), and the unadjusted and Bonferroni–Holm-corrected P values for the respective metabolites.
Figure 4.
Figure 4.
Receiver operator curves for classification of pancreatic cancer cases (sampled up to 5 y before diagnosis) and controls for (A) training set (70% of all individuals) and (B) performance testing set (30% of all individuals unseen during the variable selection). In red, the null model is shown in which only the clinical covariates (sex, age, BMI, smoking status, T2DM, and fasting status) were included in the regression. In blue, the alternative model is shown where the metabolites selected by the LASSO regression were included in addition to the clinical covariates. The AUCs are indicated, as well as the specificity (1 − false-positive rate) at 70% sensitivity.

References

    1. De Angelis R, Sant M, Coleman MP, Francisci S, Baili P, Pierannunzio D, Trama A, Visser O, Brenner H, Ardanaz E, Bielska-Lasota M, Engholm G, Nennecke A, Siesling S, Berrino F, Capocaccia R; EUROCARE-5 Working Group. Cancer survival in Europe 1999–2007 by country and age: results of EUROCARE-5—a population-based study. Lancet Oncol. 2014;15(1):23–34.
    1. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–2921.
    1. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378(9791):607–620.
    1. Fest J, Ruiter R, van Rooij FJ, van der Geest LG, Lemmens VE, Ikram MA, Coebergh JW, Stricker BH, van Eijck CH. Underestimation of pancreatic cancer in the national cancer registry—reconsidering the incidence and survival rates. Eur J Cancer. 2017;72:186–191.
    1. Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA, Velculescu VE, Kinzler KW, Vogelstein B, Iacobuzio-Donahue CA. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–1117.
    1. Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, Gavin A, Visser O, Bray F. Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer. 2018;103:356–387.
    1. Ilic M, Ilic I. Epidemiology of pancreatic cancer. World J Gastroenterol. 2016;22(44):9694–9705.
    1. De Bruijn KM, Ruiter R, de Keyser CE, Hofman A, Stricker BH, van Eijck CH. Detection bias may be the main cause of increased cancer incidence among diabetics: results from the Rotterdam Study. Eur J Cancer. 2014;50(14):2449–2455.
    1. He X, Zhong J, Wang S, Zhou Y, Wang L, Zhang Y, Yuan Y. Serum metabolomics differentiating pancreatic cancer from new-onset diabetes. Oncotarget. 2017;8(17):29116–29124.
    1. Kobayashi T, Nishiumi S, Ikeda A, Yoshie T, Sakai A, Matsubara A, Izumi Y, Tsumura H, Tsuda M, Nishisaki H, Hayashi N, Kawano S, Fujiwara Y, Minami H, Takenawa T, Azuma T, Yoshida M. A novel serum metabolomics-based diagnostic approach to pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2013;22(4):571–579.
    1. Mayers JR, Wu C, Clish CB, Kraft P, Torrence ME, Fiske BP, Yuan C, Bao Y, Townsend MK, Tworoger SS, Davidson SM, Papagiannakopoulos T, Yang A, Dayton TL, Ogino S, Stampfer MJ, Giovannucci EL, Qian ZR, Rubinson DA, Ma J, Sesso HD, Gaziano JM, Cochrane BB, Liu S, Wactawski-Wende J, Manson JE, Pollak MN, Kimmelman AC, Souza A, Pierce K, Wang TJ, Gerszten RE, Fuchs CS, Vander Heiden MG, Wolpin BM. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med. 2014;20(10):1193–1198.
    1. Mehta KY, Wu HJ, Menon SS, Fallah Y, Zhong X, Rizk N, Unger K, Mapstone M, Fiandaca MS, Federoff HJ, Cheema AK. Metabolomic biomarkers of pancreatic cancer: a meta-analysis study. Oncotarget. 2017;8(40):68899–68915.
    1. Xie G, Lu L, Qiu Y, Ni Q, Zhang W, Gao YT, Risch HA, Yu H, Jia W. Plasma metabolite biomarkers for the detection of pancreatic cancer. J Proteome Res. 2015;14(2):1195–1202.
    1. OuYang D, Xu J, Huang H, Chen Z. Metabolomic profiling of serum from human pancreatic cancer patients using 1H NMR spectroscopy and principal component analysis. Appl Biochem Biotechnol. 2011;165(1):148–154.
    1. Fukutake N, Ueno M, Hiraoka N, Shimada K, Shiraishi K, Saruki N, Ito T, Yamakado M, Ono N, Imaizumi A, Kikuchi S, Yamamoto H, Katayama K. A novel multivariate index for pancreatic cancer detection based on the plasma free amino acid profile. PLoS One. 2015;10(7):e0132223.
    1. Zhang L, Jin H, Guo X, Yang Z, Zhao L, Tang S, Mo P, Wu K, Nie Y, Pan Y, Fan D. Distinguishing pancreatic cancer from chronic pancreatitis and healthy individuals by 1H nuclear magnetic resonance-based metabonomic profiles. Clin Biochem. 2012;45(13–14):1064–1069.
    1. Leitsalu L, Haller T, Esko T, Tammesoo ML, Alavere H, Snieder H, Perola M, Ng PC, Mägi R, Milani L, Fischer K, Metspalu A. Cohort profile: Estonian Biobank of the Estonian Genome Center, University of Tartu. Int J Epidemiol. 2015;44(4):1137–1147.
    1. Pajunen P, Vartiainen E, Männistö S, Jousilahti P, Laatikainen T, Peltonen M. Intra-individual changes in body weight in population-based cohorts during four decades: the Finnish FINRISK study. Eur J Public Health. 2012;22(1):107–112.
    1. Borodulin K, Tolonen H, Jousilahti P, Jula A, Juolevi A, Koskinen S, Kuulasmaa K, Laatikainen T, Männistö S, Peltonen M, Perola M, Puska P, Salomaa V, Sundvall J, Virtanen SM, Vartiainen E. Cohort profile: the National FINRISK Study. Int J Epidemiol. 2018;47(3):696–696i.
    1. Krokstad S, Langhammer A, Hveem K, Holmen TL, Midthjell K, Stene TR, Bratberg G, Heggland J, Holmen J. Cohort profile: the HUNT Study, Norway. Int J Epidemiol. 2013;42(4):968–977.
    1. Hofman A, Grobbee DE, de Jong PT, van den Ouweland FA. Determinants of disease and disability in the elderly: the Rotterdam Elderly Study. Eur J Epidemiol. 1991;7(4):403–422.
    1. Ikram MA, Brusselle GG, Murad SD, van Duijn CM, Franco OH, Goedegebure A, Klaver CCW, Nijsten TEC, Peeters RP, Stricker BH, Tiemeier H, Uitterlinden AG, Vernooij MW, Hofman A. The Rotterdam Study: 2018 update on objectives, design and main results. Eur J Epidemiol. 2017;32(9):807–850.
    1. Bernards N, Creemers GJ, Huysentruyt CJ, de Hingh IH, van der Schelling GP, de Bruïne AP, Lemmens VE. The relevance of pathological verification in suspected pancreatic cancer. Cancer Epidemiol. 2015;39(2):250–255.
    1. Carpelan-Holmström M, Nordling S, Pukkala E, Sankila R, Lüttges J, Klöppel G, Haglund C. Does anyone survive pancreatic ductal adenocarcinoma? A nationwide study re-evaluating the data of the Finnish Cancer Registry. Gut. 2005;54(3):385–387.
    1. Lepage C, Capocaccia R, Hackl M, Lemmens V, Molina E, Pierannunzio D, Sant M, Trama A, Faivre J; EUROCARE-5 Working Group. Survival in patients with primary liver cancer, gallbladder and extrahepatic biliary tract cancer and pancreatic cancer in Europe 1999–2007: results of EUROCARE-5. Eur J Cancer. 2015;51(15):2169–2178.
    1. Soininen P, Kangas AJ, Würtz P, Suna T, Ala-Korpela M. Quantitative serum nuclear magnetic resonance metabolomics in cardiovascular epidemiology and genetics. Circ Cardiovasc Genet. 2015;8(1):192–206.
    1. Goek ON, Döring A, Gieger C, Heier M, Koenig W, Prehn C, Römisch-Margl W, Wang-Sattler R, Illig T, Suhre K, Sekula P, Zhai G, Adamski J, Köttgen A, Meisinger C. Serum metabolite concentrations and decreased GFR in the general population. Am J Kidney Dis. 2012;60(2):197–206.
    1. Westfall PH, Young SS. Resampling-Based Multiple Testing: Examples and Methods for p-Value Adjustment.New York, NY: John Wiley & Sons; 1993.
    1. Schwarzer G, Carpenter JR, Rücker G. Meta-Analysis with R. Cham, Switzerland: Springer International Publishing; 2015.
    1. Goeman JJ. L1 penalized estimation in the Cox proportional hazards model. Biom J. 2010;52(1):70–84.
    1. Fest J, Vijfhuizen LS, Goeman JJ, Veth O, Joensuu A, Perola M, Männistö S, Ness-Jensen E, Hveem K, Haller T, Tonisson N, Mikkel K, Metspalu A, van Duijn CM, Ikram A, Stricker BH, Ruiter R, van Eijck CH, van Ommen GJB, ʼt Hoen PAC. Data from: Search for early pancreatic cancer blood biomarkers in five European prospective population biobanks using metabolomics GitHub 2019 Deposited 27 February 2019 .
    1. Fest J, Vijfhuizen LS, Goeman JJ, Veth O, Joensuu A, Perola M, Männistö S, Ness-Jensen E, Hveem K, Haller T, Tonisson N, Mikkel K, Metspalu A, van Duijn CM, Ikram A, Stricker BH, Ruiter R, van Eijck CH, van Ommen GJB, ʼt Hoen PAC. Data from: Search for early pancreatic cancer blood biomarkers in five European prospective population biobanks using metabolomics GitHub 2019 Deposited 18 March 2019.
    1. Roux C, Riganti C, Borgogno SF, Curto R, Curcio C, Catanzaro V, Digilio G, Padovan S, Puccinelli MP, Isabello M, Aime S, Cappello P, Novelli F. Endogenous glutamine decrease is associated with pancreatic cancer progression. Oncotarget. 2017;8(56):95361–95376.
    1. Bi X, Henry CJ. Plasma-free amino acid profiles are predictors of cancer and diabetes development. Nutr Diabetes. 2017;7(3):e249.
    1. Gu Y, Chen T, Fu S, Sun X, Wang L, Wang J, Lu Y, Ding S, Ruan G, Teng L, Wang M. Perioperative dynamics and significance of amino acid profiles in patients with cancer. J Transl Med. 2015;13(1):35.
    1. Miyagi Y, Higashiyama M, Gochi A, Akaike M, Ishikawa T, Miura T, Saruki N, Bando E, Kimura H, Imamura F, Moriyama M, Ikeda I, Chiba A, Oshita F, Imaizumi A, Yamamoto H, Miyano H, Horimoto K, Tochikubo O, Mitsushima T, Yamakado M, Okamoto N. Plasma free amino acid profiling of five types of cancer patients and its application for early detection. PLoS One. 2011;6(9):e24143.
    1. Kanarek N, Keys HR, Cantor JR, Lewis CA, Chan SH, Kunchok T, Abu-Remaileh M, Freinkman E, Schweitzer LD, Sabatini DM. Histidine catabolism is a major determinant of methotrexate sensitivity. Nature. 2018;559(7715):632–636.

Source: PubMed

3
구독하다