Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development

Jared R Mayers, Chen Wu, Clary B Clish, Peter Kraft, Margaret E Torrence, Brian P Fiske, Chen Yuan, Ying Bao, Mary K Townsend, Shelley S Tworoger, Shawn M Davidson, Thales Papagiannakopoulos, Annan Yang, Talya L Dayton, Shuji Ogino, Meir J Stampfer, Edward L Giovannucci, Zhi Rong Qian, Douglas A Rubinson, Jing Ma, Howard D Sesso, John Michael Gaziano, Barbara B Cochrane, Simin Liu, Jean Wactawski-Wende, JoAnn E Manson, Michael N Pollak, Alec C Kimmelman, Amanda Souza, Kerry Pierce, Thomas J Wang, Robert E Gerszten, Charles S Fuchs, Matthew G Vander Heiden, Brian M Wolpin, Jared R Mayers, Chen Wu, Clary B Clish, Peter Kraft, Margaret E Torrence, Brian P Fiske, Chen Yuan, Ying Bao, Mary K Townsend, Shelley S Tworoger, Shawn M Davidson, Thales Papagiannakopoulos, Annan Yang, Talya L Dayton, Shuji Ogino, Meir J Stampfer, Edward L Giovannucci, Zhi Rong Qian, Douglas A Rubinson, Jing Ma, Howard D Sesso, John Michael Gaziano, Barbara B Cochrane, Simin Liu, Jean Wactawski-Wende, JoAnn E Manson, Michael N Pollak, Alec C Kimmelman, Amanda Souza, Kerry Pierce, Thomas J Wang, Robert E Gerszten, Charles S Fuchs, Matthew G Vander Heiden, Brian M Wolpin

Abstract

Most patients with pancreatic ductal adenocarcinoma (PDAC) are diagnosed with advanced disease and survive less than 12 months. PDAC has been linked with obesity and glucose intolerance, but whether changes in circulating metabolites are associated with early cancer progression is unknown. To better understand metabolic derangements associated with early disease, we profiled metabolites in prediagnostic plasma from individuals with pancreatic cancer (cases) and matched controls from four prospective cohort studies. We find that elevated plasma levels of branched-chain amino acids (BCAAs) are associated with a greater than twofold increased risk of future pancreatic cancer diagnosis. This elevated risk was independent of known predisposing factors, with the strongest association observed among subjects with samples collected 2 to 5 years before diagnosis, when occult disease is probably present. We show that plasma BCAAs are also elevated in mice with early-stage pancreatic cancers driven by mutant Kras expression but not in mice with Kras-driven tumors in other tissues, and that breakdown of tissue protein accounts for the increase in plasma BCAAs that accompanies early-stage disease. Together, these findings suggest that increased whole-body protein breakdown is an early event in development of PDAC.

Figures

Fig. 1. Plasma metabolites and risk of…
Fig. 1. Plasma metabolites and risk of future pancreatic cancer diagnosis
P–values of the log–transformed, continuous metabolite levels from human plasma comparing pancreatic cancer cases and controls in conditional logistic regression models conditioned on matching factors and adjusted for age at blood draw (years, continuous), fasting time (<4, 4–8, 8–12, ≥12 hours, missing) and race/ethnicity (White, Black, other, missing). The dashed green line indicates the statistically significant P–value threshold after Bonferroni correction for multiple–hypothesis testing, P–trend ≤0.0006 (0.05/83). The dashed blue line indicates P– trend of 0.05.
Fig. 2. Plasma BCAAs are elevated during…
Fig. 2. Plasma BCAAs are elevated during subclinical disease
a, Graph of odds ratio (95% confidence interval) for future pancreatic cancer diagnosis among cohort cases and matched controls comparing highest versus lowest quintiles of circulating BCAAs stratified by time from blood collection to the case's cancer diagnosis. Odds ratio (95% confidence interval) from conditional logistic regression models conditioned on matching factors and adjusted for age at blood draw (years, continuous), fasting time (<4, 4–8, 8–12, ≥12 hours, missing) and race/ethnicity (White, Black, other, missing). Red horizontal line marks an odds ratio of 1.0. b. Graph of total plasma BCAAs in KPC mice over time and in littermate controls. Each control data point is an average for one mouse over the course of the study (Supplementary Fig. 3b) and values for KPC mice living longer than 19 weeks are averaged for the >19 weeks time point. For weeks 15-17, n = 6 KPC and n = 9 control, t-test, P=0.001. c, H&E staining of pancreatic tissue obtained from KP–/–C mice and littermate controls at 3-4 weeks of age. Tissue are shown from a control mouse with histologically normal pancreas (left); a KP–/–C mouse with areas of PDAC adjacent to areas of normal pancreas (middle); and a KP–/–C mouse with areas of PDAC and pancreatic intraepithelial neoplasia (closed arrow heads) (right); scale bar = 50μM. d, Mean (±SEM) body weights at 3-4 weeks of age for KP–/–C mice and littermate controls (n = 7 KP–/–C, n = 11 control mice). e, Mean (±SEM) total plasma branched chain amino acid levels from KP–/–C mice and littermate controls at 3-4 weeks of age (n = 10 KP–/–C, n = 14 control mice, t-test, P=0.002). f, P–values for comparison of circulating amino acid levels in KP–/–C mice and littermate controls at 3-4 weeks of age, cys = cystine (n = 10 KP–/–C, n = 14 control mice), The dashed red line indicates P–value of 0.05. g, top, glucose tolerance test in KP–/–C mice and littermate controls at the time of weaning (n = 7 KP–/–C, n = 11 control mice) and bottom, insulin tolerance test in KP–/–C mice and littermate controls at four weeks of age (n = 7 KP–/–C, n = 15 control mice). h, Mean (±SEM) fasting plasma insulin levels from KP–/–C mice and littermate controls at four weeks of age (n = 7 KP–/–C, n = 11 control mice).
Fig. 3. BCAA elevations are derived from…
Fig. 3. BCAA elevations are derived from a long–term pool of amino acids
a, Plasma levels (mean ± SEM) of 13C–labeled leucine and valine normalized to food intake over time following a two–hour exposure to diets containing 13C–labeled leucine and valine. The time points correspond to the red arrowheads in the diagram. b, Diagram of experiment using labeled diets to investigate contributions to plasma BCAA levels from long–term pools. Two cohorts of mice were used for these experiments, one sacrificed in the fed state and a second sacrificed in the fasted state at the time points indicated by the red arrowheads. c, Fractional labeling of total amino acids in protein hydrolysate of gastrocnemius muscle from fasted KP–/–C mice and control littermates (n = 8 KP–/–C, n = 6 control). d, Fractional labeling of plasma amino acids in KP–/–C and control mice in the fed state (n = 3 KP–/–C, n = 4 control). e, The calculated contribution of the short– and long–term BCAA pools to the BCAAs present in plasma . f, Mean (±SEM) gastrocnemius weight (left panel, t-test, P=0.01), a predominantly fast–twitch muscle, and heart weight (right panel) normalized to body weight (n = 6 KP–/–C, n = 10 control).

References

    1. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378:607–620.
    1. Michaud DS, et al. Physical activity, obesity, height, and the risk of pancreatic cancer. JAMA. 2001;286:921–929.
    1. Stolzenberg-Solomon RZ, et al. Insulin, glucose, insulin resistance, and pancreatic cancer in male smokers. JAMA. 2005;294:2872–2878.
    1. Wolpin BM, et al. Hyperglycemia, Insulin Resistance, Impaired Pancreatic beta-Cell Function, and Risk of Pancreatic Cancer. J Natl Cancer Inst. 2013;105:1027–1035.
    1. Lynch SM, et al. Cigarette smoking and pancreatic cancer: a pooled analysis from the pancreatic cancer cohort consortium. Am J Epidemiol. 2009;170:403–413.
    1. Klein AP. Genetic susceptibility to pancreatic cancer. Mol Carcinog. 2012;51:14–24.
    1. Tan BH, Birdsell LA, Martin L, Baracos VE, Fearon KC. Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin Cancer Res. 2009;15:6973–6979.
    1. Claudino WM, Goncalves PH, di Leo A, Philip PA, Sarkar FH. Metabolomics in cancer: a bench-to-bedside intersection. Critical reviews in oncology/hematology. 2012;84:1–7.
    1. Sreekumar A, et al. Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature. 2009;457:910–914.
    1. Kobayashi T, et al. A novel serum metabolomics-based diagnostic approach to pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2013;22:571–579.
    1. Wang TJ, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–453.
    1. Brosnan JT, Brosnan ME. Branched-chain amino acids: enzyme and substrate regulation. J Nutr. 2006;136:207S–211S.
    1. Newgard CB, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell metabolism. 2009;9:311–326.
    1. Floegel A, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62:639–648.
    1. Huxley R, Ansary-Moghaddam A, Berrington de Gonzalez A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer. 2005;92:2076–2083.
    1. Hanley JA, McNeil BJ. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. 1982;143:29–36.
    1. Pencina MJ, D'Agostino RB, Sr., D'Agostino RB, Jr., Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27:157–172. discussion 207-112.
    1. Yachida S, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–1117.
    1. Hingorani SR, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–483.
    1. Bardeesy N, et al. Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. Proc Natl Acad Sci U S A. 2006;103:5947–5952.
    1. Jackson EL, et al. The differential effects of mutant p53 alleles on advanced murine lung cancer. Cancer Res. 2005;65:10280–10288.
    1. DuPage M, Dooley AL, Jacks T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nature protocols. 2009;4:1064–1072.
    1. Kirsch DG, et al. A spatially and temporally restricted mouse model of soft tissue sarcoma. Nat Med. 2007;13:992–997.
    1. Duell EJ, et al. Pancreatitis and pancreatic cancer risk: a pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol. 2012;23:2964–2970.
    1. Guerra C, et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291–302.
    1. Gidekel Friedlander SY, et al. Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell. 2009;16:379–389.
    1. Brosnan JT. Interorgan amino acid transport and its regulation. J Nutr. 2003;133:2068S–2072S.
    1. Matthews DE, Marano MA, Campbell RG. Splanchnic bed utilization of leucine and phenylalanine in humans. The American journal of physiology. 1993;264:E109–118.
    1. Cynober LA. Plasma amino acid levels with a note on membrane transport: characteristics, regulation, and metabolic significance. Nutrition. 2002;18:761–766.
    1. Acharyya S, et al. Cancer cachexia is regulated by selective targeting of skeletal muscle gene products. J Clin Invest. 2004;114:370–378.
    1. Fearon KC, Glass DJ, Guttridge DC. Cancer cachexia: mediators, signaling, and metabolic pathways. Cell metabolism. 2012;16:153–166.
    1. Lecker SH, et al. Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2004;18:39–51.
    1. Lundholm K, Bylund AC, Holm J, Schersten T. Skeletal muscle metabolism in patients with malignant tumor. Eur J Cancer. 1976;12:465–473.
    1. Heymsfield SB, McManus CB. Tissue components of weight loss in cancer patients. A new method of study and preliminary observations. Cancer. 1985;55:238–249.
    1. Dewys WD, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am J Med. 1980;69:491–497.
    1. Wigmore SJ, Plester CE, Richardson RA, Fearon KC. Changes in nutritional status associated with unresectable pancreatic cancer. Br J Cancer. 1997;75:106–109.
    1. Tsoli M, Robertson G. Cancer cachexia: malignant inflammation, tumorkines, and metabolic mayhem. Trends in endocrinology and metabolism: TEM. 2013;24:174–183.
    1. Son J, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–105.
    1. Commisso C, et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature. 2013 Epub ahead of Print May 15, 2013.
    1. Harper AE, Miller RH, Block KP. Branched-chain amino acid metabolism. Annual review of nutrition. 1984;4:409–454.
    1. Shah SH, Kraus WE, Newgard CB. Metabolomic profiling for the identification of novel biomarkers and mechanisms related to common cardiovascular diseases: form and function. Circulation. 2012;126:1110–1120.
    1. Pannala R, Basu A, Petersen GM, Chari ST. New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol. 2009;10:88–95.
    1. Rich-Edwards JW, Corsano KA, Stampfer MJ. Test of the National Death Index and Equifax Nationwide Death Search. Am J Epidemiol. 1994;140:1016–1019.
    1. Michaud DS, et al. Prediagnostic plasma C-peptide and pancreatic cancer risk in men and women. Cancer Epidemiol Biomarkers Prev. 2007;16:2101–2109.
    1. Wolpin BM, et al. Plasma 25-hydroxyvitamin D and risk of pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2012;21:82–91.
    1. Bao Y, et al. A prospective study of plasma adiponectin and pancreatic cancer risk in five US cohorts. J Natl Cancer Inst. 2013;105:95–103.
    1. Pannala R, et al. Temporal association of changes in fasting blood glucose and body mass index with diagnosis of pancreatic cancer. Am J Gastroenterol. 2009;104:2318–2325.
    1. Roberts LD, Souza AL, Gerszten RE, Clish CB. Targeted metabolomics. Curr Protoc Mol Biol. 2012 Chapter 30, Unit 30 32 31-24.
    1. Townsend MK, et al. Reproducibility of metabolomic profiles among men and women in 2 large cohort studies. Clin Chem. 2013;59:1657–1667.
    1. Bland JM, Altman DG. Multiple significance tests: the Bonferroni method. BMJ. 1995;310:170.
    1. Cochran WG. The combination of estimates from different experiments. Biometrics. 1954;10:101–129.
    1. Wacholder S, et al. Performance of common genetic variants in breast-cancer risk models. N Engl J Med. 2010;362:986–993.
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837–845.
    1. Gail MH, et al. Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. J Natl Cancer Inst. 1989;81:1879–1886.
    1. Gail MH, Benichou J. Validation studies on a model for breast cancer risk. J Natl Cancer Inst. 1994;86:573–575.
    1. Saslow D, et al. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin. 2007;57:75–89.
    1. Gail MH, et al. Weighing the risks and benefits of tamoxifen treatment for preventing breast cancer. J Natl Cancer Inst. 1999;91:1829–1846.
    1. Costantino JP, et al. Validation studies for models projecting the risk of invasive and total breast cancer incidence. J Natl Cancer Inst. 1999;91:1541–1548.
    1. Tice JA, et al. Using clinical factors and mammographic breast density to estimate breast cancer risk: development and validation of a new predictive model. Ann Intern Med. 2008;148:337–347.
    1. Barlow WE, et al. Prospective breast cancer risk prediction model for women undergoing screening mammography. J Natl Cancer Inst. 2006;98:1204–1214.
    1. Rockhill B, Spiegelman D, Byrne C, Hunter DJ, Colditz GA. Validation of the Gail et al. model of breast cancer risk prediction and implications for chemoprevention. J Natl Cancer Inst. 2001;93:358–366.
    1. Chen J, et al. Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density. J Natl Cancer Inst. 2006;98:1215–1226.
    1. Cook NR. Use and misuse of the receiver operating characteristic curve in risk prediction. Circulation. 2007;115:928–935.
    1. Pepe MS, Janes H, Longton G, Leisenring W, Newcomb P. Limitations of the odds ratio in gauging the performance of a diagnostic, prognostic, or screening marker. Am J Epidemiol. 2004;159:882–890.
    1. Cook NR, Buring JE, Ridker PM. The effect of including C-reactive protein in cardiovascular risk prediction models for women. Ann Intern Med. 2006;145:21–29.
    1. Ridker PM, Buring JE, Rifai N, Cook NR. Development and validation of improved algorithms for the assessment of global cardiovascular risk in women: the Reynolds Risk Score. Jama. 2007;297:611–619.
    1. Roberts NJ, et al. The predictive capacity of personal genome sequencing. Sci Transl Med. 2012;4:133ra158.
    1. van Ravesteyn NT, et al. Tipping the balance of benefits and harms to favor screening mammography starting at age 40 years: a comparative modeling study of risk. Ann Intern Med. 2012;156:609–617.
    1. Chatterjee N, et al. Projecting the performance of risk prediction based on polygenic analyses of genome-wide association studies. Nat Genet. 2013;45:400–405. 405e401–403.
    1. Danesh J, et al. The Emerging Risk Factors Collaboration: analysis of individual data on lipid, inflammatory and other markers in over 1.1 million participants in 104 prospective studies of cardiovascular diseases. European journal of epidemiology. 2007;22:839–869.
    1. Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med. 2000;342:836–843.
    1. Ridker PM. Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation. 2003;107:363–369.
    1. Pradhan AD, et al. Inflammatory biomarkers, hormone replacement therapy, and incident coronary heart disease: prospective analysis from the Women's Health Initiative observational study. Jama. 2002;288:980–987.
    1. Ridker PM, et al. C-reactive protein levels and outcomes after statin therapy. N Engl J Med. 2005;352:20–28.
    1. Greenland P, et al. 2010 ACCF/AHA guideline for assessment of cardiovascular risk in asymptomatic adults: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2010;56:e50–103.
    1. Ridker PM, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195–2207.
    1. Ridker PM, et al. Reduction in C-reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet. 2009;373:1175–1182.
    1. Kaptoge S, et al. C-reactive protein, fibrinogen, and cardiovascular disease prediction. N Engl J Med. 2012;367:1310–1320.
    1. Wormser D, et al. Separate and combined associations of body-mass index and abdominal adiposity with cardiovascular disease: collaborative analysis of 58 prospective studies. Lancet. 2011;377:1085–1095.
    1. Di Angelantonio E, et al. Lipid-related markers and cardiovascular disease prediction. JAMA. 2012;307:2499–2506.
    1. Ayala JE, et al. Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice. Disease models & mechanisms. 2010;3:525–534.
    1. Clasquin MF, Melamud E, Rabinowitz JD. LC-MS data processing with MAVEN: a metabolomic analysis and visualization engine. Current protocols in bioinformatics / editoral board, Andreas D. Baxevanis ... [et al.] Chapter 14, Unit14 11. 2012.
    1. Antoniewicz MR, et al. Metabolic flux analysis in a nonstationary system: fed-batch fermentation of a high yielding strain of E. coli producing 1,3-propanediol. Metabolic engineering. 2007;9:277–292.
    1. Fernandez CA, Des Rosiers C, Previs SF, David F, Brunengraber H. Correction of 13C mass isotopomer distributions for natural stable isotope abundance. Journal of mass spectrometry : JMS. 1996;31:255–262.
    1. Andrikopoulos S, Blair AR, Deluca N, Fam BC, Proietto J. Evaluating the glucose tolerance test in mice. Am J Physiol Endocrinol Metab. 2008;295:E1323–1332.
    1. Fuchs CS, et al. A prospective study of cigarette smoking and the risk of pancreatic cancer. Arch Intern Med. 1996;156:2255–2260.
    1. Schernhammer ES, et al. A prospective study of aspirin use and the risk of pancreatic cancer in women. J Natl Cancer Inst. 2004;96:22–28.
    1. Wolpin BM, et al. Circulating insulin-like growth factor binding protein-1 and the risk of pancreatic cancer. Cancer Res. 2007;67:7923–7928.
    1. Wolpin BM, et al. ABO blood group and the risk of pancreatic cancer. J Natl Cancer Inst. 2009;101:424–431.

Source: PubMed

3
S'abonner