New Onset of DiabetEs in aSsociation with pancreatic ductal adenocarcinoma (NODES Trial): protocol of a prospective, multicentre observational trial

Dóra Illés, Emese Ivány, Gábor Holzinger, Klára Kosár, M Gordian Adam, Beate Kamlage, Gábor Zsóri, Máté Tajti, Márk M Svébis, Viktor Horváth, Ilona Oláh, Katalin Márta, Szilárd Váncsa, Noémi Zádori, Andrea Szentesi, Bálint Czakó, Péter Hegyi, László Czakó, Dóra Illés, Emese Ivány, Gábor Holzinger, Klára Kosár, M Gordian Adam, Beate Kamlage, Gábor Zsóri, Máté Tajti, Márk M Svébis, Viktor Horváth, Ilona Oláh, Katalin Márta, Szilárd Váncsa, Noémi Zádori, Andrea Szentesi, Bálint Czakó, Péter Hegyi, László Czakó

Abstract

Introduction: Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis with an overall 5-year survival of approximately 8%. The success in reducing the mortality rate of PDAC is related to the discovery of new therapeutic agents, and to a significant extent to the development of early detection and prevention programmes. Patients with new-onset diabetes mellitus (DM) represent a high-risk group for PDAC as they have an eightfold higher risk of PDAC than the general population. The proposed screening programme may allow the detection of PDAC in the early, operable stage. Diagnosing more patients in the curable stage might decrease the morbidity and mortality rates of PDAC and additionally reduce the burden of the healthcare.

Methods and analysis: This is a prospective, multicentre observational cohort study. Patients ≥60 years old diagnosed with new-onset (≤6 months) diabetes will be included. Exclusion criteria are (1) Continuous alcohol abuse; (2) Chronic pancreatitis; (3) Previous pancreas operation/pancreatectomy; (4) Pregnancy; (5) Present malignant disease and (6) Type 1 DM. Follow-up visits are scheduled every 6 months for up to 36 months. Data collection is based on questionnaires. Clinical symptoms, body weight and fasting blood will be collected at each, carbohydrate antigen 19-9 and blood to biobank at every second visit. The blood samples will be processed to plasma and analysed with mass spectrometry (MS)-based metabolomics. The metabolomic data will be used for biomarker validation for early detection of PDAC in the high-risk group patients with new-onset diabetes. Patients with worrisome features will undergo MRI or endoscopic ultrasound investigation, and surgical referral depending on the radiological findings. One of the secondary end points is the incidence of PDAC in patients with newly diagnosed DM.

Ethics and dissemination: The study has been approved by the Scientific and Research Ethics Committee of the Hungarian Medical Research Council (41085-6/2019). We plan to disseminate the results to several members of the healthcare system includining medical doctors, dietitians, nurses, patients and so on. We plan to publish the results in a peer-reviewed high-quality journal for professionals. In addition, we also plan to publish it for lay readers in order to maximalise the dissemination and benefits of this trial.

Trial registration number: ClinicalTrials.gov NCT04164602.

Keywords: general diabetes; pancreatic disease; preventive medicine; protocols & guidelines.

Conflict of interest statement

Competing interests: BK and AMG are employees of Metanomics Health GmbH, Germany.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Flow chart of the study protocol. *Weight loss (except at visit 0), abdominal pain/discomfort, abnormal laboratory data, unstable glucose metabolism despite the adequate diet and medical treatment and without intercurrent infection (except at visit 0). **Fasted (overnight, at least 8 hours) patients’ blood samples at room temperature will be drawn into an EDTA tube. Within 2 hours after blood draw samples will be at 19°C–21°C. After centrifugation, the supernatant is carefully removed. After that, the plasma is transferred in 0.5 ml aliquots to tubes and stored at −80°C, in a dedicated freezer (≤6 hours from centrifuge to freezer). CA19-9, carbohydrate antigen 19–9; EUS, endoscopic ultrasound; GADA, glutamic acid decarboxylase antibodies.

References

    1. National Cancer Institute SEER cancer statistics review 1975-2006.
    1. Hariharan D, Saied A, Kocher HM. Analysis of mortality rates for pancreatic cancer across the world. HPB 2008;10:58–62. 10.1080/13651820701883148
    1. Malvezzi M, Bertuccio P, Levi F, et al. . European cancer mortality predictions for the year 2013. Ann Oncol 2013;24:792–800. 10.1093/annonc/mdt010
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016;66:7–30. 10.3322/caac.21332
    1. Rawla P, Sunkara T, Gaduputi V. Epidemiology of pancreatic cancer: global trends, etiology and risk factors. World J Oncol 2019;10:10–27. 10.14740/wjon1166
    1. Rahib L, Smith BD, Aizenberg R, et al. . Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014;74:2913–21. 10.1158/0008-5472.CAN-14-0155
    1. Gillen S, Schuster T, Meyer Zum Büschenfelde C, et al. . Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med 2010;7:e1000267. 10.1371/journal.pmed.1000267
    1. Brand RE, Lerch MM, Rubinstein WS, et al. . Advances in counselling and surveillance of patients at risk for pancreatic cancer. Gut 2007;56:1460–9. 10.1136/gut.2006.108456
    1. Chari ST, Leibson CL, Rabe KG, et al. . Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology 2005;129:504–11. 10.1016/j.gastro.2005.05.007
    1. Huxley R, Ansary-Moghaddam A, Berrington de González A, et al. . Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer 2005;92:2076–83. 10.1038/sj.bjc.6602619
    1. Ben Q, Xu M, Ning X, et al. . Diabetes mellitus and risk of pancreatic cancer: a meta-analysis of cohort studies. Eur J Cancer 2011;47:1928–37. 10.1016/j.ejca.2011.03.003
    1. Tsuchiya R, Noda T, Harada N, et al. . Collective review of small carcinomas of the pancreas. Ann Surg 1986;203:77–81. 10.1097/00000658-198601000-00013
    1. Pelaez-Luna M, Takahashi N, Fletcher JG, et al. . Resectability of presymptomatic pancreatic cancer and its relationship to onset of diabetes: a retrospective review of CT scans and fasting glucose values prior to diagnosis. Am J Gastroenterol 2007;102:2157–63. 10.1111/j.1572-0241.2007.01480.x
    1. Pannala R, Basu A, Petersen GM, et al. . New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol 2009;10:88–95. 10.1016/S1470-2045(08)70337-1
    1. Illés D, Terzin V, Holzinger G, et al. . New-onset type 2 diabetes mellitus--A high-risk group suitable for the screening of pancreatic cancer? Pancreatology 2016;16:266–71. 10.1016/j.pan.2015.12.005
    1. Hart PA, Kamada P, Rabe KG, et al. . Weight loss precedes cancer-specific symptoms in pancreatic cancer-associated diabetes mellitus. Pancreas 2011;40:768–72. 10.1097/MPA.0b013e318220816a
    1. Kim J-E, Lee KT, Lee JK, et al. . Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol 2004;19:182–6. 10.1111/j.1440-1746.2004.03219.x
    1. Maxim LD, Niebo R, Utell MJ. Screening tests: a review with examples. Inhal Toxicol 2014;26:811–28. 10.3109/08958378.2014.955932
    1. Rickes S, Unkrodt K, Neye H, et al. . Differentiation of pancreatic tumours by conventional ultrasound, unenhanced and echo-enhanced power Doppler sonography. Scand J Gastroenterol 2002;37:1313–20. 10.1080/003655202761020605
    1. Pannala R, Leirness JB, Bamlet WR, et al. . Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology 2008;134:981–7. 10.1053/j.gastro.2008.01.039
    1. Aggarwal G, Ramachandran V, Javeed N, et al. . Adrenomedullin is up-regulated in patients with pancreatic cancer and causes insulin resistance in β cells and mice. Gastroenterology 2012;143:1510–7. 10.1053/j.gastro.2012.08.044
    1. Munigala S, Singh A, Gelrud A, et al. . Predictors for pancreatic cancer diagnosis following new-onset diabetes mellitus. Clin Transl Gastroenterol 2015;6:e118. 10.1038/ctg.2015.44
    1. Choe JW, Kim JS, Kim HJ, et al. . Value of early check-up of carbohydrate antigen 19-9 levels for pancreatic cancer screening in asymptomatic new-onset diabetic patients. Pancreas 2016;45:730–4. 10.1097/MPA.0000000000000538
    1. Muniraj T, Chari ST. Diabetes and pancreatic cancer. Minerva Gastroenterol Dietol 2012;58:331–45.
    1. Chari ST, Kelly K, Hollingsworth MA, et al. . Early detection of sporadic pancreatic cancer: summative review. Pancreas 2015;44:693–712. 10.1097/MPA.0000000000000368
    1. Sah RP, Nagpal SJS, Mukhopadhyay D, et al. . New insights into pancreatic cancer-induced paraneoplastic diabetes. Nat Rev Gastroenterol Hepatol 2013;10:423–33. 10.1038/nrgastro.2013.49
    1. Mayerle J, Kalthoff H, Reszka R, et al. . Metabolic biomarker signature to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Gut 2018;67:128–37. 10.1136/gutjnl-2016-312432
    1. American Diabetes Association 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2018. Diabetes Care 2018;41:S13–27. 10.2337/dc18-S002
    1. Rabinovitz M, Pitlik SD, Leifer M, et al. . Unintentional weight loss. A retrospective analysis of 154 cases. Arch Intern Med 1986;146:186–7. 10.1001/archinte.146.1.186
    1. Jung K, Reszka R, Kamlage B, et al. . Tissue metabolite profiling identifies differentiating and prognostic biomarkers for prostate carcinoma. Int J Cancer 2013;133:2914–24. 10.1002/ijc.28303
    1. Christie WW. Rapid separation and quantification of lipid classes by high performance liquid chromatography and mass (light-scattering) detection. J Lipid Res 1985;26:507–12.
    1. Mutch DM, Fuhrmann JC, Rein D, et al. . Metabolite profiling identifies candidate markers reflecting the clinical adaptations associated with Roux-en-Y gastric bypass surgery. PLoS One 2009;4:e7905. 10.1371/journal.pone.0007905
    1. Roessner U, Wagner C, Kopka J, et al. . Technical advance: simultaneous analysis of metabolites in potato tuber by gas chromatography-mass spectrometry. Plant J 2000;23:131–42. 10.1046/j.1365-313x.2000.00774.x
    1. van Ravenzwaay B, Cunha GC-P, Leibold E, et al. . The use of metabolomics for the discovery of new biomarkers of effect. Toxicol Lett 2007;172:21–8. 10.1016/j.toxlet.2007.05.021
    1. Schmidt H, Schmidt R, Geisslinger G. LC-MS/MS-analysis of sphingosine-1-phosphate and related compounds in plasma samples. Prostaglandins Other Lipid Mediat 2006;81:162–70. 10.1016/j.prostaglandins.2006.09.003
    1. Meller S, Meyer H-A, Bethan B, et al. . Integration of tissue metabolomics, transcriptomics and immunohistochemistry reveals ERG- and Gleason score-specific metabolomic alterations in prostate cancer. Oncotarget 2016;7:1421–38. 10.18632/oncotarget.6370
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B 1995;57:289–300. 10.1111/j.2517-6161.1995.tb02031.x
    1. Gart JJ, Nam J. Approximate interval estimation of the ratio of binomial parameters: a review and corrections for skewness. Biometrics 1988;44:323–38. 10.2307/2531848
    1. Scherer R, Schaarschmidt F, Prescher S, et al. . Simultaneous confidence intervals for comparing biodiversity indices estimated from overdispersed count data. Biom J 2013;55:246–63. 10.1002/bimj.201200157
    1. Ewald N, Bretzel RG. Diabetes mellitus secondary to pancreatic diseases (Type 3c)--are we neglecting an important disease? Eur J Intern Med 2013;24:203–6. 10.1016/j.ejim.2012.12.017
    1. Hritz I, Hegyi P. Early achievable severity (easy) index for simple and accurate expedite risk stratification in acute pancreatitis. J Gastrointestin Liver Dis 2015;24:177–82. 10.15403/jgld.2014.1121.242.easy
    1. Párniczky A, Mosztbacher D, Zsoldos F, et al. . Analysis of pediatric pancreatitis (APPLE trial): Pre-study protocol of a multinational prospective clinical trial. Digestion 2016;93:105–10. 10.1159/000441353
    1. Zsoldos F, Párniczky A, Mosztbacher D, et al. . Pain in the early phase of pediatric pancreatitis (PINEAPPLE trial): Pre-Study protocol of a multinational prospective clinical trial. Digestion 2016;93:121–6. 10.1159/000441352
    1. Dubravcsik Z, Madácsy L, Gyökeres T, et al. . Preventive pancreatic stents in the management of acute biliary pancreatitis (PREPAST trial): pre-study protocol for a multicenter, prospective, randomized, interventional, controlled trial. Pancreatology 2015;15:115–23. 10.1016/j.pan.2015.02.007
    1. Márta K, Szabó AN, Pécsi D, et al. . High versus low energy administration in the early phase of acute pancreatitis (GOULASH trial): protocol of a multicentre randomised double-blind clinical trial. BMJ Open 2017;7:e015874. 10.1136/bmjopen-2017-015874
    1. Dubravcsik Z, Farkas G, Hegyi P, et al. . [Autoimmune pancreatitis. Evidence based management guidelines of the Hungarian Pancreatic Study Group]. Orv Hetil 2015;156:292–307. 10.1556/OH.2015.30061
    1. Hritz I, Czakó L, Dubravcsik Z, et al. . [Acute pancreatitis. Evidence-based practice guidelines, prepared by the Hungarian Pancreatic Study Group]. Orv Hetil 2015;156:244–61. 10.1556/OH.2015.30059
    1. Párniczky A, Czakó L, Dubravcsik Z, et al. . Pediatric pancreatitis. Evidence based management guidelines of the Hungarian Pancreatic Study Group. Orv Hetil 2015;156:308–25. 10.1556/OH.2015.30062
    1. Takács T, Czakó L, Dubravcsik Z, et al. . [Chronic pancreatitis. Evidence based management guidelines of the Hungarian Pancreatic Study Group]. Orv Hetil 2015;156:262–88. 10.1556/OH.2015.30060

Source: PubMed

3
Prenumerera