Combined microRNA and mRNA microfluidic TaqMan array cards for the diagnosis of malignancy of multiple types of pancreatico-biliary tumors in fine-needle aspiration material

Thomas M Gress, Ludwig Lausser, Lyn-Rouven Schirra, Lisa Ortmüller, Ramona Diels, Bo Kong, Christoph W Michalski, Thilo Hackert, Oliver Strobel, Nathalia A Giese, Miriam Schenk, Rita T Lawlor, Aldo Scarpa, Hans A Kestler, Malte Buchholz, Thomas M Gress, Ludwig Lausser, Lyn-Rouven Schirra, Lisa Ortmüller, Ramona Diels, Bo Kong, Christoph W Michalski, Thilo Hackert, Oliver Strobel, Nathalia A Giese, Miriam Schenk, Rita T Lawlor, Aldo Scarpa, Hans A Kestler, Malte Buchholz

Abstract

Pancreatic ductal adenocarcinoma (PDAC) continues to carry the lowest survival rates among all solid tumors. A marked resistance against available therapies, late clinical presentation and insufficient means for early diagnosis contribute to the dismal prognosis. Novel biomarkers are thus required to aid treatment decisions and improve patient outcomes. We describe here a multi-omics molecular platform that allows for the first time to simultaneously analyze miRNA and mRNA expression patterns from minimal amounts of biopsy material on a single microfluidic TaqMan Array card. Expression profiles were generated from 113 prospectively collected fine needle aspiration biopsies (FNAB) from patients undergoing surgery for suspect masses in the pancreas. Molecular classifiers were constructed using support vector machines, and rigorously evaluated for diagnostic performance using 10×10fold cross validation. The final combined miRNA/mRNA classifier demonstrated a sensitivity of 91.7%, a specificity of 94.5%, and an overall diagnostic accuracy of 93.0% for the differentiation between PDAC and benign pancreatic masses, clearly outperfoming miRNA-only classifiers. The classification algorithm also performed very well in the diagnosis of other types of solid tumors (acinar cell carcinomas, ampullary cancer and distal bile duct carcinomas), but was less suited for the diagnostic analysis of cystic lesions. We thus demonstrate that simultaneous analysis of miRNA and mRNA biomarkers from FNAB samples using multi-omics TaqMan Array cards is suitable to differentiate suspect solid pancreatic masses with high precision.

Keywords: fine needle aspiration biopsy; microfluidic TaqMan arrays; molecular diagnostics; pancreatic cancer; pancreatico-biliary tumors.

Conflict of interest statement

CONFLICTS OF INTEREST The authors declare no conflicts of interest.

Figures

Figure 1. Study design
Figure 1. Study design
Figure 2. Schematic representation of 10×10 cross…
Figure 2. Schematic representation of 10×10 cross validation procedure
The original dataset is split into ten subsets of approximately equal size. Nine of these subsets are used for training the classification model (SVM). The tenth subset is used as an independent subset for evaluating the performance of the trained classifier. This procedure is repeated for each subset and an overall accuracy is calculated as the mean accuracy over ten permutations of the original dataset.
Figure 3. Exhaustive evaluation of all miRNA…
Figure 3. Exhaustive evaluation of all miRNA marker combinations
All possible combinations of miRNA markers, from single miRNAs to the combination of all 9 markers (29-1 = 511 possible combinations), were tested for their diagnostic performances on the set of PDAC and CP samples. Each combination was evaluated by a linear SVM and sensitivity, specificity and diagnostic accuracy determined by 10×10-fold cross validation. Results are plotted in decreasing order according to accuracy. Note that none of the resulting classifiers exceeded a diagnostic accuracy of 75%, with the best-performing combination achieving a sensitivity of 58.5% and a specificity of 88.3%. “CP” = chronic pancreatitis; “PDAC” = pancreatic ductal adenocarcinoma; “Acc” = accuracy; “Sens” = sensitivity; “Spec” = specificity.
Figure 4. Unsupervised clustering of expression profiles…
Figure 4. Unsupervised clustering of expression profiles from PDAC and CP biopsies
Individual samples are shown in columns; genes are shown in rows. Both the samples and the genes were hierarchically clustered (complete linkage clustering) using uncentered Pearson correlation as the similarity measure. Red cells indicate high expression, black intermediate expression, and green low expression of a gene in the respective group. Clusters of CP (blue bars) and PDAC samples (yellow bars) are readily apparent. “CP” = chronic pancreatitis; “PDAC” = pancreatic ductal adenocarcinoma.
Figure 5. Training of mixed miRNA /…
Figure 5. Training of mixed miRNA / mRNA classifiers
Combined profiles of miRNA and mRNA markers (Panel i) were analyzed in a sequence of marker selection and classification experiment. First, a subset of candidate markers was selected via the univariate TNoMcw score (Panel ii). The reduced marker profiles were then used to train a multivariate SVM (Panel iii). The final decision can be projected to univariate space (Panel iv).
Figure 6. Evaluation of the combined miRNA/mRNA…
Figure 6. Evaluation of the combined miRNA/mRNA marker profiles
Accuracy, sensitivity and specificity gained in the 10×10 cross validation experiments with the combined miRNA/mRNA marker profiles. Classification models vary in the number of markers that were selected during their training phases. Results are given in increasing order from 1 to 88 selected biomarkers. Accuracies ≥ 90% are marked in yellow. “CP” = chronic pancreatitis; “PDAC” = pancreatic ductal adenocarcinoma.
Figure 7. Performance of the final classifier…
Figure 7. Performance of the final classifier in identifying malignancy on biopsy samples from pancreatobiliary tumors other than PDAC
Retraining of the optimal model (77 markers) on all available samples. Prediction of PDAC and CP samples and categorization of all remaining tissue-types is given. “CP” = chronic pancreatitis; “PDAC” = pancreatic ductal adenocarcinoma; “CA” = carcinoma; “IPMN” = intraductal papillary mucinous neoplasm; “SPT” = solid pseudopapillary neoplasia.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7–30.
    1. Michl P, Gress TM. Current concepts and novel targets in advanced pancreatic cancer. Gut. 2013;62:317–326.
    1. Hartwig W, Werner J, Jager D, Debus J, Buchler MW. Improvement of surgical results for pancreatic cancer. Lancet Oncol. 2013;14:e476–485.
    1. Hackeng WM, Hruban RH, Offerhaus GJ, Brosens LA. Surgical and molecular pathology of pancreatic neoplasms. Diagn Pathol. 2016;11:47.
    1. Kamisawa T, Chari ST, Lerch MM, Kim MH, Gress TM, Shimosegawa T. Recent advances in autoimmune pancreatitis: type 1 and type 2. Gut. 2013;62:1373–1380.
    1. Shimosegawa T, Chari ST, Frulloni L, Kamisawa T, Kawa S, Mino-Kenudson M, Kim MH, Kloppel G, Lerch MM, Lohr M, Notohara K, Okazaki K, Schneider A, Zhang L. International consensus diagnostic criteria for autoimmune pancreatitis: guidelines of the International Association of Pancreatology. Pancreas. 2011;40:352–358.
    1. Puli SR, Bechtold ML, Buxbaum JL, Eloubeidi MA. How good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: a meta-analysis and systematic review. Pancreas. 2013;42:20–26.
    1. Eloubeidi MA, Decker GA, Chandrasekhara V, Chathadi KV, Early DS, Evans JA, Fanelli RD, Fisher DA, Foley K, Hwang JH, Jue TL, Lightdale JR, Pasha SF, et al. The role of endoscopy in the evaluation and management of patients with solid pancreatic neoplasia. Gastrointest Endosc. 2016;83:17–28.
    1. Storm AC, Lee LS. Endoscopic ultrasound-guided techniques for diagnosing pancreatic mass lesions: can we do better? World J Gastroenterol. 2016;22:8658–8669.
    1. Agarwal B, Abu-Hamda E, Molke KL, Correa AM, Ho L. Endoscopic ultrasound-guided fine needle aspiration and multidetector spiral CT in the diagnosis of pancreatic cancer. Am J Gastroenterol. 2004;99:844–850.
    1. Gress TM, Kestler HA, Lausser L, Fiedler L, Sipos B, Michalski CW, Werner J, Giese N, Scarpa A, Buchholz M. Differentiation of multiple types of pancreatico-biliary tumors by molecular analysis of clinical specimens. J Mol Med (Berl) 2012;90:457–464.
    1. Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, Cooc J, Weinkle J, Kim GE, Jakkula L, Feiler HS, Ko AH, Olshen AB, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17:500–503.
    1. Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, Rashid NU, Williams LA, Eaton SC, Chung AH, Smyla JK, Anderson JM, Kim HJ, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47:1168–1178.
    1. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, Miller DK, Christ AN, Bruxner TJ, Quinn MC, Nourse C, Murtaugh LC, Harliwong I, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.
    1. Haider S, Wang J, Nagano A, Desai A, Arumugam P, Dumartin L, Fitzgibbon J, Hagemann T, Marshall JF, Kocher HM, Crnogorac-Jurcevic T, Scarpa A, Lemoine NR, Chelala C. A multi-gene signature predicts outcome in patients with pancreatic ductal adenocarcinoma. Genome Med. 2014;6:105.
    1. D'Angelo B, Benedetti E, Cimini A, Giordano A. MicroRNAs: a puzzling tool in cancer diagnostics and therapy. Anticancer Res. 2016;36:5571–5575.
    1. Brunetti O, Russo A, Scarpa A, Santini D, Reni M, Bittoni A, Azzariti A, Aprile G, Delcuratolo S, Signorile M, Gnoni A, Palermo L, Lorusso V, et al. MicroRNA in pancreatic adenocarcinoma: predictive/prognostic biomarkers or therapeutic targets? Oncotarget. 2015;6:23323–23341. .
    1. Huang J, Liu J, Chen-Xiao K, Zhang X, Lee WN, Go VL, Xiao GG. Advance in microRNA as a potential biomarker for early detection of pancreatic cancer. Biomark Res. 2016;4:20.
    1. Hernandez YG, Lucas AL. MicroRNA in pancreatic ductal adenocarcinoma and its precursor lesions. World J Gastrointest Oncol. 2016;8:18–29.
    1. Cote GA, Gore AJ, McElyea SD, Heathers LE, Xu H, Sherman S, Korc M. A pilot study to develop a diagnostic test for pancreatic ductal adenocarcinoma based on differential expression of select miRNA in plasma and bile. Am J Gastroenterol. 2014;109:1942–1952.
    1. Brand RE, Adai AT, Centeno BA, Lee LS, Rateb G, Vignesh S, Menard C, Wiechowska-Kozlowska A, Boldys H, Hartleb M, Sanders MK, Munding JB, Tannapfel A, et al. A microRNA-based test improves endoscopic ultrasound-guided cytologic diagnosis of pancreatic cancer. Clin Gastroenterol Hepatol. 2014;12:1717–1723.
    1. Frampton AE, Krell J, Prado MM, Gall TM, Abbassi-Ghadi N, Del Vecchio Blanco G, Funel N, Giovannetti E, Castellano L, Basyouny M, Habib NA, Kaltsidis H, Vlavianos P, et al. Prospective validation of microRNA signatures for detecting pancreatic malignant transformation in endoscopic-ultrasound guided fine-needle aspiration biopsies. Oncotarget. 2016;7:28556–28569. .
    1. Ferlinz A, Miller C, Formosa R, Lee KY. Gene expression analysis of both mRNA and miRNA on the same TaqMan(R) Array Card: development of a pancreatic tumor tissue classification methodology. Methods. 2013;59:S11–S15.
    1. Szafranska AE, Davison TS, John J, Cannon T, Sipos B, Maghnouj A, Labourier E, Hahn SA. MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene. 2007;26:4442–4452.
    1. Szafranska AE, Doleshal M, Edmunds HS, Gordon S, Luttges J, Munding JB, Barth RJ, Jr, Gutmann EJ, Suriawinata AA, Marc Pipas J, Tannapfel A, Korc M, Hahn SA, et al. Analysis of microRNAs in pancreatic fine-needle aspirates can classify benign and malignant tissues. Clin Chem. 2008;54:1716–1724.
    1. Munding JB, Adai AT, Maghnouj A, Urbanik A, Zollner H, Liffers ST, Chromik AM, Uhl W, Szafranska-Schwarzbach AE, Tannapfel A, Hahn SA. Global microRNA expression profiling of microdissected tissues identifies miR-135b as a novel biomarker for pancreatic ductal adenocarcinoma. Int J Cancer. 2012;131:E86–E95.
    1. Biankin AV, Maitra A. Subtyping pancreatic cancer. Cancer Cell. 2015;28:411–413.
    1. Neesse A, Algul H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64:1476–1484.
    1. Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goere D, Seufferlein T, Haustermans K, Van Laethem JL, Conroy T, Arnold D, ESMO Guidelines Committee Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26:v56–68.
    1. Seufferlein T, Bachet JB, Van CE Rougier P. Pancreatic adenocarcinoma: ESMO-ESDO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012;23:vii33–40.
    1. Jenssen C, Alvarez-Sanchez MV, Napoleon B, Faiss S. Diagnostic endoscopic ultrasonography: assessment of safety and prevention of complications. World J Gastroenterol. 2012;18:4659–4676.
    1. Ngamruengphong S, Swanson KM, Shah ND, Wallace MB. Preoperative endoscopic ultrasound-guided fine needle aspiration does not impair survival of patients with resected pancreatic cancer. Gut. 2015;64:1105–1110.
    1. Garrido-Laguna I, Hidalgo M. Pancreatic cancer: from state-of-the-art treatments to promising novel therapies. Nat Rev Clin Oncol. 2015;12:319–334.
    1. Ielpo B, Duran H, Diaz E, Fabra I, Caruso R, Ferri V, Malave L, Hidalgo M, Alvarez R, Plaza C, Quijano Y, Vicente E. Preoperative treatment with gemcitabine plus nab-paclitaxel is a safe and effective chemotherapy for pancreatic adenocarcinoma. Eur J Surg Oncol. 2016;42:1394–1400.
    1. Hackert T, Sachsenmaier M, Hinz U, Schneider L, Michalski CW, Springfeld C, Strobel O, Jager D, Ulrich A, Buchler MW. Locally advanced pancreatic cancer: neoadjuvant therapy with folfirinox results in resectability in 60% of the patients. Ann Surg. 2016;264:457–463.
    1. Hackert T, Ulrich A, Buchler MW. Borderline resectable pancreatic cancer. Cancer Lett. 2016;375:231–237.
    1. Petrelli F, Coinu A, Borgonovo K, Cabiddu M, Ghilardi M, Lonati V, Aitini E, Barni S. FOLFIRINOX-based neoadjuvant therapy in borderline resectable or unresectable pancreatic cancer: a meta-analytical review of published studies. Pancreas. 2015;44:515–521.
    1. Cioffi M, Trabulo SM, Sanchez-Ripoll Y, Miranda-Lorenzo I, Lonardo E, Dorado J, Reis Vieira C, Ramirez JC, Hidalgo M, Aicher A, Hahn S, Sainz B, Jr, Heeschen C. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem cells. Gut. 2015;64:1936–1948.
    1. Matthaei H, Wylie D, Lloyd MB, Dal Molin M, Kemppainen J, Mayo SC, Wolfgang CL, Schulick RD, Langfield L, Andruss BF, Adai AT, Hruban RH, Szafranska-Schwarzbach AE, Maitra A. miRNA biomarkers in cyst fluid augment the diagnosis and management of pancreatic cysts. Clin Cancer Res. 2012;18:4713–4724.
    1. Wang J, Paris PL, Chen J, Ngo V, Yao H, Frazier ML, Killary AM, Liu CG, Liang H, Mathy C, Bondada S, Kirkwood K, Sen S. Next generation sequencing of pancreatic cyst fluid microRNAs from low grade-benign and high grade-invasive lesions. Cancer Lett. 2015;356:404–409.
    1. Springer S, Wang Y, Dal Molin M, Masica DL, Jiao Y, Kinde I, Blackford A, Raman SP, Wolfgang CL, Tomita T, Niknafs N, Douville C, Ptak J, et al. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology. 2015;149:1501–1510.
    1. Jones M, Zheng Z, Wang J, Dudley J, Albanese E, Kadayifci A, Dias-Santagata D, Le L, Brugge WR, Fernandez-del Castillo C, Mino-Kenudson M, Iafrate AJ, Pitman MB. Impact of next-generation sequencing on the clinical diagnosis of pancreatic cysts. Gastrointest Endosc. 2016;83:140–148.
    1. Lausser L, Müssel C, Maucher M, Kestler HA. Measuring and visualizing the stability of biomarker selection techniques. Comput Stat. 2013;28:51–65.
    1. Schirra LR, Lausser L, Kestler HA. Selection Stability as a Means of Biomarker Discovery in Classification. In: Wilhelm AFX, Kestler HA, editors. Analysis of Large and Complex Data. Springer International Publishing; 2016. pp. 79–89.
    1. Kestler HA, Lausser L, Lindner W, Palm G. On the fusion of threshold classifiers for categorization and dimensionality reduction. Comput Stat. 2011;26:321–340.
    1. Ben-Dor A, Bruhn L, Friedman N, Nachman I, Schummer M, Yakhini Z. Tissue classification with gene expression profiles. J Comput Biol. 2000;7:559–583.
    1. Vapnik VN. Statistical Learning Theory. New York: Wiley; 1998.
    1. Shah M, Japkowicz N. Evaluating Learning Algorithms: A Classification Perspective. New York, NY: US Cambridge University Press; 2011.
    1. Müssel C, Lausser L, Maucher M, Kestler HA. Multi-Objective Parameter Selection for Classifiers. J Stat Softw. 2012;46:1–27.

Source: PubMed

3
Abonnere