The identification of gut neuroendocrine tumor disease by multiple synchronous transcript analysis in blood

Irvin M Modlin, Ignat Drozdov, Mark Kidd, Irvin M Modlin, Ignat Drozdov, Mark Kidd

Abstract

Gastroenteropancreatic (GEP) neuroendocrine neoplasms (NENs) are increasing in both incidence and prevalence. A delay in correct diagnosis is common for these lesions. This reflects the absence of specific blood biomarkers to detect NENs. Measurement of the neuroendocrine secretory peptide Chromogranin A (CgA) is used, but is a single value, is non-specific and assay data are highly variable. To facilitate tumor detection, we developed a multi-transcript molecular signature for PCR-based blood analysis. NEN transcripts were identified by computational analysis of 3 microarray datasets: NEN tissue (n = 15), NEN peripheral blood (n = 7), and adenocarcinoma (n = 363 tumors). The candidate gene signature was examined in 130 blood samples (NENs: n = 63) and validated in two independent sets (Set 1 [n = 115, NENs: n = 72]; Set 2 [n = 120, NENs: n = 58]). Comparison with CgA (ELISA) was undertaken in 176 samples (NENs: n = 81). 51 significantly elevated transcript markers were identified. Gene-based classifiers detected NENs in independent sets with high sensitivity (85-98%), specificity (93-97%), PPV (95-96%) and NPV (87-98%). The AUC for the NEN gene-based classifiers was 0.95-0.98 compared to 0.64 for CgA (Z-statistic 6.97-11.42, p<0.0001). Overall, the gene-based classifier was significantly (χ(2) = 12.3, p<0.0005) more accurate than CgA. In a sub-analysis, pancreatic NENs and gastrointestinal NENs could be identified with similar efficacy (79-88% sensitivity, 94% specificity), as could metastases (85%). In patients with low CgA, 91% exhibited elevated transcript markers. A panel of 51 marker genes differentiates NENs from controls with a high PPV and NPV (>90%), identifies pancreatic and gastrointestinal NENs with similar efficacy, and confirms GEP-NENs when CgA levels are low. The panel is significantly more accurate than the CgA assay. This reflects its utility to identify multiple diverse biological components of NENs. Application of this sensitive and specific PCR-based blood test to NENs will allow accurate detection of disease, and potentially define disease progress enabling monitoring of treatment efficacy.

Conflict of interest statement

Competing Interests: We received funding from a commercial source: Clifton Life Sciences. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Computational pipeline used to derive…
Figure 1. Computational pipeline used to derive a set of 51 markers that identify GEP-NEN disease.
Step 1: Gene co-expression networks inferred from GEP-NEN-A and GEP-NEN-B datasets are intersected, producing the GEP-NEN network. Step 2: Co-expression networks from neoplastic and normal tissue microarray datasets are combined to produce the normal and neoplastic networks. Step 3: Links present in normal and neoplastic networks are subtracted from the GEP-NEN network. Step 4: Concordantly regulated genes in GEP-NEN-A and GEP-NEN-B networks are retained; other genes are eliminated from the GEP-NEN network, producing the Consensus GEP-NEN network. Step 5: Upregulated genes in both the GEP-NEN-A and GEP-NEN-B dataset are mapped to the Consensus GEP-NEN network. Step 6: Topological filtering, expression profiling, and literature-curation of putative tissue-based markers, yielding 21 putative genes further examined by RT-PCR. Step 7: Identification of mutually up-regulated genes in GEP-NEN blood transcriptome and GEP-NEN-A and GEP-NEN-B datasets, yielding 32 putative genes further examined by RT-PCR. Step 8: Literature-curation and cancer mutation database search, yielding a panel of 22 putative marker genes for further RT-PCR analysis.
Figure 2. GEP-NEN gene co-expression network.
Figure 2. GEP-NEN gene co-expression network.
A) Visualization of the GEP-NEN gene co-expression network (2545 genes, 30249 edges). Each node represents a gene, while a link represents a GEP-NEN-specific co-expression. Nodes that localized to the same network community are marked in the same color. The community structure of the GEP-NEN network is further visualized in the 3 dimensional inset, whereby each node represents a community while edges are drawn between communities that contain co-expressed genes. Larger nodes indicate bigger gene communities. B) Heatmap visualizing enrichment for over-represented Gene Ontology (GO) Biological Process (BP) terms assigned to the 10 largest clusters (>20 genes). Heatmap colors represent the significance of the enrichment.
Figure 3. Utility of the 51 marker…
Figure 3. Utility of the 51 marker gene signature for identification of GEP-NEN disease.
A) Unsupervised hierarchical clustering of the 130 samples in the training set (n = 67 controls, n = 63 GEP-NENs). Tree was generated with an average agglomeration method and 1-(sample correlation) was used as a measure of dissimilarity. Unique colors under the dendrogram represent sample cluster assignments, computed by cutting the hierarchical tree at height = 0.99 (black line), 0.85 (blue line), or 0.50 (red line) using a dynamic tree cutting approach . B) Prediction accuracy of each classifier using sequential addition of 27 significantly up-regulated genes (p<0.05) in the GEP-NEN samples obtained using results of the 10-fold cross validation. C) Receiver operating characteristic (ROC) curves for “majority vote” classifier applied to validation sets 1 (AUC = 0.98, p<0.0001) and 2 (AUC = 0.95, p<0.0001) compared to ROC curve for utility of the plasma CgA values (AUC = 0.64, p<0.002). Direct comparisons of AUCs between set 1 or set 2 and CgA identified estimated Z-scores of 10.57 and 11.42 respectively, confirming the significant differences between the two detection systems (calculations detailed in Supplementary Methods).
Figure 4. Comparison of the 51 marker…
Figure 4. Comparison of the 51 marker gene signature with Chromogranin A (CgA) for detecting GEP-NENs.
A) CgA levels were significantly elevated in the GEP-NEN group (n = 176; *p<0.002) but an overlap with normal values was identified. B) Comparison of the PCR-based approach with CgA protein measurement identified that call rates were significantly higher for the PCR-based test (*p<0.0005, χ2 = 12.3). The PCR blood test was significantly more accurate than measurement of CgA levels to detect GEP-NENs. ULN = upper limit of normal (19U/L – DAKO).
Figure 5. Utility of the 51 marker…
Figure 5. Utility of the 51 marker gene signature for detecting P-NENs, metastases and in patients with low Chromogranin A (CgA).
A) The sensitivity and specificity of the test to detect GI-NENs (90%, 94%) and P-NENs (80%, 94%) was similar. B) The PCR-based approach could detect patients with no metastases as well as patients with metastases. C) The PCR-based test could accurately identify GEP-NENs even when plasma CgA were low (<19U/L). Overall, the PCR blood test was significantly more accurate than measurement of CgA levels to detect GEP-NENs (*p<10−13, χ2>50).

References

    1. Modlin IM, Oberg K, Chung DC, Jensen RT, de Herder WW, et al. (2008) Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol 9: 61–72.
    1. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, et al. (2008) One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 26: 3063–3072.
    1. Kulke MH, Siu LL, Tepper JE, Fisher G, Jaffe D, et al. (2011) Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol 29: 934–943.
    1. Yao JC, Pavel M, Phan AT, Kulke MH, Hoosen S, et al. (2011) Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. J Clin Endocrinol Metab 96: 3741–3749. Epub 2011 Oct 3712.
    1. AACC (2010) Chromogranin A. Lab Tests Online: American Association for Clinical Chemistry. pp. Details regarding Chromogranin A as a test.
    1. van ’t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, et al. (2002) Gene expression profiling predicts clinical outcome of breast cancer. Nature 415: 530–536.
    1. Hess KR, Anderson K, Symmans WF, Valero V, Ibrahim N, et al. (2006) Pharmacogenomic predictor of sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide in breast cancer. J Clin Oncol 24: 4236–4244. Epub 2006 Aug 4238.
    1. Frederiksen CM, Knudsen S, Laurberg S, Orntoft TF (2003) Classification of Dukes’ B and C colorectal cancers using expression arrays. J Cancer Res Clin Oncol 129: 263–271.
    1. Kidd M, Modlin IM, Mane SM, Camp RL, Eick GN, et al. (2006) Utility of molecular genetic signatures in the delineation of gastric neoplasia. Cancer 106: 1480–1488.
    1. Modlin IM, Kidd M, Latich I, Zikusoka MN, Eick GN, et al. (2006) Genetic differentiation of appendiceal tumor malignancy: a guide for the perplexed. Ann Surg 244: 52–60.
    1. Kidd M, Modlin IM, Mane SM, Camp RL, Eick G, et al. (2006) The role of genetic markers–NAP1L1, MAGE-D2, and MTA1–in defining small-intestinal carcinoid neoplasia. Ann Surg Oncol 13: 253–262. Epub 2006 Jan 2020.
    1. Drozdov I, Kidd M, Nadler B, Camp RL, Mane SM, et al. (2009) Predicting neuroendocrine tumor (carcinoid) neoplasia using gene expression profiling and supervised machine learning. Cancer 115: 1638–1650.
    1. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, et al. (2005) Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst 97: 1180–1184.
    1. Parkinson H, Kapushesky M, Kolesnikov N, Rustici G, Shojatalab M, et al. (2009) ArrayExpress update–from an archive of functional genomics experiments to the atlas of gene expression. Nucleic Acids Res 37: D868–872.
    1. Kidd M, Nadler B, Mane S, Eick G, Malfertheiner M, et al. (2007) GeneChip, geNorm, and gastrointestinal tumors: novel reference genes for real-time PCR. Physiological genomics 30: 363–370.
    1. Leja J, Essaghir A, Essand M, Wester K, Oberg K, et al. (2009) Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas. Mod Pathol 22: 261–272.
    1. Shou J, Dotson C, Qian HR, Tao W, Lin C, et al. (2005) Optimized blood cell profiling method for genomic biomarker discovery using high-density microarray. Biomarkers 10: 310–320.
    1. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, et al. (2003) Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4: 249–264.
    1. Drozdov I, Ouzounis CA, Shah AM, Tsoka S (2011) Functional Genomics Assistant (FUGA): a toolbox for the analysis of complex biological networks. BMC research notes 4: 462.
    1. Modlin IM, Gustafsson BI, Drozdov I, Nadler B, Pfragner R, et al. (2009) Principal component analysis, hierarchical clustering, and decision tree assessment of plasma mRNA and hormone levels as an early detection strategy for small intestinal neuroendocrine (carcinoid) tumors. Ann Surg Oncol 16: 487–498.
    1. Sarac OS, Atalay V, Cetin-Atalay R (2010) GOPred: GO molecular function prediction by combined classifiers. PloS one 5: e12382.
    1. Horvath S, Dong J (2008) Geometric interpretation of gene coexpression network analysis. PLoS computational biology 4: e1000117.
    1. Stuart JM, Segal E, Koller D, Kim SK (2003) A gene-coexpression network for global discovery of conserved genetic modules. Science 302: 249–255.
    1. Blondel VD, Guillaume J-L, Lambiotte R, Lefebvre E (2008) Fast unfolding of communities in large network. J Stat Mech P10008.
    1. Ippolito JE, Xu J, Jain S, Moulder K, Mennerick S, et al. (2005) An integrated functional genomics and metabolomics approach for defining poor prognosis in human neuroendocrine cancers. Proceedings of the National Academy of Sciences of the United States of America 102: 9901–9906.
    1. Li L, Zhang K, Lee J, Cordes S, Davis DP, et al. (2009) Discovering cancer genes by integrating network and functional properties. BMC Med Genomics 2: 61.
    1. Muscarella LA, D’Alessandro V, la Torre A, Copetti M, De Cata A, et al. (2011) Gene expression of somatostatin receptor subtypes SSTR2a, SSTR3 and SSTR5 in peripheral blood of neuroendocrine lung cancer affected patients. Cell Oncol 19: 19.
    1. Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, et al. (2011) COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic acids research 39: D945–950.
    1. Zatelli MC, Torta M, Leon A, Ambrosio MR, Gion M, et al. (2007) Chromogranin A as a marker of neuroendocrine neoplasia: an Italian Multicenter Study. Endocr Relat Cancer 14: 473–482.
    1. Ramachandran R, Bech P, Murphy KG, Dhillo WS, Meeran KM, et al. (2012) Improved diagnostic accuracy for neuroendocrine neoplasms using two chromogranin A assays. Clin Endocrinol (Oxf) 76: 831–836 doi:.
    1. Palmieri G, Pirastu M, Strazzullo M, Ascierto PA, Satriano SM, et al. (2001) Clinical significance of PCR-positive mRNA markers in peripheral blood and regional nodes of malignant melanoma patients. Melanoma Cooperative Group. Recent Results Cancer Res 158: 200–203.
    1. Van der Auwera I, Peeters D, Benoy IH, Elst HJ, Van Laere SJ, et al. (2010) Circulating tumour cell detection: a direct comparison between the CellSearch System, the AdnaTest and CK-19/mammaglobin RT-PCR in patients with metastatic breast cancer. Br J Cancer 102: 276–284. Epub 2009 Dec 2001.
    1. Stridsberg M, Eriksson B, Oberg K, Janson ET (2003) A comparison between three commercial kits for chromogranin A measurements. J Endocrinol 177: 337–341.
    1. Aggarwal A, Guo DL, Hoshida Y, Yuen ST, Chu KM, et al. (2006) Topological and functional discovery in a gene coexpression meta-network of gastric cancer. Cancer Res 66: 232–241.
    1. Vischioni B, Oudejans JJ, Vos W, Rodriguez JA, Giaccone G (2006) Frequent overexpression of aurora B kinase, a novel drug target, in non-small cell lung carcinoma patients. Molecular cancer therapeutics 5: 2905–2913.
    1. Glotsos D, Tohka J, Ravazoula P, Cavouras D, Nikiforidis G (2005) Automated diagnosis of brain tumours astrocytomas using probabilistic neural network clustering and support vector machines. Int J Neural Syst 15: 1–11.
    1. Mattfeldt T, Gottfried HW, Wolter H, Schmidt V, Kestler HA, et al. (2003) Classification of prostatic carcinoma with artificial neural networks using comparative genomic hybridization and quantitative stereological data. Pathol Res Pract 199: 773–784.
    1. Zander T, Hofmann A, Staratschek-Jox A, Classen S, Debey-Pascher S, et al. (2011) Blood-based gene expression signatures in non-small cell lung cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 17: 3360–3367.
    1. Parry RM, Jones W, Stokes TH, Phan JH, Moffitt RA, et al. (2010) k-Nearest neighbor models for microarray gene expression analysis and clinical outcome prediction. The pharmacogenomics journal 10: 292–309.
    1. Demsar J, Zupan B, Kattan MW, Beck JR, Bratko I (1999) Naive Bayesian-based nomogram for prediction of prostate cancer recurrence. Studies in health technology and informatics 68: 436–441.
    1. Nebozhyn M, Loboda A, Kari L, Rook AH, Vonderheid EC, et al. (2006) Quantitative PCR on 5 genes reliably identifies CTCL patients with 5% to 99% circulating tumor cells with 90% accuracy. Blood 107: 3189–3196. Epub 2006 Jan 3110.
    1. Stridsberg M, Oberg K, Li Q, Stridsberg M, Eriksson B, et al. (1995) Measurement of chromogranin A, chromogranin B (secretogranin I), chromogranin C (secretogranin II) and pancreastatin in plasma and urine from patients with carcinoid tumours and endocrine pancreatic tumours. Journal of Endocrinology 144: 49–59.
    1. Modlin IM, Gustafsson BI, Moss SF, Pavel M, Tsolakis AV, et al. (2010) Chromogranin A–biological function and clinical utility in neuro endocrine tumor disease. Ann Surg Oncol 17: 2427–2443.
    1. Jianu CS, Fossmark R, Syversen U, Hauso O, Waldum HL (2010) A meal test improves the specificity of chromogranin A as a marker of neuroendocrine neoplasia. Tumour Biol 31: 373–380. Epub 2010 May 2018.
    1. Namwongprom S, Wong FC, Tateishi U, Kim EE, Boonyaprapa S (2008) Correlation of chromogranin A levels and somatostatin receptor scintigraphy findings in the evaluation of metastases in carcinoid tumors. Ann Nucl Med 22: 237–243. Epub 2008 Jun 2006.
    1. Arnold R, Wilke A, Rinke A, Mayer C, Kann PH, et al. (2008) Plasma chromogranin A as marker for survival in patients with metastatic endocrine gastroenteropancreatic tumors. Clin Gastroenterol Hepatol 6: 820–827. Epub 2008 Jun 2010.
    1. Sciarra A, Monti S, Gentile V, Salciccia S, Gomez AM, et al. (2005) Chromogranin A expression in familial versus sporadic prostate cancer. Urology 66: 1010–1014.
    1. Giusti M, Sidoti M, Augeri C, Rabitti C, Minuto F (2004) Effect of short-term treatment with low dosages of the proton-pump inhibitor omeprazole on serum chromogranin A levels in man. Eur J Endocrinol 150: 299–303.
    1. Hsiao RJ, Mezger MS, O’Connor DT (1990) Chromogranin A in uremia: progressive retention of immunoreactive fragments. Kidney Int 37: 955–964.
    1. Campana D, Nori F, Piscitelli L, Morselli-Labate AM, Pezzilli R, et al. (2007) Chromogranin A: is it a useful marker of neuroendocrine tumors? J Clin Oncol 25: 1967–1973.
    1. Chou WC, Hung YS, Hsu JT, Chen JS, Lu CH, et al. (2012) Chromogranin A is a reliable biomarker for gastroenteropancreatic neuroendocrine tumors in an Asian population of patients. Neuroendocrinology 95: 344–350 doi:. Epub 000332012 Feb 000333814
    1. Yates DR, Roupret M, Drouin SJ, Comperat E, Ricci S, et al. (2012) Quantitative RT-PCR analysis of PSA and prostate-specific membrane antigen mRNA to detect circulating tumor cells improves recurrence-free survival nomogram prediction after radical prostatectomy. Prostate 6: 22488.
    1. Mead R, Duku M, Bhandari P, Cree IA (2011) Circulating tumour markers can define patients with normal colons, benign polyps, and cancers. Br J Cancer 105: 239–245 doi: Epub 2011 Jun 1028
    1. Bai VU, Hwang O, Divine GW, Barrack ER, Menon M, et al. (2012) Averaged differential expression for the discovery of biomarkers in the blood of patients with prostate cancer. PLoS One 7: e34875. Epub 32012 Apr 34876.
    1. Alymani NA, Smith MD, Williams DJ, Petty RD (2010) Predictive biomarkers for personalised anti-cancer drug use: discovery to clinical implementation. Eur J Cancer 46: 869–879.
    1. Schuster R, Max N, Mann B, Heufelder K, Thilo F, et al. (2004) Quantitative real-time RT-PCR for detection of disseminated tumor cells in peripheral blood of patients with colorectal cancer using different mRNA markers. Int J Cancer 108: 219–227.
    1. Wu DY, Ugozzoli L, Pal BK, Qian J, Wallace RB (1991) The effect of temperature and oligonucleotide primer length on the specificity and efficiency of amplification by the polymerase chain reaction. DNA Cell Biol 10: 233–238.
    1. Dingemans AM, Brakenhoff RH, Postmus PE, Giaccone G (1997) Detection of cytokeratin-19 transcripts by reverse transcriptase-polymerase chain reaction in lung cancer cell lines and blood of lung cancer patients. Lab Invest 77: 213–220.
    1. Zippelius A, Kufer P, Honold G, Kollermann MW, Oberneder R, et al. (1997) Limitations of reverse-transcriptase polymerase chain reaction analyses for detection of micrometastatic epithelial cancer cells in bone marrow. J Clin Oncol 15: 2701–2708.
    1. Henke W, Loening SA (1998) Detection of illegitimate transcripts of prostate-specific antigen mRNA in blood by reverse transcription-polymerase chain reaction. Int J Cancer 77: 164–165.
    1. Lambrechts AC, van ’t Veer LJ, Rodenhuis S (1998) The detection of minimal numbers of contaminating epithelial tumor cells in blood or bone marrow: use, limitations and future of RNA-based methods. Ann Oncol 9: 1269–1276.
    1. Bustin SA (2002) Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems. J Mol Endocrinol 29: 23–39.
    1. Bustin SA (2000) Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays. J Mol Endocrinol 25: 169–193.
    1. Schmittgen TD, Zakrajsek BA, Mills AG, Gorn V, Singer MJ, et al. (2000) Quantitative reverse transcription-polymerase chain reaction to study mRNA decay: comparison of endpoint and real-time methods. Anal Biochem 285: 194–204.
    1. Ding C, Cantor CR (2003) A high-throughput gene expression analysis technique using competitive PCR and matrix-assisted laser desorption ionization time-of-flight MS. Proc Natl Acad Sci U S A 100: 3059–3064. Epub 2003 Mar 3056.
    1. Liu W, Saint DA (2002) Validation of a quantitative method for real time PCR kinetics. Biochem Biophys Res Commun 294: 347–353.
    1. Lekanne Deprez RH, Fijnvandraat AC, Ruijter JM, Moorman AF (2002) Sensitivity and accuracy of quantitative real-time polymerase chain reaction using SYBR green I depends on cDNA synthesis conditions. Anal Biochem 307: 63–69.
    1. Keilholz U, Willhauck M, Rimoldi D, Brasseur F, Dummer W, et al. (1998) Reliability of reverse transcription-polymerase chain reaction (RT-PCR)-based assays for the detection of circulating tumour cells: a quality-assurance initiative of the EORTC Melanoma Cooperative Group. Eur J Cancer 34: 750–753.
    1. Jung R, Ahmad-Nejad P, Wimmer M, Gerhard M, Wagener C, et al. (1997) Quality management and influential factors for the detection of single metastatic cancer cells by reverse transcriptase polymerase chain reaction. Eur J Clin Chem Clin Biochem 35: 3–10.
    1. Schittek B, Blaheta HJ, Florchinger G, Sauer B, Garbe C (1999) Increased sensitivity for the detection of malignant melanoma cells in peripheral blood using an improved protocol for reverse transcription-polymerase chain reaction. Br J Dermatol 141: 37–43.
    1. Saveanu A, Muresan M, De Micco C, Taieb D, Germanetti AL, et al. (2011) Expression of somatostatin receptors, dopamine D(2) receptors, noradrenaline transporters, and vesicular monoamine transporters in 52 pheochromocytomas and paragangliomas. Endocr Relat Cancer 18: 287–300 doi:. Print 2011 Apr.
    1. Reubi JC, Waser B (2003) Concomitant expression of several peptide receptors in neuroendocrine tumours: molecular basis for in vivo multireceptor tumour targeting. Eur J Nucl Med Mol Imaging 30: 781–793. Epub 2003 Apr 2018.
    1. Dilley WG, Kalyanaraman S, Verma S, Cobb JP, Laramie JM, et al. (2005) Global gene expression in neuroendocrine tumors from patients with the MEN1 syndrome. Mol Cancer 4: 9.
    1. Lapuk AV, Wu C, Wyatt AW, McPherson A, McConeghy BJ, et al. (2012) From sequence to molecular pathology, and a mechanism driving the neuroendocrine phenotype in prostate cancer. J Pathol 227: 286–297 doi:.
    1. Sun MH (2004) Neuroendocrine differentiation in sporadic CRC and hereditary nonpolyosis colorectal cancer. Dis Markers 20: 283–288.
    1. Indinnimeo M, Cicchini C, Memeo L, Stazi A, Provenza C, et al. (2002) Correlation between chromogranin-A expression and pathological variables in human colon carcinoma. Anticancer Res 22: 395–398.
    1. Langfelder P, Zhang B, Horvath S (2008) Defining clusters from a hierarchical cluster tree: the Dynamic Tree Cut package for R. Bioinformatics. 24: 719–720.

Source: PubMed

3
Iratkozz fel