Abstract

Recently we identified a molecular basis for differentiating benign and malignant follicular thyroid tumors. The purpose of these studies was to determine whether molecular analysis can be used to differentiate papillary thyroid carcinomas from benign thyroid nodules. Gene expression patterns of 14 papillary thyroid carcinomas and 21 benign tumors were analyzed by oligonucleotide array analysis. The carcinomas included seven classical papillary thyroid carcinomas (PTC) and seven follicular variant of PTC (FVPTC), and the benign tumors included 14 follicular adenomas and seven hyperplastic nodules. A hierarchical clustering analysis was performed to examine the groups for potential differences. The combined PTC and FVPTC groups had a distinct gene expression profile compared with the benign lesions. The sensitivity for a diagnosis of carcinoma was 93%, with a 100% specificity (one FVPTC clustered with the benign nodules). Cancer gene profiles contained both known (Met and galectin-3) and previously unidentified genes. Gene profiling is a reliable means of distinguishing PTC, FVPTC, and benign tumors of the thyroid. These gene profiles may provide insight into the pathogenesis of papillary thyroid carcinoma and may ultimately enhance the preoperative diagnosis of thyroid nodules on a molecular basis.

PAPILLARY THYROID CANCER (PTC) is the most common thyroid malignancy, with an estimated incidence of approximately 16,000 cases/yr in the U.S. (13). Nevertheless, benign thyroid nodules are far more common, and distinguishing benign and malignant thyroid nodules is a common diagnostic dilemma (47). The introduction of fine needle aspiration (FNA) in the 1970s simplified the evaluation of thyroid nodules and decreased the likelihood of surgery for a thyroid nodule by approximately 50% (8). Despite the ability to identify PTC with some reliability on FNA, clinicians frequently have to make a decision regarding the care of patients with thyroid nodules on the basis of equivocal information. For PTC, this diagnostic dilemma is particularly evident in the evaluation of nodules that are potentially follicular variant of papillary thyroid carcinoma (FVPTC) (4, 5). Given such difficulties, these studies were undertaken to analyze PTC, both classical and FVPTC, to determine whether molecular profiling can distinguish these cancers from benign thyroid tumors.

Most large studies have found that between 5 and 29% of thyroid nodules will be deemed a follicular lesion on FNA (6, 9). Many of the nuclear findings associated with PTC, including nuclear grooves, hypochromasia, and pear-shaped or ovoid nuclei, are not readily apparent on cytology due to artifact or sampling of an area that does not include nuclei with those changes (9, 10). Recently, studies evaluating follicular lesions on FNA have found upward of 21% of follicular lesions to be follicular variant of PTC (FVPTC) (5, 7). Baloch et al. (5) noted that 30% of lesions that are FVPTC cannot be diagnosed by FNA. Recent data suggest that FVPTC may be the dominant malignant phenotype of follicular lesions seen in FNA (5, 9, 11). Thus, better techniques are needed to differentiate FVPTC from benign follicular lesions.

Many attempts have been made to identify markers for thyroid carcinoma that can preoperatively distinguish between benign and malignant lesions. Most have not been proven to be specific enough, including PPAR-γ, Met, galectin-3, hTERT, and HBME-1 (1219). Recently, our laboratory reported the ability to differentiate follicular thyroid carcinoma from follicular adenoma (FA) by means of molecular profiling as well as use this technique to elucidate novel genes that may play an important role in the tumorigenesis of thyroid carcinoma (20). This study reports the ability to differentiate PTC from benign nodules using microarray technology and molecular profiling. This technology may ultimately prove useful in the diagnostic evaluation of thyroid nodules.

Materials and Methods

Tissue procurement

Thyroid tumor tissue was obtained at surgery from patients undergoing thyroidectomy (including hemi, subtotal, and total thyroidectomy). A pathologist dissected the tissue, and a small tumor tissue block from the dominant or suspicious nodule was snap-frozen in liquid nitrogen and stored at −80 C. The size and location of tumor samples were recorded in detail. All tumor samples were obtained with permission of and in accordance with the guidelines of our institutional review board, and informed consent was obtained from all patients. Histological classification was confirmed, the diagnosis was obtained from the final pathology report, and this diagnosis was reviewed and confirmed by an endocrine pathologist.

RNA extraction, purification, labeling, and hybridization

The methods described by Barden et al. (20) were employed for RNA extraction, purification, labeling, and hybridization. In brief, frozen tumor tissue was homogenized by sonication in TRIzol reagent (Invitrogen, Carlsbad, CA), and total RNA was prepared according to the manufacturer’s specifications. A total of 42 samples were analyzed by gene chip array (GeneChip Hu95 array, Affymetrix, Inc., Santa Clara, CA). The carcinoma samples included seven PTC and seven FVPTC. The benign samples consisted of 14 FA and seven hyperplastic nodules. An additional seven unknown samples were processed (blinded to preparer). All samples were processed in the same manner following the Affymetrix protocol. cDNA was synthesized from 8 μg sample RNA using T7 (dT)24 primer (GENSET Corp., La Jolla, CA). Second strand cDNA was then produced and purified. Biotin-labeled cRNA was made and used for hybridization to the Affymetrix oligonucleotide arrays. A sample aliquot was first hybridized to an Affymetrix test chip to confirm that the cRNA quality was adequate. All samples were of good quality. After staining with streptavidin-phycoerythrin, the chips were scanned in an HP ChipScanner (Affymetrix, Inc.) to detect hybridization signals.

Data analysis

The data were analyzed using MicroArray Suite version 5.0 (Affymetrix, Inc.). The intensity of each probe set of the array was captured, and the average intensity was calculated. Quantitative expression levels were calculated using intrachip-positive controls. Normalization of data was performed to account for variability in hybridization among duplicate probe sets and other hybridization artifacts. Transcripts were designated reliably detected (present) or not detected (absent) using the above analysis.

Data analysis was performed to identify genes that were differentially expressed between the papillary carcinoma (PTC and FVPTC) and benign groups (FA and hyperplasia). Data from the 21 benign tumors and 14 carcinomas that comprised the training set were used. First, the data were screened to identify signals counted as present by the Affymetrix software. These results were exported to GeneSpring (Silicon Genetics, Redwood City, CA), then analyzed with a parametric t test and multiple testing correction (Benjamini and Hochberg False Discovery Rate, with the P value set at <0.05), producing a gene list of 1149 genes differentially expressed. This list of 1149 differentially expressed genes was then used for unsupervised hierarchical clustering and statistical analysis. Cluster analysis was used to group the tumors based upon their similarities measured across the expression of 1149 genes.

To determine whether FVPTC could be differentiated from benign thyroid nodules, a second analysis, comparing only FVPTC tumors to benign thyroid nodules, was performed to identify genes differentially expressed between these groups. The data were exported to GeneSpring (Silicon Genetics) and using a nonparametric t test with a P value set at less than 0.01, the data were screened to produce a gene list of 843 differentially expressed genes between FVPTC and benign lesions. Finally, a similar analysis was performed to compare PTC to benign lesions. A total of 483 genes were differentially expressed between PTC and benign lesions. As described above, these gene lists were used for unsupervised hierarchical clustering and statistical analysis.

Evaluation of unknown samples

Once the hierarchical cluster analysis was established using gene expression profiles of differentially expressed genes in 35 tumors, the same analysis was performed on seven thyroid tumors (one PTC, four FVPTC, and two hyperplastic nodules) with investigators blinded to the final diagnosis. Gene profiles of the seven unknown tumors were produced using the 1149 differentially expressed genes for comparison. The unknown sample profiles were then added to the original 35 samples to create a combined group of 42 samples, which then underwent an unsupervised hierarchical clustering analysis.

To confirm the validity of the subset analysis of FVPTC vs. benign tumors, the unknown samples’ individual gene profiles were analyzed (excluding the one classical PTC), using the 843 differentially expressed genes produced by the analysis of FVPTC vs. benign tumors. The gene profiles of unknown samples combined with the test set (FVPTC vs. benign) samples underwent an unsupervised hierarchical cluster analysis, producing a dendrogram for the 34 samples. This same technique was repeated to analyze the PTC vs. benign tumor gene list. Unknown samples’ gene expression profiles were analyzed (excluding the four FVPTC) using the gene profile for 438 genes differentially expressed in the PTC vs. benign analysis. Once again the unknown samples were combined with the PTC vs. benign samples, and an unsupervised hierarchical clustering was performed. A dendrogram of the 31 samples (PTC vs. benign plus unknowns) was produced.

Semiquantitative RT-PCR

To verify the results of the Affymetrix gene chip array, a total of five genes were chosen that had expression levels with more than 2-fold difference, were implicated in the molecular pathogenesis of cancer, and had readily available primers or were made from the known mRNA sequences using Primer3 (21). One microgram of sample RNA was reverse transcribed with oligo(dT) primer in a total volume of 50 μl. A 1-μl aliquot of cDNA was used for PCR, and the product was electrophoresed in a 2.0% agarose gel and visualized with ethidium bromide and UV light. The band intensity for each sample was calculated using EagleSight software (version 3.2, Stratagene, La Jolla, CA), and a ratio of the intensity of the gene of interest to that of the housekeeping gene was calculated for each sample. These normalized intensity levels were then analyzed using a t test. The genes and primers are listed in Table 1.

TABLE 1.

RT-PCR genes and primers

 SenseAntisenseSize (bp)Ref. no.
AdrenomedullinAAGAAGTGGAATAAGTGGGCTTGGCTTAGAAGACACCAGAGT41040
TROP-2CCAGTTCCTTGATCTCCACCTTCTTCTGCTCCACGCTGACCTCCAAGTGT75354
METGATTTTAGTCATCCCAATGTCCATCCAGCATACAGTTTCTTGC22655
NRP-2CAAACACTGTGGGAACATCGTTCTCAGGAAACCCAGGAGA19843
TrefoilTTCAAGCCCCTGACTAGGAAGAGCATGGGACCTTTATTCG205 
GAPDHGAAGGTGAAGGTCGGAGTCGAAGATGGTGATGGGATTTC225 
 SenseAntisenseSize (bp)Ref. no.
AdrenomedullinAAGAAGTGGAATAAGTGGGCTTGGCTTAGAAGACACCAGAGT41040
TROP-2CCAGTTCCTTGATCTCCACCTTCTTCTGCTCCACGCTGACCTCCAAGTGT75354
METGATTTTAGTCATCCCAATGTCCATCCAGCATACAGTTTCTTGC22655
NRP-2CAAACACTGTGGGAACATCGTTCTCAGGAAACCCAGGAGA19843
TrefoilTTCAAGCCCCTGACTAGGAAGAGCATGGGACCTTTATTCG205 
GAPDHGAAGGTGAAGGTCGGAGTCGAAGATGGTGATGGGATTTC225 

GAPDH, Glyceraldehyde-3-phosphate dehydrogenase

TABLE 1.

RT-PCR genes and primers

 SenseAntisenseSize (bp)Ref. no.
AdrenomedullinAAGAAGTGGAATAAGTGGGCTTGGCTTAGAAGACACCAGAGT41040
TROP-2CCAGTTCCTTGATCTCCACCTTCTTCTGCTCCACGCTGACCTCCAAGTGT75354
METGATTTTAGTCATCCCAATGTCCATCCAGCATACAGTTTCTTGC22655
NRP-2CAAACACTGTGGGAACATCGTTCTCAGGAAACCCAGGAGA19843
TrefoilTTCAAGCCCCTGACTAGGAAGAGCATGGGACCTTTATTCG205 
GAPDHGAAGGTGAAGGTCGGAGTCGAAGATGGTGATGGGATTTC225 
 SenseAntisenseSize (bp)Ref. no.
AdrenomedullinAAGAAGTGGAATAAGTGGGCTTGGCTTAGAAGACACCAGAGT41040
TROP-2CCAGTTCCTTGATCTCCACCTTCTTCTGCTCCACGCTGACCTCCAAGTGT75354
METGATTTTAGTCATCCCAATGTCCATCCAGCATACAGTTTCTTGC22655
NRP-2CAAACACTGTGGGAACATCGTTCTCAGGAAACCCAGGAGA19843
TrefoilTTCAAGCCCCTGACTAGGAAGAGCATGGGACCTTTATTCG205 
GAPDHGAAGGTGAAGGTCGGAGTCGAAGATGGTGATGGGATTTC225 

GAPDH, Glyceraldehyde-3-phosphate dehydrogenase

Protein purification and Western blotting

Samples were chosen for immunoblot assay based on adequate tumor tissue availability. Frozen tissue was thawed in ice-cold homogenization buffer containing 50 mm Tris-buffered saline (pH 7.4), 2 mm EGTA, 150 mm NaCl, 2 mm MgCl2, 1 mm diethyldithiocarbamate, 1 mm phenylmethylsulfonylfluoride, 10 μg/ml leupeptin, 10 μg/ml aprotinin, 5 μg/ml pepstatin, 3 mm hydrogen peroxide, 50 mm sodium fluoride, 1 mm sodium orthovandate, 10 mm sodium molybdate (Sigma-Aldrich Corp., St. Louis, MO), 10% glycerol (Invitrogen), and 1% Triton-X (Bio-Rad Laboratories, Hercules, CA). Tissues were homogenized using a glass-on-glass tissue homogenizer. Homogenates were centrifuged at 11,750 × g for 10 min at 4 C to remove the particulate material. The protein concentration of the supernatant was measured using the Lowry protein assay kit (Sigma-Aldrich Corp.). Immunoblot analysis for neuropilin-2 (NRP2) was performed using a monoclonal antibody to NRP2 (catalogue no. SC13117, Santa Cruz Biotechnology, Inc., Santa Cruz, CA). One hundred micrograms per lane of protein from tissue were loaded on a 12% SDS-PAGE gel; after transfer, the membrane was blocked in 3% BSA overnight and incubated for 3 h with primary antibody. β-Actin was used as an internal control (Sigma-Aldrich Corp.).

Results

Differentially expressed genes

The level of gene expression in PTC samples (classical and follicular variant) was compared with gene expression in all benign tissue samples (FA and hyperplasia). The resulting gene expression profiles generated a list of differentially expressed genes. A total of 1149 genes were differentially expressed, of which 262 were greater than 2-fold differentially expressed. Seventy-six genes were relatively overexpressed in the carcinoma samples, and 186 genes were relatively overexpressed in the benign tissues (Table 2).

TABLE 2.

Gene list

Gene nameSymbolFold changeAccession no.
Overexpressed in carcinomas   
    GA733-1 protein precursorTROP211.4J04152
    Similar to LDL receptor-like protein: tSXV motifMEGF78.6AB011540
    Tenascin-CTNC7.9X78565
    FibronectinFN17.3M10905
    Dioxin-inducible cytochrome P450GLC3A7.3U03688
    α1-AntitrypsinAAT6.9X01683
    Melanocyte-specific gene 1MSG16.3U65092
    Intermediate conductance calcium-activated potassium channelhSK45.3AF022797
    Dual specificity protein phosphataseHVH35.2U15932
    NRP-2NRP24.9D83018
    MAP kinase phosphataseMKP-24.7U48807
    Keratin 19KRT194.7Y00503
    S plasma proteinPROS4.5M15036
    Galectin-3GALBP4.4AB006780
    Glutamine cyclotransferaseGCT4.2X71125
    Tissue inhibitor of metalloproteinasesTIMP4.1D11139
    G Protein-coupled receptorGABABR24.0AF056085
    Human cytokine (GRO-β)MIP2A3.9M36820
    Precursor alkaline phosphataseHUSI-I3.8X04470
    TGFαTGFA3.8X70340
    Signal sequence for secretion by HUVECSIG133.5AF015287
    DystrophinDMD3.4M18533
    KJAA0942 proteinKIAA09423.4AB023159
    LipocortinLPC13.3X05908
    Virally, encoded semaphorin protein receptorPLXN-C13.3AF030339
    AdrenomedullinAM3.3D14874
    Preprocathepsin HCPSB3.1X16832
    Breast epithelial antigen 46BA463.0U58516
    SkyRSE2.9D17517
    Ryudocan core proteinSYND42.9D79206
    Skeletal muscle ryanodine receptor geneCCO2.9U48508
    Epidermal growth factor receptor kinase substrateEps82.9U12535
    MET protooncogene proteinHGFR2.7J02958
    HER3 protein precursorHER32.7M34309
    Cathepsin CCPP12.6X87212
    Nuclear factor RIP140RIP1402.5X84373
    TGFβ-stimulated clone 22TSC-222.5AJ222700
    α1 (VIII) collagenCOL8A12.5X57527
    Aryl hydrocarbon receptorAHR2.4L19872
    SOX-4 proteinSOX-42.4X70683
    Aldehyde dehydrogenaseALDH42.2U10868
    Small GTP-binding proteinRAB272.2U57094
    N-Acetylglucosamine-6-O-sulfotransferaseGN6ST2.2AB014679
    Single-strand DNA-binding proteinHSPC1162.1AL080076
    GM3 synthaseSIATGM3S2.1AB018356
    MHC class II HLA-DQ-betaHLA-DQ-β2.1M81141
    Dihydrouracil dehydrogenaseDPD2.1U20938
    Serine protease hepsinTMPRSS12.1X07732
Overexpressed in benign tumors   
    Collagen α3 type IXEDM355.7L41162
    Intestinal trefoil factor, humanTFF37.6AI985964
    Hair keratinhHb57.3X99140
    Creatine kinase BCKBB5.7X15334
    Thyroid peroxidaseTPX5.4J02969
    Cellular retinoic acid-binding protein-1CRABPI5.2S74445
    Thrombospondin-4THBS44.5Z19585
    IgG Fc-binding proteinFC(γ)BP4.1D84239
    Dystrophin-binding intracellular membrane-associated muscle proteinSNT14.0U40571
    Creatine kinase BCKBB3.9X15334
    Cocaine- and amphetamine-regulated transcripthCART3.9U20325
    Recombination-activating protein-2RAG23.7M94633
    GTPase-activating proteinrap1GAP3.5M64788
    Matrilin-2 precursorMATN23.5U69263
    Human metallothionein-9fMT13.4M10943
    SURF-1SURF13.3Z35093
    Hepatic dihydrodiol dehydrogenaseDDH3.3U05861
    Aldolase AALDOA3.2X05236
Gene nameSymbolFold changeAccession no.
Overexpressed in carcinomas   
    GA733-1 protein precursorTROP211.4J04152
    Similar to LDL receptor-like protein: tSXV motifMEGF78.6AB011540
    Tenascin-CTNC7.9X78565
    FibronectinFN17.3M10905
    Dioxin-inducible cytochrome P450GLC3A7.3U03688
    α1-AntitrypsinAAT6.9X01683
    Melanocyte-specific gene 1MSG16.3U65092
    Intermediate conductance calcium-activated potassium channelhSK45.3AF022797
    Dual specificity protein phosphataseHVH35.2U15932
    NRP-2NRP24.9D83018
    MAP kinase phosphataseMKP-24.7U48807
    Keratin 19KRT194.7Y00503
    S plasma proteinPROS4.5M15036
    Galectin-3GALBP4.4AB006780
    Glutamine cyclotransferaseGCT4.2X71125
    Tissue inhibitor of metalloproteinasesTIMP4.1D11139
    G Protein-coupled receptorGABABR24.0AF056085
    Human cytokine (GRO-β)MIP2A3.9M36820
    Precursor alkaline phosphataseHUSI-I3.8X04470
    TGFαTGFA3.8X70340
    Signal sequence for secretion by HUVECSIG133.5AF015287
    DystrophinDMD3.4M18533
    KJAA0942 proteinKIAA09423.4AB023159
    LipocortinLPC13.3X05908
    Virally, encoded semaphorin protein receptorPLXN-C13.3AF030339
    AdrenomedullinAM3.3D14874
    Preprocathepsin HCPSB3.1X16832
    Breast epithelial antigen 46BA463.0U58516
    SkyRSE2.9D17517
    Ryudocan core proteinSYND42.9D79206
    Skeletal muscle ryanodine receptor geneCCO2.9U48508
    Epidermal growth factor receptor kinase substrateEps82.9U12535
    MET protooncogene proteinHGFR2.7J02958
    HER3 protein precursorHER32.7M34309
    Cathepsin CCPP12.6X87212
    Nuclear factor RIP140RIP1402.5X84373
    TGFβ-stimulated clone 22TSC-222.5AJ222700
    α1 (VIII) collagenCOL8A12.5X57527
    Aryl hydrocarbon receptorAHR2.4L19872
    SOX-4 proteinSOX-42.4X70683
    Aldehyde dehydrogenaseALDH42.2U10868
    Small GTP-binding proteinRAB272.2U57094
    N-Acetylglucosamine-6-O-sulfotransferaseGN6ST2.2AB014679
    Single-strand DNA-binding proteinHSPC1162.1AL080076
    GM3 synthaseSIATGM3S2.1AB018356
    MHC class II HLA-DQ-betaHLA-DQ-β2.1M81141
    Dihydrouracil dehydrogenaseDPD2.1U20938
    Serine protease hepsinTMPRSS12.1X07732
Overexpressed in benign tumors   
    Collagen α3 type IXEDM355.7L41162
    Intestinal trefoil factor, humanTFF37.6AI985964
    Hair keratinhHb57.3X99140
    Creatine kinase BCKBB5.7X15334
    Thyroid peroxidaseTPX5.4J02969
    Cellular retinoic acid-binding protein-1CRABPI5.2S74445
    Thrombospondin-4THBS44.5Z19585
    IgG Fc-binding proteinFC(γ)BP4.1D84239
    Dystrophin-binding intracellular membrane-associated muscle proteinSNT14.0U40571
    Creatine kinase BCKBB3.9X15334
    Cocaine- and amphetamine-regulated transcripthCART3.9U20325
    Recombination-activating protein-2RAG23.7M94633
    GTPase-activating proteinrap1GAP3.5M64788
    Matrilin-2 precursorMATN23.5U69263
    Human metallothionein-9fMT13.4M10943
    SURF-1SURF13.3Z35093
    Hepatic dihydrodiol dehydrogenaseDDH3.3U05861
    Aldolase AALDOA3.2X05236
TABLE 2.

Gene list

Gene nameSymbolFold changeAccession no.
Overexpressed in carcinomas   
    GA733-1 protein precursorTROP211.4J04152
    Similar to LDL receptor-like protein: tSXV motifMEGF78.6AB011540
    Tenascin-CTNC7.9X78565
    FibronectinFN17.3M10905
    Dioxin-inducible cytochrome P450GLC3A7.3U03688
    α1-AntitrypsinAAT6.9X01683
    Melanocyte-specific gene 1MSG16.3U65092
    Intermediate conductance calcium-activated potassium channelhSK45.3AF022797
    Dual specificity protein phosphataseHVH35.2U15932
    NRP-2NRP24.9D83018
    MAP kinase phosphataseMKP-24.7U48807
    Keratin 19KRT194.7Y00503
    S plasma proteinPROS4.5M15036
    Galectin-3GALBP4.4AB006780
    Glutamine cyclotransferaseGCT4.2X71125
    Tissue inhibitor of metalloproteinasesTIMP4.1D11139
    G Protein-coupled receptorGABABR24.0AF056085
    Human cytokine (GRO-β)MIP2A3.9M36820
    Precursor alkaline phosphataseHUSI-I3.8X04470
    TGFαTGFA3.8X70340
    Signal sequence for secretion by HUVECSIG133.5AF015287
    DystrophinDMD3.4M18533
    KJAA0942 proteinKIAA09423.4AB023159
    LipocortinLPC13.3X05908
    Virally, encoded semaphorin protein receptorPLXN-C13.3AF030339
    AdrenomedullinAM3.3D14874
    Preprocathepsin HCPSB3.1X16832
    Breast epithelial antigen 46BA463.0U58516
    SkyRSE2.9D17517
    Ryudocan core proteinSYND42.9D79206
    Skeletal muscle ryanodine receptor geneCCO2.9U48508
    Epidermal growth factor receptor kinase substrateEps82.9U12535
    MET protooncogene proteinHGFR2.7J02958
    HER3 protein precursorHER32.7M34309
    Cathepsin CCPP12.6X87212
    Nuclear factor RIP140RIP1402.5X84373
    TGFβ-stimulated clone 22TSC-222.5AJ222700
    α1 (VIII) collagenCOL8A12.5X57527
    Aryl hydrocarbon receptorAHR2.4L19872
    SOX-4 proteinSOX-42.4X70683
    Aldehyde dehydrogenaseALDH42.2U10868
    Small GTP-binding proteinRAB272.2U57094
    N-Acetylglucosamine-6-O-sulfotransferaseGN6ST2.2AB014679
    Single-strand DNA-binding proteinHSPC1162.1AL080076
    GM3 synthaseSIATGM3S2.1AB018356
    MHC class II HLA-DQ-betaHLA-DQ-β2.1M81141
    Dihydrouracil dehydrogenaseDPD2.1U20938
    Serine protease hepsinTMPRSS12.1X07732
Overexpressed in benign tumors   
    Collagen α3 type IXEDM355.7L41162
    Intestinal trefoil factor, humanTFF37.6AI985964
    Hair keratinhHb57.3X99140
    Creatine kinase BCKBB5.7X15334
    Thyroid peroxidaseTPX5.4J02969
    Cellular retinoic acid-binding protein-1CRABPI5.2S74445
    Thrombospondin-4THBS44.5Z19585
    IgG Fc-binding proteinFC(γ)BP4.1D84239
    Dystrophin-binding intracellular membrane-associated muscle proteinSNT14.0U40571
    Creatine kinase BCKBB3.9X15334
    Cocaine- and amphetamine-regulated transcripthCART3.9U20325
    Recombination-activating protein-2RAG23.7M94633
    GTPase-activating proteinrap1GAP3.5M64788
    Matrilin-2 precursorMATN23.5U69263
    Human metallothionein-9fMT13.4M10943
    SURF-1SURF13.3Z35093
    Hepatic dihydrodiol dehydrogenaseDDH3.3U05861
    Aldolase AALDOA3.2X05236
Gene nameSymbolFold changeAccession no.
Overexpressed in carcinomas   
    GA733-1 protein precursorTROP211.4J04152
    Similar to LDL receptor-like protein: tSXV motifMEGF78.6AB011540
    Tenascin-CTNC7.9X78565
    FibronectinFN17.3M10905
    Dioxin-inducible cytochrome P450GLC3A7.3U03688
    α1-AntitrypsinAAT6.9X01683
    Melanocyte-specific gene 1MSG16.3U65092
    Intermediate conductance calcium-activated potassium channelhSK45.3AF022797
    Dual specificity protein phosphataseHVH35.2U15932
    NRP-2NRP24.9D83018
    MAP kinase phosphataseMKP-24.7U48807
    Keratin 19KRT194.7Y00503
    S plasma proteinPROS4.5M15036
    Galectin-3GALBP4.4AB006780
    Glutamine cyclotransferaseGCT4.2X71125
    Tissue inhibitor of metalloproteinasesTIMP4.1D11139
    G Protein-coupled receptorGABABR24.0AF056085
    Human cytokine (GRO-β)MIP2A3.9M36820
    Precursor alkaline phosphataseHUSI-I3.8X04470
    TGFαTGFA3.8X70340
    Signal sequence for secretion by HUVECSIG133.5AF015287
    DystrophinDMD3.4M18533
    KJAA0942 proteinKIAA09423.4AB023159
    LipocortinLPC13.3X05908
    Virally, encoded semaphorin protein receptorPLXN-C13.3AF030339
    AdrenomedullinAM3.3D14874
    Preprocathepsin HCPSB3.1X16832
    Breast epithelial antigen 46BA463.0U58516
    SkyRSE2.9D17517
    Ryudocan core proteinSYND42.9D79206
    Skeletal muscle ryanodine receptor geneCCO2.9U48508
    Epidermal growth factor receptor kinase substrateEps82.9U12535
    MET protooncogene proteinHGFR2.7J02958
    HER3 protein precursorHER32.7M34309
    Cathepsin CCPP12.6X87212
    Nuclear factor RIP140RIP1402.5X84373
    TGFβ-stimulated clone 22TSC-222.5AJ222700
    α1 (VIII) collagenCOL8A12.5X57527
    Aryl hydrocarbon receptorAHR2.4L19872
    SOX-4 proteinSOX-42.4X70683
    Aldehyde dehydrogenaseALDH42.2U10868
    Small GTP-binding proteinRAB272.2U57094
    N-Acetylglucosamine-6-O-sulfotransferaseGN6ST2.2AB014679
    Single-strand DNA-binding proteinHSPC1162.1AL080076
    GM3 synthaseSIATGM3S2.1AB018356
    MHC class II HLA-DQ-betaHLA-DQ-β2.1M81141
    Dihydrouracil dehydrogenaseDPD2.1U20938
    Serine protease hepsinTMPRSS12.1X07732
Overexpressed in benign tumors   
    Collagen α3 type IXEDM355.7L41162
    Intestinal trefoil factor, humanTFF37.6AI985964
    Hair keratinhHb57.3X99140
    Creatine kinase BCKBB5.7X15334
    Thyroid peroxidaseTPX5.4J02969
    Cellular retinoic acid-binding protein-1CRABPI5.2S74445
    Thrombospondin-4THBS44.5Z19585
    IgG Fc-binding proteinFC(γ)BP4.1D84239
    Dystrophin-binding intracellular membrane-associated muscle proteinSNT14.0U40571
    Creatine kinase BCKBB3.9X15334
    Cocaine- and amphetamine-regulated transcripthCART3.9U20325
    Recombination-activating protein-2RAG23.7M94633
    GTPase-activating proteinrap1GAP3.5M64788
    Matrilin-2 precursorMATN23.5U69263
    Human metallothionein-9fMT13.4M10943
    SURF-1SURF13.3Z35093
    Hepatic dihydrodiol dehydrogenaseDDH3.3U05861
    Aldolase AALDOA3.2X05236
TABLE 2A.

Continued

    Protein C precursorPROC3.2X02750
    Human IgG Fc receptorFCGRT3.1U12255
    Dipeptidyl aminopeptidase-like proteinDPPX3.1M96860
    Angiopoietin-1ANG13.1U83508
    Inhibitor of DNA binding 1Idt3.0X77956
    Metallothionein-1AMT12.9K01383
    SelenocysteineTXD122.9AF093774
    Heart (R)-3-hydroxybutyrate dehydrogenaseBDH2.9M93107
    Glutathione transferase-ζ1GSTZ12.9U86529
    Metallothionein IGMT12.8J03910
    KIAA0777 gene productKIAA07772.8AF049884
    Metallothionein IIIGIFB2.8M93311
    Very-low-density lipoprotein receptorVLDLR2.7D16532
    Plasma glutamate carboxypeptidasePGCP2.7W28330
    Protein kinase X1PKX12.7X85545
    Endoplasmic reticulum-associated amyloid-β-peptide-binding proteinERAB2.7AF035555
    KIAA0281 geneKIAA02812.7D87457
    25-Hydroxyvitamin D3 1-α-hydroxylasePDDR2.7AB005038
    Extracellular proteinase inhibitor homologueHE42.6X63187
    Ribosomal protein L26RPL262.6X69392
    Human homolog of a mouse imprinted gene, Peg3KIAA02872.6AB006625
    K1AA0062 geneKIAA00622.6D31887
    Fibrillin-like (S1-5)FBNL2.6U03877
    Membrane-type matrix metalloproteinase 2MT2-MMP2.6Z48482
    Human ATX1 homologHAH12.6U70660
    Pituitary tumor-transforming genePTTG12.5AA203476
    Tetranectin (plasminogen-kringle-4-binding protein)TN2.5X64559
    Human homolog of mouse Tis11d early response geneERF22.5X78992
    Human selenium-binding proteinHSBP2.5U29091
    Carbonic anhydrase IVCAIV2.5M83670
    17β-Hydroxysteroid dehydrogenase type 3EDH17B32.5U05659
    Skeletal muscle LIM protein 1SLIM12.5AF063002
    PLA2 receptor precursorPLA2R2.5U17034
    KIAA0403 mRNAKIAA04032.5AB007863
    Human metallothionein 1EMTIE2.4R92331
    Metallothionein 1BMTI2.4M13485
    Monocyte chemotactic protein-2MCP-22.4Y16645
    A28-RGS14p mRNAA28-RGS142.4U70426
    Type II transmembrane protein: Golga enzymeTPST22.4AF049891
    Secreted cyclophilin-like proteinCYPB2.3M63573
    Nuclear aconitaseACO22.3NM_001098
    Pax8PAX82.3X69699
    FK506-binding proteinFKBP122.3M34539
    Electron transfer flavoprotein β-subunitETFB2.3X71129
    Leukemia inhibitory factor receptorLIFR2.3X61615
    T Complex-associated testis-expressed 1-like 1CW-12.3D50663
    Mitochondrial ATP synthase c subunitATP5G2.3X69907
    Translation initiation factor e1F-2B β-subunitEIF2B22.3AF035280
    Type 1 inositol 1,4,5-trisphosphate receptorINSP3R12.3D26070
    PepsinogenPEP2.3J00287
    Associated with mitotic spindle apparatusDEEPEST2.3AF063308
    Ribonuclease ARNS12.3D26129
    Synaptic glycoprotein 2SC22.2AF038958
    Myo-inositol monophosphatase 2IMPA22.2AF014398
    Histone H1xH1FX2.2D64142
    P43EFTU2.2S75463
    Protein kinase CθPKC2.2L07032
    Human phosphogluconate dehydrogenasehPGDH2.2U30255
    ProlylcarboxypeptidasePCP2.1L13977
    Phospholipase CPLC-L2.1D42108
    Cyclophilin isoformCYP32.1M80254
    Phosphatidylinositol 4,5-bisphosphate 5-phosphatase homologPIPP2.1U45975
    Phosphoribosylpyrophosphate synthetasePRS 12.1D00860
    Fibroblast growth factor receptorK-sam-12.1M87770
    Cytochrome e-1CYC12.1J04444
    MegalinGP3302.1U33837
    Lysosomal α-glucosidaseLYAG2.0X55079
    Protein C precursorPROC3.2X02750
    Human IgG Fc receptorFCGRT3.1U12255
    Dipeptidyl aminopeptidase-like proteinDPPX3.1M96860
    Angiopoietin-1ANG13.1U83508
    Inhibitor of DNA binding 1Idt3.0X77956
    Metallothionein-1AMT12.9K01383
    SelenocysteineTXD122.9AF093774
    Heart (R)-3-hydroxybutyrate dehydrogenaseBDH2.9M93107
    Glutathione transferase-ζ1GSTZ12.9U86529
    Metallothionein IGMT12.8J03910
    KIAA0777 gene productKIAA07772.8AF049884
    Metallothionein IIIGIFB2.8M93311
    Very-low-density lipoprotein receptorVLDLR2.7D16532
    Plasma glutamate carboxypeptidasePGCP2.7W28330
    Protein kinase X1PKX12.7X85545
    Endoplasmic reticulum-associated amyloid-β-peptide-binding proteinERAB2.7AF035555
    KIAA0281 geneKIAA02812.7D87457
    25-Hydroxyvitamin D3 1-α-hydroxylasePDDR2.7AB005038
    Extracellular proteinase inhibitor homologueHE42.6X63187
    Ribosomal protein L26RPL262.6X69392
    Human homolog of a mouse imprinted gene, Peg3KIAA02872.6AB006625
    K1AA0062 geneKIAA00622.6D31887
    Fibrillin-like (S1-5)FBNL2.6U03877
    Membrane-type matrix metalloproteinase 2MT2-MMP2.6Z48482
    Human ATX1 homologHAH12.6U70660
    Pituitary tumor-transforming genePTTG12.5AA203476
    Tetranectin (plasminogen-kringle-4-binding protein)TN2.5X64559
    Human homolog of mouse Tis11d early response geneERF22.5X78992
    Human selenium-binding proteinHSBP2.5U29091
    Carbonic anhydrase IVCAIV2.5M83670
    17β-Hydroxysteroid dehydrogenase type 3EDH17B32.5U05659
    Skeletal muscle LIM protein 1SLIM12.5AF063002
    PLA2 receptor precursorPLA2R2.5U17034
    KIAA0403 mRNAKIAA04032.5AB007863
    Human metallothionein 1EMTIE2.4R92331
    Metallothionein 1BMTI2.4M13485
    Monocyte chemotactic protein-2MCP-22.4Y16645
    A28-RGS14p mRNAA28-RGS142.4U70426
    Type II transmembrane protein: Golga enzymeTPST22.4AF049891
    Secreted cyclophilin-like proteinCYPB2.3M63573
    Nuclear aconitaseACO22.3NM_001098
    Pax8PAX82.3X69699
    FK506-binding proteinFKBP122.3M34539
    Electron transfer flavoprotein β-subunitETFB2.3X71129
    Leukemia inhibitory factor receptorLIFR2.3X61615
    T Complex-associated testis-expressed 1-like 1CW-12.3D50663
    Mitochondrial ATP synthase c subunitATP5G2.3X69907
    Translation initiation factor e1F-2B β-subunitEIF2B22.3AF035280
    Type 1 inositol 1,4,5-trisphosphate receptorINSP3R12.3D26070
    PepsinogenPEP2.3J00287
    Associated with mitotic spindle apparatusDEEPEST2.3AF063308
    Ribonuclease ARNS12.3D26129
    Synaptic glycoprotein 2SC22.2AF038958
    Myo-inositol monophosphatase 2IMPA22.2AF014398
    Histone H1xH1FX2.2D64142
    P43EFTU2.2S75463
    Protein kinase CθPKC2.2L07032
    Human phosphogluconate dehydrogenasehPGDH2.2U30255
    ProlylcarboxypeptidasePCP2.1L13977
    Phospholipase CPLC-L2.1D42108
    Cyclophilin isoformCYP32.1M80254
    Phosphatidylinositol 4,5-bisphosphate 5-phosphatase homologPIPP2.1U45975
    Phosphoribosylpyrophosphate synthetasePRS 12.1D00860
    Fibroblast growth factor receptorK-sam-12.1M87770
    Cytochrome e-1CYC12.1J04444
    MegalinGP3302.1U33837
    Lysosomal α-glucosidaseLYAG2.0X55079
TABLE 2A.

Continued

    Protein C precursorPROC3.2X02750
    Human IgG Fc receptorFCGRT3.1U12255
    Dipeptidyl aminopeptidase-like proteinDPPX3.1M96860
    Angiopoietin-1ANG13.1U83508
    Inhibitor of DNA binding 1Idt3.0X77956
    Metallothionein-1AMT12.9K01383
    SelenocysteineTXD122.9AF093774
    Heart (R)-3-hydroxybutyrate dehydrogenaseBDH2.9M93107
    Glutathione transferase-ζ1GSTZ12.9U86529
    Metallothionein IGMT12.8J03910
    KIAA0777 gene productKIAA07772.8AF049884
    Metallothionein IIIGIFB2.8M93311
    Very-low-density lipoprotein receptorVLDLR2.7D16532
    Plasma glutamate carboxypeptidasePGCP2.7W28330
    Protein kinase X1PKX12.7X85545
    Endoplasmic reticulum-associated amyloid-β-peptide-binding proteinERAB2.7AF035555
    KIAA0281 geneKIAA02812.7D87457
    25-Hydroxyvitamin D3 1-α-hydroxylasePDDR2.7AB005038
    Extracellular proteinase inhibitor homologueHE42.6X63187
    Ribosomal protein L26RPL262.6X69392
    Human homolog of a mouse imprinted gene, Peg3KIAA02872.6AB006625
    K1AA0062 geneKIAA00622.6D31887
    Fibrillin-like (S1-5)FBNL2.6U03877
    Membrane-type matrix metalloproteinase 2MT2-MMP2.6Z48482
    Human ATX1 homologHAH12.6U70660
    Pituitary tumor-transforming genePTTG12.5AA203476
    Tetranectin (plasminogen-kringle-4-binding protein)TN2.5X64559
    Human homolog of mouse Tis11d early response geneERF22.5X78992
    Human selenium-binding proteinHSBP2.5U29091
    Carbonic anhydrase IVCAIV2.5M83670
    17β-Hydroxysteroid dehydrogenase type 3EDH17B32.5U05659
    Skeletal muscle LIM protein 1SLIM12.5AF063002
    PLA2 receptor precursorPLA2R2.5U17034
    KIAA0403 mRNAKIAA04032.5AB007863
    Human metallothionein 1EMTIE2.4R92331
    Metallothionein 1BMTI2.4M13485
    Monocyte chemotactic protein-2MCP-22.4Y16645
    A28-RGS14p mRNAA28-RGS142.4U70426
    Type II transmembrane protein: Golga enzymeTPST22.4AF049891
    Secreted cyclophilin-like proteinCYPB2.3M63573
    Nuclear aconitaseACO22.3NM_001098
    Pax8PAX82.3X69699
    FK506-binding proteinFKBP122.3M34539
    Electron transfer flavoprotein β-subunitETFB2.3X71129
    Leukemia inhibitory factor receptorLIFR2.3X61615
    T Complex-associated testis-expressed 1-like 1CW-12.3D50663
    Mitochondrial ATP synthase c subunitATP5G2.3X69907
    Translation initiation factor e1F-2B β-subunitEIF2B22.3AF035280
    Type 1 inositol 1,4,5-trisphosphate receptorINSP3R12.3D26070
    PepsinogenPEP2.3J00287
    Associated with mitotic spindle apparatusDEEPEST2.3AF063308
    Ribonuclease ARNS12.3D26129
    Synaptic glycoprotein 2SC22.2AF038958
    Myo-inositol monophosphatase 2IMPA22.2AF014398
    Histone H1xH1FX2.2D64142
    P43EFTU2.2S75463
    Protein kinase CθPKC2.2L07032
    Human phosphogluconate dehydrogenasehPGDH2.2U30255
    ProlylcarboxypeptidasePCP2.1L13977
    Phospholipase CPLC-L2.1D42108
    Cyclophilin isoformCYP32.1M80254
    Phosphatidylinositol 4,5-bisphosphate 5-phosphatase homologPIPP2.1U45975
    Phosphoribosylpyrophosphate synthetasePRS 12.1D00860
    Fibroblast growth factor receptorK-sam-12.1M87770
    Cytochrome e-1CYC12.1J04444
    MegalinGP3302.1U33837
    Lysosomal α-glucosidaseLYAG2.0X55079
    Protein C precursorPROC3.2X02750
    Human IgG Fc receptorFCGRT3.1U12255
    Dipeptidyl aminopeptidase-like proteinDPPX3.1M96860
    Angiopoietin-1ANG13.1U83508
    Inhibitor of DNA binding 1Idt3.0X77956
    Metallothionein-1AMT12.9K01383
    SelenocysteineTXD122.9AF093774
    Heart (R)-3-hydroxybutyrate dehydrogenaseBDH2.9M93107
    Glutathione transferase-ζ1GSTZ12.9U86529
    Metallothionein IGMT12.8J03910
    KIAA0777 gene productKIAA07772.8AF049884
    Metallothionein IIIGIFB2.8M93311
    Very-low-density lipoprotein receptorVLDLR2.7D16532
    Plasma glutamate carboxypeptidasePGCP2.7W28330
    Protein kinase X1PKX12.7X85545
    Endoplasmic reticulum-associated amyloid-β-peptide-binding proteinERAB2.7AF035555
    KIAA0281 geneKIAA02812.7D87457
    25-Hydroxyvitamin D3 1-α-hydroxylasePDDR2.7AB005038
    Extracellular proteinase inhibitor homologueHE42.6X63187
    Ribosomal protein L26RPL262.6X69392
    Human homolog of a mouse imprinted gene, Peg3KIAA02872.6AB006625
    K1AA0062 geneKIAA00622.6D31887
    Fibrillin-like (S1-5)FBNL2.6U03877
    Membrane-type matrix metalloproteinase 2MT2-MMP2.6Z48482
    Human ATX1 homologHAH12.6U70660
    Pituitary tumor-transforming genePTTG12.5AA203476
    Tetranectin (plasminogen-kringle-4-binding protein)TN2.5X64559
    Human homolog of mouse Tis11d early response geneERF22.5X78992
    Human selenium-binding proteinHSBP2.5U29091
    Carbonic anhydrase IVCAIV2.5M83670
    17β-Hydroxysteroid dehydrogenase type 3EDH17B32.5U05659
    Skeletal muscle LIM protein 1SLIM12.5AF063002
    PLA2 receptor precursorPLA2R2.5U17034
    KIAA0403 mRNAKIAA04032.5AB007863
    Human metallothionein 1EMTIE2.4R92331
    Metallothionein 1BMTI2.4M13485
    Monocyte chemotactic protein-2MCP-22.4Y16645
    A28-RGS14p mRNAA28-RGS142.4U70426
    Type II transmembrane protein: Golga enzymeTPST22.4AF049891
    Secreted cyclophilin-like proteinCYPB2.3M63573
    Nuclear aconitaseACO22.3NM_001098
    Pax8PAX82.3X69699
    FK506-binding proteinFKBP122.3M34539
    Electron transfer flavoprotein β-subunitETFB2.3X71129
    Leukemia inhibitory factor receptorLIFR2.3X61615
    T Complex-associated testis-expressed 1-like 1CW-12.3D50663
    Mitochondrial ATP synthase c subunitATP5G2.3X69907
    Translation initiation factor e1F-2B β-subunitEIF2B22.3AF035280
    Type 1 inositol 1,4,5-trisphosphate receptorINSP3R12.3D26070
    PepsinogenPEP2.3J00287
    Associated with mitotic spindle apparatusDEEPEST2.3AF063308
    Ribonuclease ARNS12.3D26129
    Synaptic glycoprotein 2SC22.2AF038958
    Myo-inositol monophosphatase 2IMPA22.2AF014398
    Histone H1xH1FX2.2D64142
    P43EFTU2.2S75463
    Protein kinase CθPKC2.2L07032
    Human phosphogluconate dehydrogenasehPGDH2.2U30255
    ProlylcarboxypeptidasePCP2.1L13977
    Phospholipase CPLC-L2.1D42108
    Cyclophilin isoformCYP32.1M80254
    Phosphatidylinositol 4,5-bisphosphate 5-phosphatase homologPIPP2.1U45975
    Phosphoribosylpyrophosphate synthetasePRS 12.1D00860
    Fibroblast growth factor receptorK-sam-12.1M87770
    Cytochrome e-1CYC12.1J04444
    MegalinGP3302.1U33837
    Lysosomal α-glucosidaseLYAG2.0X55079

Two-way cluster analysis

GeneSpring software produces a two-dimensional dendrogram that displays the relatedness of the tumor sample types by both the length and the subdivisions of its clustering. Two distinctly clustered groups are seen in the comparison of PTC (combined PTC and FVPTC) and benign groups (Fig. 1). The top cluster represents only carcinoma samples, whereas the bottom cluster, which represents the benign group, included one FVPTC. Thus, this analysis detected cancer with 93% sensitivity and 100% specificity.

Fig. 1.

Gene cluster 1. Dendrogram of cluster analysis of 14 PTCs (seven PTC and seven FVPTC) and 21 benign nodules (FAs and hyperplastic nodules) based upon the pattern of expression of 1149 differentially expressed genes. Red indicates relatively high expression; blue indicates low levels of expression. Sample names are listed at the right of the diagram.

Using the list of differentially expressed genes from the test sets, seven unknown samples were added and assigned to a group based on similarity to other samples by hierarchical cluster analysis. In the combined papillary vs. benign analysis, all seven of the unknown samples were correctly clustered (Fig. 2).

Fig. 2.

Gene cluster 1 and unknowns. Dendrogram of cluster analysis of 14 PTCs (classic and follicular variant), 21 benign nodules, and seven unknown samples (UNK 1–2 are hyperplastic nodules, UNK 3 is PTC, and UNK 4–7 are FVPTC) based upon the pattern of expression of 1149 differentially expressed genes. Red indicates relatively high expression; blue indicates low levels of expression. All unknowns clustered correctly. Sample names are listed at the right of the diagram.

Subset analyses of FVPTC vs. benign nodules and PTC vs. benign nodules was then performed to determine whether the gene profiles could discriminate between the two types of PTC and benign nodules independently. In the analysis of FVPTC vs. benign nodules, 10 of 11 FVPTC clustered together as a distinct cancer group (one FVPTC clustered with the benign nodules). All benign nodules clustered together as a group, yielding a sensitivity of 91% and a specificity of 100% for the diagnosis of cancer (Fig. 3). It is known that FVPTC can be a difficult diagnosis to make even on histopathological review. Thus, a second analysis of the FVPTC vs. benign samples was undertaken using the gene list generated from the classic PTC vs. benign analysis. This second analysis of the FVPTC vs. benign samples revealed results identical to the initial comparison (data not shown). Finally, cluster analysis of PTC vs. benign tumors was 100% sensitive and specific for the diagnosis of cancer (Fig. 4).

Fig. 3.

Gene cluster 2. Dendrogram of cluster analysis of 11 FVPTC (including three unknowns labeled UNK 4–6) and 23 benign nodules (FAs, hyperplastic nodules, and UNK 1–2) based upon the pattern of expression of 873 differentially expressed genes. Red indicates relatively high expression; blue indicates low levels of expression. Sample names are listed at the right of the diagram.

Fig. 4.

Gene cluster 3. Dendrogram of cluster analysis of eight PTC (including UNK 3) and 23 benign nodules (FAs, hyperplastic nodules, and UNK 1–2) based upon the pattern of expression of 483 differentially expressed genes. Red indicates relative high expression; blue indicates low levels of expression. Sample names are listed at the right of the diagram.

Corroboration of gene chip analysis

Validation of the gene chip analysis was performed by semiquantitative RT-PCR of five of the most highly differentially expressed genes (Table 2). A total of 26 of the 42 samples were tested, 14 papillary carcinomas (PTC and FVPTC), six FA, and six hyperplasia. The Met oncogene is a gene known to be involved in thyroid tumorigenesis; however, the remaining four genes have not been previously associated with thyroid carcinomas. For each gene tested, the level of expression by RT-PCR correlated with the data obtained by microarray analysis, and all differences seen were statistically significant (P < 0.05; Fig. 5). Additionally, differential expression of the protein NRP2 was confirmed by immunoblot (Fig. 6). The expression of NRP2 was greater in the papillary carcinomas (classic and follicular variant) than in the benign tumors.

Fig. 5.

Semiquantitative RT-PCR. Verification of highly differentially expressed genes specific for carcinoma or benign tissue. Adrenomedullin (AM), TROP-2, MET, and NRP2 were differentially overexpressed in the carcinomas. Trefoil (intestinal trefoil factor) was relatively overexpressed in benign nodules. Primers specific for glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a housekeeping gene, were used as controls. All carcinoma samples (PTC and FVPTC) and 12 of the benign nodules [FA and hyperplastic nodules (HYP)] are displayed. Band intensity levels were normalized to the housekeeping gene and compared using a t test. All differences seen are statistically significant (P < 0.05).

Fig. 6.

Immunoblot. The differential expression of NRP2 was confirmed by Western blot analysis. One hundred micrograms of protein per lane were loaded. Blot A, The first eight samples (left) are from carcinomas (PTC and FVPTC); the next eight samples (right) are from benign nodules (FA and hyperplastic nodules (HYP)]. Blot B, β-Actin was used as an internal control.

Discussion

PTC constitutes 80% of all differentiated thyroid carcinomas, accounting for almost 16,000 new cases of cancer each year (13). Here we have shown that molecular analysis by gene profiling can readily distinguish between PTC, both classical and follicular variant, and benign nodules. These data will provide insight into the molecular pathogenesis of PTCs and may ultimately offer new leads into the treatment of thyroid carcinomas. Additionally, it may provide a molecular basis for the final classification of thyroid tumors that are diagnostic dilemmas for even experienced endocrine pathologists. Finally, it can serve as a springboard for the advancement of diagnosis of thyroid nodules by FNA.

Currently, FNA is sensitive and specific for classic PTC, given the consistency of nuclear findings, including grooving and central clearing (9, 10). However, the ability to define FVPTC by FNA is difficult and limited (5, 911). Combining these difficulties with the problems with follicular lesions, FNA is not the optimal preoperative diagnostic tool. Attempts at finding a molecular marker that will readily distinguish benign from malignant thyroid lesions have previously fallen short of the mark. Although many important genetic alterations have been found, including galectin-3, PAX8-PPAR-γ, and MET/HGF receptor, most have not yet proven to be sensitive or specific enough to be translated into clinically useful markers (13, 1619). Molecular profiling of tumors allows for a quantitative analysis of a panel of genes that are differentially expressed in thyroid nodules, which may lead to the classification of thyroid nodules as either benign or malignant (22, 23). Because of the larger number of genes analyzed, we hypothesized that the profiles generated would overcome the major pitfall of single gene analysis, specifically that in all cases there is overlap of gene expression between benign and malignant tumors. The data presented suggest that our hypothesis is, in fact, true.

Comparison of PTC samples (PTC and FVPTC) with benign lesions by microarray analysis produced a gene list of 262 genes that were significantly differentially expressed. To validate the microarray analysis, we selected five of the 262 genes identified to be differentially expressed between PTC and benign thyroid lesions. Most of these genes have not previously been implicated in thyroid carcinoma pathogenesis and were selected due to their high level of differential expression. Four of these genes were confirmed to be preferentially expressed in PTC, and one was expressed preferentially in the benign tumors. One of the four genes preferentially expressed in papillary carcinoma, the Met gene, has been previously reported to be overexpressed in PTC by both classic molecular techniques and microarray analysis (22, 24). Intestinal trefoil factor, which we found to be overexpressed in the benign group, has previously been reported to be both a tumor suppressor as well as a tumor promoter in other epithelial cell lines (25, 26). The remaining three genes analyzed hold interesting potential as mediators of thyroid carcinogenesis, and these are reviewed below.

TROP2 is a transmembrane receptor protein that has a phosphatidylinositol 4,5-bisphosphate binding sequence on the intracellular portion and is thought to be a receptor-gated calcium channel, integral to calcium homeostasis and possibly cell cycle activation (2729). Recently, TROP2 has been found to be overexpressed in human epithelial cell tumors, including lung, colon, breast, and ovarian cancers (2832). Activation of the receptor causes an increase in intracellular calcium levels, and this Ca+ spike is postulated to be a mediator of uncontrolled cell growth (29).

Highly homologous to the cell-cell adhesion molecule TROP1, TROP2 may be involved in cell migration and possibly cell metastasis (29, 33). TROP1 has been used for immunotherapy in colon cancer patients, reducing mortality and relapse rates in resected colon cancer patients, signifying its role in epithelial cell tumors (34, 35). Recently, our laboratory has shown that TROP2 is not highly expressed in follicular thyroid carcinomas (data not shown). Most follicular thyroid carcinomas spread hematogenously, unlike PTC, which mainly metastasizes to lymph nodes (1, 3). Given this distribution of expression between tumors that metastasize differently, TROP2 may be a potential marker for lymphatic metastasis.

Adrenomedullin has been shown to play a role in cancer cell growth and survival (36). A potent angiogenic factor, adrenomedullin, has also been postulated to be a cell mitogen (37). Blocking the activity of adrenomedullin by the use of monoclonal antibodies has shown to suppress tumor cell growth in vitro (38). Recent clinical studies have shown that overexpression of adrenomedullin is correlated with poor prognosis in ovarian cancer (39). Previously our laboratory showed that adrenomedullin was up-regulated in follicular thyroid carcinomas and may be a potential marker for thyroid carcinomas in general (20). Other laboratories have found adrenomedullin abundantly expressed in epithelial tumor cell lines, including lung, breast, ovary, colon, and prostate (40).

Another proangiogenic receptor, NRP2, was found to be overexpressed in PTC. Initially thought only to be a receptor for the semaphorin-3 (SEMA3) family of proteins and expressed during axonal extension, NRP2 has recently been shown to also be a receptor for the vascular endothelial growth factor family of angiogenic factors and has been found to be expressed in NSCLC, osteosarcomas, and pancreatic islet cell and endocrine tumors (4145). Some postulate that SEMA3F and vascular endothelial growth factor compete for binding to NRP2, and a reduction in SEMA3F leads to increased vascularity, loss of apoptosis, and increased tumor growth (44, 46). Analysis of these three genes (TROP2, adrenomedullin, and NRP2) demonstrates the powerful potential that microarray analysis provides in the identification of new genes that contribute to the pathogenesis of thyroid carcinoma.

The ability to differentiate both classic PTC and FVPTC individually from benign thyroid lesions is clinically significant. Hyperplastic nodules are often difficult to distinguish from FAs on pathological review (47, 48). FVPTC is mistaken for adenomatous lesions, including hyperplasia, on both cytological and histological evaluation, making it difficult to distinguish from benign lesions even on permanent sectioning (4749). This difficulty has led to the use of multiple immunohistochemical markers, including cytokeratins, to help in differentiating FVPTC from benign lesions (50, 51). Although many markers are positive in FVPTC, they are also positive in benign lesions and ultimately do not significantly increase the sensitivity or specificity in diagnosing these difficult lesions (48, 50, 51). Our ability to differentiate FVPTC from all benign lesions based on the molecular profile with greater than 90% sensitivity and specificity may improve the future accuracy of a diagnosis of cancer.

We have shown in this paper that both classic papillary and the follicular variant of papillary thyroid tumors can be distinguished from benign thyroid nodules based on their molecular profile with excellent sensitivity and specificity. Although classification of tumors based upon their molecular profile is an emerging technology, it has significant clinical potential. Recently, microarray analysis has been performed on FNA biopsies of the breast with hopes of modifying treatment strategies based on FNA molecular analysis (52, 53). Similar potential exists for future preoperative molecular classification of thyroid nodules based on our findings.

This work was supported by a G. Tom Shires Faculty Scholar Award.

Abbreviations:

     
  • FA,

    Follicular adenoma;

  •  
  • FNA,

    fine needle aspiration;

  •  
  • FVPTC,

    follicular variant of papillary thyroid carcinoma;

  •  
  • NRP2,

    neuropilin-2;

  •  
  • PTC,

    papillary thyroid carcinoma;

  •  
  • SEMA3,

    semaphorin-3.

References

1

Sherman
SI
2003
Thyroid carcinoma.
Lancet
361
:
501
511

2

Wang
C
,
Crapo
LM
1997
The epidemiology of thyroid disease and implications for screening.
Endocrinol Metab Clin North Am
26
:
189
218

3

Gimm
O
2001
Thyroid cancer.
Cancer Lett
163
:
143
156

4

Caraway
NP
,
Sneige
N
,
Samaan
NA
1993
Diagnostic pitfalls in thyroid fine-needle aspiration: a review of 394 cases.
Diagn Cytopathol
9
:
345
350

5

Baloch
ZW
,
Fleisher
S
,
LiVolsi
VA
,
Gupta
PK
2002
Diagnosis of “follicular neoplasm:” a gray zone in thyroid fine-needle aspiration cytology.
Diagn Cytopathol
26
:
41
44

6

Baloch
ZW
,
Sack
MJ
,
Yu
GH
,
Livolsi
VA
,
Gupta
PK
1998
Fine-needle aspiration of thyroid: an institutional experience.
Thyroid
8
:
565
569

7

Yang
GC
,
Liebeskind
D
,
Messina
AV
2001
Ultrasound-guided fine-needle aspiration of the thyroid assessed by Ultrafast Papanicolaou stain: data from 1135 biopsies with a two- to six-year follow-up.
Thyroid
11
:
581
589

8

Hamberger
B
,
Gharib
H
,
Melton III
LJ
,
Goellner
JR
,
Zinsmeister
AR
1982
Fine-needle aspiration biopsy of thyroid nodules. Impact on thyroid practice and cost of care.
Am J Med
73
:
381
384

9

Greaves
TS
,
Olvera
M
,
Florentine
BD
,
Raza
AS
,
Cobb
CJ
,
Tsao-Wei
DD
,
Groshen
S
,
Singer
P
,
Lopresti
J
,
Martin
SE
2000
Follicular lesions of thyroid: a 5-year fine-needle aspiration experience.
Cancer
90
:
335
341

10

Mesonero
CE
,
Jugle
JE
,
Wilbur
DC
,
Nayar
R
1998
Fine-needle aspiration of the macrofollicular and microfollicular subtypes of the follicular variant of papillary carcinoma of the thyroid.
Cancer
84
:
235
244

11

Yang
GC
,
Liebeskind
D
,
Messina
AV
2003
Should cytopathologists stop reporting follicular neoplasms on fine-needle aspiration of the thyroid?
Cancer
99
:
69
74

12

Aogi
K
,
Kitahara
K
,
Buley
I
,
Backdahl
M
,
Tahara
H
,
Sugino
T
,
Tarin
D
,
Goodison
S
1998
Telomerase activity in lesions of the thyroid: application to diagnosis of clinical samples including fine-needle aspirates.
Clin Cancer Res
4
:
1965
1970

13

Kroll
TG
,
Sarraf
P
,
Pecciarini
L
,
Chen
CJ
,
Mueller
E
,
Spiegelman
BM
,
Fletcher
JA
2000
PAX8-PPARγ1 fusion oncogene in human thyroid carcinoma.
Science
289
:
1357
1360

14

Inohara
H
,
Honjo
Y
,
Yoshii
T
,
Akahani
S
,
Yoshida
J
,
Hattori
K
,
Okamoto
S
,
Sawada
T
,
Raz
A
,
Kubo
T
1999
Expression of galectin-3 in fine-needle aspirates as a diagnostic marker differentiating benign from malignant thyroid neoplasms.
Cancer
85
:
2475
2484

15

Ippolito
A
,
Vella
V
,
La Rosa
GL
,
Pellegriti
G
,
Vigneri
R
,
Belfiore
A
2001
Immunostaining for Met/HGF receptor may be useful to identify malignancies in thyroid lesions classified suspicious at fine-needle aspiration biopsy.
Thyroid
11
:
783
787

16

Saggiorato
E
,
Cappia
S
,
De Giuli
P
,
Mussa
A
,
Pancani
G
,
Caraci
P
,
Angeli
A
,
Orlandi
F
2001
Galectin-3 as a presurgical immunocytodiagnostic marker of minimally invasive follicular thyroid carcinoma.
J Clin Endocrinol Metab
86
:
5152
5158

17

Casey
MB
,
Lohse
CM
,
Lloyd
RV
2003
Distinction between papillary thyroid hyperplasia and papillary thyroid carcinoma by immunohistochemical staining for cytokeratin 19, galectin-3, and HBME-1.
Endocr Pathol
14
:
55
60

18

Raphael
SJ
2002
The meanings of markers: ancillary techniques in diagnosis of thyroid neoplasia.
Endocr Pathol
13
:
301
311

19

Mase
T
,
Funahashi
H
,
Koshikawa
T
,
Imai
T
,
Nara
Y
,
Tanaka
Y
,
Nakao
A
2003
HBME-1 immunostaining in thyroid tumors especially in follicular neoplasm.
Endocr J
50
:
173
177

20

Barden
CB
,
Shister
KW
,
Zhu
B
,
Guiter
G
,
Greenblatt
DY
,
Zeiger
MA
,
Fahey III
TJ
2003
Classification of follicular thyroid tumors by molecular signature: results of gene profiling.
Clin Cancer Res
9
:
1792
1800

21

Rozen
S
,
Skaletsky
H
2000
Primer3 on the WWW for general users and for biologist programmers.
Methods Mol Biol
132
:
365
386

22

Huang
Y
,
Prasad
M
,
Lemon
WJ
,
Hampel
H
,
Wright
FA
,
Kornacker
K
,
LiVolsi
V
,
Frankel
W
,
Kloos
RT
,
Eng
C
,
Pellegata
NS
,
de la Chapelle
A
2001
Gene expression in papillary thyroid carcinoma reveals highly consistent profiles.
Proc Natl Acad Sci USA
98
:
15044
15049

23

Chen
KT
,
Lin
JD
,
Chao
TC
,
Hsueh
C
,
Chang
CA
,
Weng
HF
,
Chan
EC
2001
Identifying differentially expressed genes associated with metastasis of follicular thyroid cancer by cDNA expression array.
Thyroid
11
:
41
46

24

Trovato
M
,
Villari
D
,
Bartolone
L
,
Spinella
S
,
Simone
A
,
Violi
MA
,
Trimarchi
F
,
Batolo
D
,
Benvenga
S
1998
Expression of the hepatocyte growth factor and c-met in normal thyroid, non-neoplastic, and neoplastic nodules.
Thyroid
8
:
125
131

25

Leung
WK
,
Yu
J
,
Chan
FK
,
To
KF
,
Chan
MW
,
Ebert
MP
,
Ng
EK
,
Chung
SC
,
Malfertheiner
P
,
Sung
JJ
2002
Expression of trefoil peptides (TFF1, TFF2, and TFF3) in gastric carcinomas, intestinal metaplasia, and non-neoplastic gastric tissues.
J Pathol
197
:
582
588

26

Uchino
H
,
Kataoka
H
,
Itoh
H
,
Hamasuna
R
,
Koono
M
2000
Overexpression of intestinal trefoil factor in human colon carcinoma cells reduces cellular growth in vitro and in vivo.
Gastroenterology
118
:
60
69

27

El Sewedy
T
,
Fornaro
M
,
Alberti
S
1998
Cloning of the murine TROP2 gene: conservation of a PIP2-binding sequence in the cytoplasmic domain of TROP-2.
Int J Cancer
75
:
324
330

28

Fornaro
M
,
Dell’Arciprete
R
,
Stella
M
,
Bucci
C
,
Nutini
M
,
Capri
MG
,
Alberti
S
1995
Cloning of the gene encoding Trop-2, a cell-surface glycoprotein expressed by human carcinomas.
Int J Cancer
62
:
610
618

29

Ripani
E
,
Sacchetti
A
,
Corda
D
,
Alberti
S
1998
Human Trop-2 is a tumor-associated calcium signal transducer.
Int J Cancer
76
:
671
676

30

Miotti
S
,
Canevari
S
,
Menard
S
,
Mezzanzanica
D
,
Porro
G
,
Pupa
SM
,
Regazzoni
M
,
Tagliabue
E
,
Colnaghi
MI
1987
Characterization of human ovarian carcinoma-associated antigens defined by novel monoclonal antibodies with tumor-restricted specificity.
Int J Cancer
39
:
297
303

31

Fradet
Y
,
Cordon-Cardo
C
,
Thomson
T
,
Daly
ME
,
Whitmore Jr
WF
,
Lloyd
KO
,
Melamed
MR
,
Old
LJ
1984
Cell surface antigens of human bladder cancer defined by mouse monoclonal antibodies.
Proc Natl Acad Sci USA
81
:
224
228

32

Gerritsen
ME
,
Soriano
R
,
Yang
S
,
Ingle
G
,
Zlot
C
,
Toy
K
,
Winer
J
,
Draksharapu
A
,
Peale
F
,
Wu
TD
,
Williams
PM
2002
In silico data filtering to identify new angiogenesis targets from a large in vitro gene profiling data set.
Physiol Genomics
10
:
13
20

33

Mangino
G
,
Grazia Capri
M
,
Barnaba
V
,
Alberti
S
2002
Presentation of native TROP-2 tumor antigens to human cytotoxic T lymphocytes by engineered antigen-presenting cells.
Int J Cancer
101
:
353
359

34

Trebak
M
,
Begg
GE
,
Chong
JM
,
Kanazireva
EV
,
Herlyn
D
,
Speicher
DW
2001
Oligomeric state of the colon carcinoma-associated glycoprotein GA733–2 (Ep-CAM/EGP40) and its role in GA733-mediated homotypic cell-cell adhesion.
J Biol Chem
276
:
2299
2309

35

Kirman
I
,
Maydelman
A
,
Asi
Z
,
Whelan
RL
2003
Effect of surgical trauma on epithelial cell adhesion molecule (GA-733) vaccine-induced tumor resistance.
Surg Endosc
17
:
505
509

36

Cuttitta
F
,
Pio
R
,
Garayoa
M
,
Zudaire
E
,
Julian
M
,
Elsasser
TH
,
Montuenga
LM
,
Martinez
A
2002
Adrenomedullin functions as an important tumor survival factor in human carcinogenesis.
Microsc Res Tech
57
:
110
119

37

Zudaire
E
,
Martinez
A
,
Cuttitta
F
2003
Adrenomedullin and cancer.
Regul Pept
112
:
175
183

38

Ouafik
L
,
Sauze
S
,
Boudouresque
F
,
Chinot
O
,
Delfino
C
,
Fina
F
,
Vuaroqueaux
V
,
Dussert
C
,
Palmari
J
,
Dufour
H
,
Grisoli
F
,
Casellas
P
,
Brunner
N
,
Martin
PM
2002
Neutralization of adrenomedullin inhibits the growth of human glioblastoma cell lines in vitro and suppresses tumor xenograft growth in vivo.
Am J Pathol
160
:
1279
1292

39

Hata
K
,
Takebayashi
Y
,
Akiba
S
,
Fujiwaki
R
,
Iida
K
,
Nakayama
K
,
Nakayama
S
,
Fukumoto
M
,
Miyazaki
K
2000
Expression of the adrenomedullin gene in epithelial ovarian cancer.
Mol Hum Reprod
6
:
867
872

40

Miller
MJ
,
Martinez
A
,
Unsworth
EJ
,
Thiele
CJ
,
Moody
TW
,
Elsasser
T
,
Cuttitta
F
1996
Adrenomedullin expression in human tumor cell lines. Its potential role as an autocrine growth factor.
J Biol Chem
271
:
23345
23351

41

Klagsbrun
M
,
Takashima
S
,
Mamluk
R
2002
The role of neuropilin in vascular and tumor biology.
Adv Exp Med Biol
515
:
33
48

42

Handa
A
,
Tokunaga
T
,
Tsuchida
T
,
Lee
YH
,
Kijima
H
,
Yamazaki
H
,
Ueyama
Y
,
Fukuda
H
,
Nakamura
M
2000
Neuropilin-2 expression affects the increased vascularization and is a prognostic factor in osteosarcoma.
Int J Oncol
17
:
291
295

43

Kawakami
T
,
Tokunaga
T
,
Hatanaka
H
,
Kijima
H
,
Yamazaki
H
,
Abe
Y
,
Osamura
Y
,
Inoue
H
,
Ueyama
Y
,
Nakamura
M
2002
Neuropilin 1 and neuropilin 2 co-expression is significantly correlated with increased vascularity and poor prognosis in nonsmall cell lung carcinoma.
Cancer
95
:
2196
2201

44

Lantuejoul
S
,
Constantin
B
,
Drabkin
H
,
Brambilla
C
,
Roche
J
,
Brambilla
E
2003
Expression of VEGF, semaphorin SEMA3F, and their common receptors neuropilins NP1 and NP2 in preinvasive bronchial lesions, lung tumours, and cell lines.
J Pathol
200
:
336
347

45

Cohen
T
,
Herzog
Y
,
Brodzky
A
,
Greenson
JK
,
Eldar
S
,
Gluzman-Poltorak
Z
,
Neufeld
G
,
Resnick
MB
2002
Neuropilin-2 is a novel marker expressed in pancreatic islet cells and endocrine pancreatic tumours.
J Pathol
198
:
77
82

46

Bagnard
D
,
Vaillant
C
,
Khuth
ST
,
Dufay
N
,
Lohrum
M
,
Puschel
AW
,
Belin
MF
,
Bolz
J
,
Thomasset
N
2001
Semaphorin 3A-vascular endothelial growth factor-165 balance mediates migration and apoptosis of neural progenitor cells by the recruitment of shared receptor.
J Neurosci
21
:
3332
3341

47

Baloch
ZW
,
Livolsi
VA
2002
Follicular-patterned lesions of the thyroid: the bane of the pathologist.
Am J Clin Pathol
117
:
143
150

48

Kesmodel
SB
,
Terhune
KP
,
Canter
RJ
,
Mandel
SJ
,
Livolsi
VA
,
Baloch
ZW
,
Fraker
DL
2003
The diagnostic dilemma of follicular variant of papillary thyroid carcinoma.
Surgery
134
:
1005
1012

49

Baloch
ZW
,
Gupta
PK
,
Yu
GH
,
Sack
MJ
,
LiVolsi
VA
1999
Follicular variant of papillary carcinoma. Cytologic and histologic correlation.
Am J Clin Pathol
111
:
216
222

50

Miettinen
M
,
Kovatich
AJ
,
Karkkainen
P
1997
Keratin subsets in papillary and follicular thyroid lesions. A paraffin section analysis with diagnostic implications.
Virchows Arch
431
:
407
413

51

Baloch
ZW
,
Abraham
S
,
Roberts
S
,
LiVolsi
VA
1999
Differential expression of cytokeratins in follicular variant of papillary carcinoma: an immunohistochemical study and its diagnostic utility.
Hum Pathol
30
:
1166
1171

52

Pusztai
L
,
Ayers
M
,
Stec
J
,
Clark
E
,
Hess
K
,
Stivers
D
,
Damokosh
A
,
Sneige
N
,
Buchholz
TA
,
Esteva
FJ
,
Arun
B
,
Cristofanilli
M
,
Booser
D
,
Rosales
M
,
Valero
V
,
Adams
C
,
Hortobagyi
GN
,
Symmans
WF
2003
Gene expression profiles obtained from fine-needle aspirations of breast cancer reliably identify routine prognostic markers and reveal large-scale molecular differences between estrogen-negative and estrogen-positive tumors.
Clin Cancer Res
9
:
2406
2415

53

Symmans
WF
,
Ayers
M
,
Clark
EA
,
Stec
J
,
Hess
KR
,
Sneige
N
,
Buchholz
TA
,
Krishnamurthy
S
,
Ibrahim
NK
,
Buzdar
AU
,
Theriault
RL
,
Rosales
MF
,
Thomas
ES
,
Gwyn
KM
,
Green
MC
,
Syed
AR
,
Hortobagyi
GN
,
Pusztai
L
2003
Total RNA yield and microarray gene expression profiles from fine-needle aspiration biopsy and core-needle biopsy samples of breast carcinoma.
Cancer
97
:
2960
2971

54

Wonderling
RS
,
Owens
RA
1996
The Rep68 protein of adeno-associated virus type 2 stimulates expression of the platelet-derived growth factor B c-sis proto-oncogene.
J Virol
70
:
4783
4786

55

Shin
JH
,
Chung
J
,
Kim
HO
,
Kim
YH
,
Hur
YM
,
Rhim
JH
,
Chung
HK
,
Park
SC
,
Park
JG
,
Yang
HK
2002
Detection of cancer cells in peripheral blood of stomach cancer patients using RT-PCR amplification of tumour-specific mRNAs
.
Aliment Pharmacol Ther
16
(
Suppl 2
):
137
144