MOLECULAR SIGNATURE OF LIVER TUMOR GRADE AND USE TO EVALUATE PROGNOSIS AND THERAPEUTIC REGIMEN

Abstract
The present invention concerns a method to determine the gene expression profile on a sample previously obtained from a patient diagnosed for a liver tumor, comprising assaying the expression of a set of genes in this sample and determining the gene expression profile (signature). In a particular embodiment, said method enables to determine the grade of liver tumor, such as hepatoblastoma (HB) or a hepatocellular carcinoma (HCC). The invention is also directed to kits comprising a plurality of pairs of primers or a plurality of probes specific for a set of genes, as well as to solid support or composition comprising a set of probes specific for a set of genes. These methods are useful to determine the grade of a liver tumor in sample obtained from a patient, to determine the risk of developing metastasis and/or to define the therapeutic regimen to apply to a patient.
Description

The present invention relates to a method to in vitro determine the grade of a liver tumor in a sample previously obtained from a patient, using a molecular signature based on the expression of a set of genes comprising at least 2, especially has or consist of 2 to 16 genes, preferably a set of 16 genes. In a particular embodiment, the method focuses on hepatoblastoma (HB) or hepatocellular carcinoma (HCC), in adults or in children. The invention is also directed to sets of primers, sets of probes, compositions, kits or arrays, comprising primers or probes specific for a set of genes comprising at least 2 genes, especially has or consists of 2 to 16 genes, preferably exactly 16 genes. Said sets, kits and arrays are tools suitable to determine the grade of a liver tumor in a patient.


The liver is a common site of metastases from a variety of organs such as lung, breast, colon and rectum. However, liver is also a site of different kinds of cancerous tumors that start in the liver (primary liver cancers). The most frequent is the Hepatocellular Carcinoma (HCC) (about 3 out of 4 primary liver cancers are this type) and is mainly diagnosed in adults. In the United States approximately 10,000 new patients are diagnosed with hepatocellular carcinoma each year. Less frequent liver tumours are cholangiocarcinoma (CC) in adults and hepatoblastoma (HB) in children.


The prognosis and treatment options associated with these different kinds of cancers is difficult to predict, and is dependent in particular on the stage of the cancer (such as the size of the tumor, whether it affects part or all of the liver, has spread to other places in the body or its aggressiveness). Therefore, it is important for clinicians and physicians to establish a classification of primary liver cancers (HCC or HB) to propose the most appropriate treatment and adopt the most appropriate surgery strategy. Some factors are currently used (degree of local invasion, histological types of cancer with specific grading, tumour markers and general status of the patient) but have been found to not be accurate and sufficient enough to ensure a correct classification.


As far as the HB is concerned, the PRETEXT (pre-treatment extent of disease) system designed by the International Childhood Liver Tumor Strategy Group (SIOPEL) is a non invasive technique commonly used by clinicians, to assess the extent of liver cancer, to determine the time of surgery and to adapt the treatment protocol. This system is based on the division of the liver in four parts and the determination of the number of liver sections that are free of tumor (Aronson et al. 2005; Journal of Clinical Oncology; 23 (6): 1245-1252). A revised staging system taking into account other criteria, such as caudate lobe involvement, extrahepatic abdominal disease, tumor focality, tumor rupture or intraperitoneal haemorrhage, distant metastases, lymph node metastases, portal vein involvement and involvement of the IVC (inferior vena cava) and/or hepatic veins, has been recently proposed (Roebuck; 2007; Pediatr Radiol; 37: 123-132). However, the PRETEXT system, even if reproducible and providing good prognostic value, is based on imaging and clinical symptoms, making this system dependent upon the technicians and clinicians. There is thus a need for a system, complementary to the PRETEXT system, based on genetic and molecular features of the liver tumors.


The present invention concerns a method or process of profiling gene expression for a set of genes, in a sample previously obtained from a patient diagnosed for a liver tumor. In a particular embodiment said method is designed to determine the grade of a liver tumor in a patient.


By “liver tumor” or “hepatic tumor”, it is meant a tumor originating from the liver of a patient, which is a malignant tumor (comprising cancerous cells), as opposed to a benign tumor (non cancerous) which is explicitly excluded. Malignant liver tumors encompass two main kinds of tumors: hepatoblastoma (HB) or hepatocellular carcinoma (HCC). These two tumor types can be assayed for the presently reported molecular signature. However, the present method may also be used to assay malignant liver tumors which are classified as unspecified (non-HB, non-HCC).


The present method may be used to determine the grade of a liver tumor or several liver tumors of the same patient, depending on the extent of the liver cancer. For convenience, the expression “a liver tumor” will be used throughout the specification to possibly apply to “one or several liver tumor(s)”. The term “neoplasm” may also be used as a synonymous of “tumor”.


In a particular embodiment, the tumor whose grade has to be determined is located in the liver. The presence of the tumor(s) in the liver may be diagnosed by ultrasound scan, x-rays, blood test, CT scans (computerised tomography) and/or MRI scans (magnetic resonance imaging).


In a particular embodiment, the tumor, although originating from the liver, has extended to other tissues or has given rise to metastasis.


In a particular embodiment, the patient is a child i.e., a human host who is under 20 years of age according to the present application. Therefore, in a particular embodiment, the liver tumor is a paediatric HB or a paediatric HCC. In another embodiment, the liver tumor is an adult HCC.


A grade is defined as a subclass of the liver tumor, corresponding to prognostic factors, such as tumor status, liver function and general health status. The present method of the invention allows or at least contributes to differentiating liver tumors having a good prognosis from tumors with a bad prognosis, in terms of evolution of the patient's disease. A good prognosis tumor is defined as a tumor with good survival probability for the patient (more than 80% survival at two years for HB and more than 50% survival at two years for HCC), low probability of metastases and good response to treatment for the patient. In contrast, a bad prognosis tumor is defined as a tumor with an advanced stage, such as one having vascular invasion or/and extrahepatic metastasis, and associated with a low survival probability for the patient (less than 50% survival in two years).


The method of the invention is carried out on a sample isolated from the patient who has previously been diagnosed for the tumor(s) and who, optionally, may have been treated by surgery. In a preferred embodiment, the sample is the liver tumor (tumoral tissue) or of one of the liver tumors identified by diagnosis imaging and obtained by surgery or a biopsy of this tumor. The tumor located in the liver tumor is called the primary tumor.


In another embodiment, the sample is not the liver tumor, but is representative of this tumor. By “representative”, it is meant that the sample is regarded as having the same features as the primary tumors, when considering the gene expression profile assayed in the present invention. Therefore, the sample may also consist of metastatic cells (secondary tumors spread into different part(s) of the body) or of a biological fluid containing cancerous cells (such as blood).


The sample may be fixed, for example in formalin (formalin fixed). In addition or alternatively, the sample may be embedded in paraffin (paraffin-embedded) or equivalent products. In particular, the tested sample is a formalin-fixed, paraffin-embedded (FFPE) sample.


One advantage of the method of the present invention is that, despite the possible heterogeneity of some liver tumors (comprising epithelial tumor cells at different stages of liver differentiation within the same tumor), the assay has proved to be reproducible and efficient on liver tumor biopsies obtained from any part of the whole tumor. Therefore, there is no requirement for the isolation of cells presenting particular features except from the fact that they are obtained from a liver tumor or are representative thereof, to carry out the gene expression profile assay.


In a particular embodiment, the tumor originates from a patient having a Caucasian origin, in particular European, North American, Australian, New-Zealander or Afrikaners.


In a first step, the method or process of the invention comprises assaying the expression level of a set of genes in a sample, in order to get an expression profile thereof.


By “expression of a set of genes” (or “gene expression”), it is meant assaying, in particular detecting, the product or several products resulting from the expression of a gene, this product being in the form of a nucleic acid, especially RNA, mRNA, cDNA, polypeptide, protein or any other formats. In a particular embodiment, the assay of the gene expression profile comprises detecting a set of nucleotide targets, each nucleotide target corresponding to the expression product of a gene encompassed in the set.


The expression “nucleotide target” means a nucleic acid molecule whose expression must be measured, preferably quantitatively measured. By “expression measured”, it is meant that the expression product(s), in particular the transcription product(s) of a gene, are measured. By “quantitative” it is meant that the method is used to determine the quantity or the number of copies of the expression products, in particular the transcription products or nucleotide targets, originally present in the sample. This must be opposed to the qualitative measurement, whose aim is to determine the presence or absence of said expression product(s) only.


A nucleotide target is in particular a RNA, and most particularly a total RNA. In a preferred embodiment, the nucleotide target is mRNA or transcripts. According to the methods used to measure the gene expression level, the mRNA initially present in the sample may be used to obtain cDNA or cRNA, which is then detected and possibly measured.


In an embodiment, the expression of the gene is assayed directly on the sample, in particular in the tumor. In an alternative embodiment, the expression products or the nucleotide targets are prepared from the sample, in particular are isolated or even purified. When the nucleotide targets are mRNA, a further step comprising or consisting in the retro-transcription of said mRNA into cDNA (complementary DNA) may also be performed prior to the step of detecting expression. Optionally, the cDNA may also be transcribed in vitro to provide cRNA.


During the step of preparation, and before assaying the expression, the expression product(s) or the nucleotide target(s) may be labelled, with isotopic (such as radioactive) or non isotopic (such as fluorescent, coloured, luminescent, affinity, enzymatic, magnetic, thermal or electrical) markers or labels.


It is noteworthy that steps carried out for assaying the gene expression must not alter the qualitative or the quantitative expression (number of copies) of the expression product(s) or of the nucleotide target(s), or must not interfere with the subsequent step comprising assaying the qualitative or the quantitative expression of said expression product(s) or nucleotide target(s).


The step of profiling gene expression comprises determining the expression of a set of genes. Such a set is defined as a group of genes that must be assayed for one test, and especially performed at the same time, on the same patient's sample. A set comprises at least 2 and has especially from 2 to 16 genes, said 2 to 16 genes being chosen from the 16 following genes: alpha-fetoprotein (AFP), aldehyde dehydrogenase 2 (ALDH2), amyloid P component serum (APCS), apolipoprotein C-IV (APOC4), aquaporin 9 (AQP9), budding uninhibited by benzimidazoles 1 (BUB1), complement componant 1 (C1S), cytochrome p450 2E1 (CYP2E1), discs large homolog 7 (DLG7), dual specificity phosphatase 9 (DUSP9), E2F5 transcription factor (E2F5), growth hormone receptor (GHR), 4-hydroxyphenylpyruvase dioxygenase (HPD), immunoglogulin superfamily member 1 (IGSF1), Notchless homolog 1 (NLE1) and the ribosomal protein L10a (RPL10A) genes.


A complete description of these 16 genes is given in Table 1. This table lists, from left to right, the symbol of the gene, the complete name of the gene, the number of the SEQ ID provided in the sequence listing, the Accession Number from the NCBI database on June 2008, the human chromosomal location and the reported function (when known).


A set of genes comprises at least 2 out the 16 genes of Table 1, and particularly at least or exactly 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 out of the 16 genes of Table 1. In a particular embodiment, the set comprises or consists of the 16 genes of Table 1 i.e. the set of genes comprises or consists of AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes. Accordingly, unless otherwise stated when reference is made in the present application to a set of 2 to 16 genes of Table 1, it should be understood as similarly applying to any number of genes within said 2 to 16 range.


In other particular embodiments, the set of genes comprises or consists of one of the following sets: (a) the E2F5 and HPD genes, (b) the APCS, BUB1, E2F5, GHR and HPD genes, (c) the ALDH2, APCS, APOC4, BUB1, C1S, CYP2E1, E2F5, GHR and HPD genes, (d) the ALDH2, APCS, APOC4, AQP9, BUB1, C1S, DUSP9, E2F5 and RPL10A genes, or (e) the ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, E2F5, GHR, IGSF1 and RPL10A genes.


As indicated by the expression “comprises from 2 to 16 genes of Table 1”, the set may, besides the specific genes of Table 1, contain additional genes not listed in Table 1. This means that the set must comprises from 2 to 16 genes of Table 1, i.e. 2 to 16 genes of Table 1 (in particular 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 16 genes), and optionally comprises one or more additional genes. Said set may also be restricted to said 2 to 16 genes of Table 1.


Additional genes may be selected for the difference of expression observed between the various grades of liver cancer, in particular between a tumor of good prognosis and a tumor of poor prognosis.

TABLE 1mRNAAccessionProteinsymbolGene nameSEQ IDNoLocationFunctionSEQ IDAFPalpha-fetoprotein1NM_0011344q11-q13plasma protein synthesized2by the fetal liverALDH2aldehyde dehydrogenase 23NM_00069012q24.2liver enzyme involved in4family (mitochondrial)alcohol metabolismAPCSamyloid P component, serum5NM_0016391q21-q23secreted glycoprotein6AP0C4apolipoprotein C-IV7NM_00164619q13.2secreted liver protein8AQP9aquaporin 99NM_02098015q22.1-22.2water-selective membrane channel10BUB1BUB1 budding uninhibited11AF0432942q14kinase involved in spindle12by benzimidazoles 1 homologcheckpoint(yeast)C1Scomplement component 1, s13M1876712p13component of the cleavage and14subcomponentpolyadenylation specificityfactor complexCYP2E1cytochrome P450, family 2,15AF18227610q24.3-qtercytochrome P450 family membersubfamily E, polypeptide 1involved in drug metabolismDLG7discs, large homolog717NM_01475014q22.3cell cycle regulator involved18(Drosophila) (DLGAP5)in kinetocore formationDUSP9dual specificity phosphatase 919NM_001395Xq28phosphatase involved in20regulation of MAP KinasesE2F5E2F transcription factor 5,21U156428q21.2transcription factor involved in cell22p130-bindingcycle regulationGHRGrowth hormone receptor23NM_0001635p13-p12transmembrane receptor for24growth hormoneHPD4-hydroxyphenylpyruvate25NM_00215012q24-qterenzyme involved in amino-acid26dioxygenasedegradationIGSF1immunoglobulin superfamily,27NM_001555Xq25cell recognition and28member 1regulation of cell behaviorNLE1notchless homolog 129NM_0809617q12unknown30(Drosophila)RPL10Aribosomal protein L10a31NM_0071046p21.3-p21.2ribosomal protein of 60S subunit32


The invention also relates to a set of genes comprising or consisting of the 16 genes of Table 1 (i.e., AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes), in which 1, 2, 3, 4 or 5 genes out of the 16 genes are substituted by a gene presenting the same features in terms of difference of expression between a tumor of a good prognosis and a tumor of poor prognosis.


In a particular embodiment, the number of genes of the set does not exceed 100, particularly 50, 30, 20, more particularly 16 and even more particularly is maximum 5, 6, 7, 8, 9 or 10.


When considering adding or substituting a gene or several genes to the disclosed set, the person skilled in the art will consider one or several of the following features:

    • (a) the added gene(s) and/or the substituted gene(s) of Table 1 must present the same features in terms of difference of expression between a tumor of a good prognosis and a tumor of poor prognosis as the genes of Table 1 when taken as a whole. Thus, the expression of the added gene or of the substituted gene in a tumor of a good prognosis is either overexpressed or underexpressed of a factor of at least 2, preferably of at least 5, and more preferably of at least 10, as compared to its expression in a tumor of poor prognosis.
    • (b) besides presenting the feature in a), the added gene and/or the substituted gene may also provide, in combination with the other genes of the set, discriminant results with respect to the grade of the liver tumors; this discrimination is reflected by the homogeneity of expression profile of this gene in the tumors of a good prognosis on the one hand, and the tumors of poor prognosis in the other hand; and
    • (c) finally, besides features of a) and/or b), the added gene and/or the substituted gene is optionally chosen among genes that are involved in liver differentiation, in particular having a specific expression in fetal liver, or genes that are involved in proliferation, for example in mitosis or associated with ribosomes.


Examples of genes which can be added or may replace genes of the set may be identified in following Table 2.

TABLE 2list of genes according to p value.GenemeanmeanratioParametricsymbolrC1rC2rC2/rC1p-valueFDRDescriptionIPO4123.7248.32.02.00E−070.00036importin 4CPSF1467.81010.72.22.00E-070.00036cleavage and polyadenylation specificfactor 1, 160 kDaMCM425.890.73.51.10E−060.00115MCM4 minichromosome maintenancedeficient 4 (S. cerevisise)EIF3S313192601.22.01.20E−060.00119eukaryotic translation initiation factor 3,subunit 3 gamma, 40 kDaNCL13192655.62.01.30E−060.00122nucleolinCDC25C35.799.32.81.40E−060.00124cell division cycle 25CCENPA28.278.42.81.50E−060.00124centromere protein A, 17 kDaKIF1424.754.22.21.50E−060.00124kinesin family member 14IPW145.7397.62.71.90E−060.0015imprinted in Prader-Willi syndromeKNTC226.865.12.42.20E−060.00157kinetochore associated 2TMEM4826471.72.72.30E−060.00157transmembrane protein 48BOP187.2270.93.12.30E−060.00157block of proliferation 1EIF3S9170372.42.22.30E−060.00157eukaryotic translation initiation factor 3,subunit 9 eta, 116 kDaPH-4340.9168.20.52.40E−060.00158hypoxia-inducible factor prolyl 4-hydroxylaseSMC4L1151.5359.32.42.50E−060.0016SMC4 structural maintenance ofchromosomes 4-like 1 (yeast)TTK23.774.23.12.60E−060.00161TTK protein kinaseLAMA3696136.30.22.80E−060.00168laminin, alpha 3C10orf72192.667.70.42.90E−060.00169Chromosome 10 open reading frame 72TPX273.4401.55.53.10E−060.00171TPX2, microtubule-associated, homolog(Xenopus laevis)MSH275.5212.12.83.20E−060.00171mutS homolog 2, colon cancer,nonpolyposis type 1 (E. coli)DKC1358.1833.52.33.20E−060.00171dyskeratosis congenita 1, dyskerinSTK686.4395.34.63.30E−060.00172serine/threonine kinase 6CCT6A200.5526.62.63.50E−060.00173chaperonin containing TCP1, subunit 6A(zeta 1)SULT1C167.5314.84.73.50E−060.00173sulfotransferase family, cytosolic, 1C,member 1ILF3142.3294.52.13.70E−060.00174interleukin enhancer binding factor 3,90 kDaIMPDH2916.92385.62.63.70E−060.00174IMP (inosine monophosphate)dehydrogenase 2HIC263.4208.83.33.90E−060.00179hypermethylated in cancer 2AFM1310.3237.40.24.10E−060.00184afaminMCM7187.3465.32.54.30E−060.00189MCM7 minichromosome maintenancedeficient 7 (S. cerevisiae)CNAP170.2177.52.54.40E−060.00189chromosome condensation-related SMC-associated protein 1CBARA19584750.54.60E−060.00194calcium binding atopy-relatedautoantigen 1PLA2G4C123.351.20.44.90E−060.00194phospholipase A2, group IVC (cytosolic,calcium-independent)CPSF1301.96162.05.00E−060.00194cleavage and polyadenylation specificfactor 1, 160 kDaSNRPN30.9100.63.35.00E−060.00194Small nuclear ribonucleoproteinpolypeptide NRPL52754.849611.85.20E−060.00194ribosomal protein L5C1R1446.5366.40.35.30E−060.00194complement component 1, rsubcomponentC16orf34630.41109.61.85.30E−060.00194chromosome 16 open reading frame 34PHB309.3915.13.05.30E−060.00194prohibitinBZW2387.4946.42.45.40E−060.00194basic leucine zipper and W2 domains 2ALAS11075.8466.50.45.50E−060.00194aminolevulinate, delta-, synthase 1FLJ2036448.6112.42.35.70E−060.00198hypothetical protein FLJ20364RANBP1593.71168.12.05.90E−060.00201RAN binding protein 1SKB1354.7687.41.96.20E−060.00208SKB1 homolog (S. pombe)ABHD6402.2196.90.56.50E−060.00213abhydrolase domain containing 6CCNB160.43305.56.60E−060.00213cyclin B1NOL5A246.9716.22.97.00E−060.00213nucleolar protein 5A (56 kDa with KKE/Drepeat)RPL83805.77390.51.97.00E−060.00213ribosomal protein L8BLNK211.139.80.27.10E−060.00213B-cell linkerBYSL167.3269.71.67.10E−060.00213bystin-likeUBE1L247.6142.30.67.20E−060.00213ubiquitin-activating enzyme E1-likeCHD7118.63122.67.40E−060.00215chromodomain helicase DNA bindingprotein 7DKFZp762E170.2219.43.17.60E−060.00218hypothetical protein DKFZp762E1312312(HJURP)NUP210178.4284.91.87.70E−060.00218nucleoporin 210 kDaPLK172.8185.22.57.90E−060.0022polo-like kinase 1 (Drosophila)ENPEP116.229.40.38.00E−060.0022glutamyl aminopeptidase(aminopeptidase A)HCAP-G17.757.83.38.40E−060.00228chromosome condensation protein GUGT2B41117.8246.70.29.20E−060.00245UDP glucuronosyltransferase 2 family,polypeptide B4C20orf27129.7245.31.99.30E−060.00245chromosome 20 open reading frame 27C6orf149178.7491.12.79.40E−060.00245chromosome 6 open reading frame 149(LYRM4)
The Accession Numbers of the genes of Table 2. as found in NCBI database in June 2008, are the following: IPO4 (BC136759), CPSF1 (NM_013291), MCM4 (NM_005914.2; NM_182746.1; two accession numbers for the same gene correspond to 2 different isoforms of the gene), EIF3S3 (NM_003756.2), NCL (NM_005381.2), CDC25C (NM_001790.3), CENPA (NM_001809.3; NM_001042426.1), K1F14 (BC113742), IPW
# (U12897), KNTC2 (AK313184), TMEM48 (NM_018087), BOP1 (NM_015201), EIF3S9 (NM_003751; NM_001037283). PH-4 (NM _177939), SMC4L1 (NM_005496; NM_001002800), TTK (AK315696), LAMA3 (NM_198129), C10orf72 (NM_001031746; NM_144984), TPX2 (NM_012112), MSH2 (NM_000251), DKC1 (NM_001363), STK6 (AY892410), CCT6A (NM_001762; # NM_001009186), SULT1C1 (AK313193), ILF3 (NM_012218; NM_004516), IMPDH2 (NM_000884), HIC2 (NM_015094), AFM (NM_001133), MCM7 (NM_005916; NM_182776), CNAP1(AK128354), CBARA1 (AK225695), PLA2G4C (NM_003706), CPSF1(NM_013291), SNRPN (BC000611), RPL5 (AK314720), C1R (NM_001733), C16orf34 (CH471112), PHB (AK312649), BZW2 (BC017794), ALAS1(AK312566), # FLJ20364 (NM_017785), RANBP1 (NM_002882), SKB1 (AF015913), ABHD6 (NM_020676), CCNB1 (NM_031966), NOL5A (NM_006392), RPL8 (NM_000973; NM_033301), BLNK (NM_013314; NM_001114094), BYSL (NM_004053), UBE1L(AY889910), CHD7 (NM_017780), DKFZp762E1312 (NM_018410), NUP210(NM_024923), PLK1(NM_005030), ENPEP(NM_001977), # HCAP-G(NM_022346), UGT2B4 (NM_021139), C20orf27 (NM_001039140) and C6orf149 (NM_020408).


In a particular embodiment of the invention, the set of genes of the invention is designed to determine the grade of hepatoblastoma, in particular paediatric hepatoblastoma. In another embodiment, the set of genes is designed to determine the grade of hepatocellular carcinoma, in particular paediatric HCC or adult HCC.


The expression of the genes of the set may be assayed by any conventional methods, in particular any conventional methods known to measure the quantitative expression of RNA, preferably mRNA.


The expression may be measured after carrying out an amplification process, such as by PCR, quantitative PCR (qPCR) or real-time PCR. Kits designed for measuring expression after an amplification step are disclosed below.


The expression may be measured using hybridization method, especially with a step of hybridizing on a solid support, especially an array, a macroarray or a microarray or in other conditions especially in solution. Arrays and kits of the invention, designed for measuring expression by hybridization method are disclosed below.


The expression of a gene may be assayed in two manners:

    • to determine absolute gene expression that corresponds to the number of copies of the product of expression of a gene, in particular the number of copies of a nucleotide target, in the sample; and
    • to determine the relative expression that corresponds to the number of copies of the product of expression of a gene, in particular the number of copies of a nucleotide target, in the sample over the number of copies of the expression product or the number of copies of a nucleotide target of a different gene (calculation also known as normalisation). This different gene is not one of the genes contained in the set to be assayed. This different gene is assayed on the same sample and at the same time as the genes of the set to be assayed, and is called an invariant gene or a normalizer. The invariant gene is generally selected for the fact that its expression is steady whatever the sample to be tested. The expression “steady whatever the sample” means that the expression of an invariant gene does not vary significantly between a normal liver cell and the corresponding tumor cell in a same patient and/or between different liver tumor samples in a same patient. In the present specification, a gene is defined as invariant when its absolute expression does not vary in function of the grade of the liver tumors, in particular does not vary in function of the grade of the HB or HCC tumor, and/or does not vary between liver tumor and normal liver cells.


In the present invention, the expression which is assayed is preferably the relative expression of each gene, calculated with reference to at least one (preferably 1, 2, 3 or 4) invariant gene(s). Invariant genes, suitable to perform the invention, are genes whose expression is constant whatever the grade of the liver tumors, such as for example ACTG1, EFF1A1, PNN and RHOT2 genes, whose features are summarized in Table 3. In a particular embodiment preferred, the relative expression is calculated with respect to at least the RHOT2 gene or with respect to the RHOT2 gene.


In another advantageous embodiment, the relative expression is calculated with respect to at least the PNN gene or with respect to the PNN gene. It may be calculated with respect to the RHOT2 and PNN genes.


The calculation of the absolute expression or of the relative expression of each gene of the set and of each invariant gene being assayed with the same method from the same sample, preferably at the same time, enables to determine for each sample a gene expression profile.

TABLE 3Features of invariant genes. ACTG1, EEF1A1, PNN and RHOT2proteins are defined in SEQ ID NOs: 34, 36, 38 and 40 respectively.symbolGene nameSEQ 10*Accession NoLocationFunctionACTG1actin, gamma 133NM_00161417q25cytoplasmic actincytoskeleton innonmuscle cellsEEF1A1eukaryotic translation35NM13 001 4026q14.1enzymatic delivery ofelongation factor 1aminoacyl tRNAs toalpha 1the ribosomePNNpinin, desmosome37NM_00268714q21.1transcriptionalassociated proteincorepressor, RNAsplicing regulatorRHOT2ras homolog gene39NM_13876916p13.3Signaling by Rhofamily, member T2GTPases,mitochondrial protein


An additional step of the method or process comprises the determination of the grade of said liver tumor, referring to the gene expression profile that has been assayed. In a particular embodiment of the invention, the method is designed to determine the grade of hepatoblastoma, in particular paediatric hepatoblastoma. In another embodiment, the method is designed to determine the grade of hepatocellular carcinoma, in particular paediatric HCC or adult HCC.


According to a particular embodiment of the invention, in the step of the method which is performed to determine the grade of the liver tumor, a gene expression profile or a signature (preferably obtained after normalization), which is thus specific for each sample, is compared to the gene expression profile of a reference sample or to the gene expression profiles of each sample of a collection of reference samples (individually tested) whose grade is known, so as to determine the grade of said liver tumor. This comparison step is carried out with at least one prediction algorithm. In a particular embodiment, the comparison step is carried out with 1, 2, 3, 4, 5 or 6 prediction algorithms chosen in the following prediction algorithms: Compound Covariate Predictor (CCP), Linear Discriminator Analysis (LDA), One Nearest Neighbor (1NN), Three Nearest Neighbor (3NN), Nearest Centroid (NC) and Support Vector Machine (SVM). These six algorithms are part of the “Biometric Research Branch (BRB) Tools” developed by the National Cancer Institut (NCI) and are available on http://linus.nci.nih.gov/BRB-ArrayTools.html. Equivalent algorithms may be used instead of or in addition to the above ones. Each algorithm classifies tumors within either of the two groups, defined as tumors with good prognosis (such as C1) or tumors with bad prognosis (such as C2); each group comprises the respective reference samples used for comparison, and one of these two groups also comprises the tumor to be classified.


Therefore, when 6 algorithms are used, the grade of a tumor sample may be assigned with certainty to the class of good prognosis or to the class of bad prognosis, when 5 or 6 of the above algorithms classified the tumor sample in the same group. In contrast, when less than 5 of the above algorithms classify a tumor sample in the same group, it provides an indication of the grade rather than a definite classification.


Reference samples which can be used for comparison with the gene expression profile of a tumor to be tested are one or several sample(s) representative for tumor with poor prognosis (such as C2), one or several sample(s) representative of tumor with good prognosis (such as C1), one or several sample(s) of a normal adult liver and/or one or several sample(s) of a fetal liver.


Table 4 lists the level of expression of each gene of Table 1 depending upon the status of the reference sample i.e., robust tumor with poor prognostic and robust tumor with good prognostic. Examples of methods to identify such robust tumors are provided in the examples. The present invention provides a new classification method in this respect, which is based on discretization of continuous values.

TABLE 4Level of expression of the genes of Table 1, with respectto the status of the robust tumorsNucleotideExpression status in robust tumortargetwith poor prognosiswith good prognosisAFPoverexpressedunderexpressedALDH2underexpressedoverexpressedAPCSunderexpressedovorexpressedAPOC4underexpressedoverexpressedAQP9underexpressedoverexpressedBUB1overexpressedunderexpressedC1SunderexpressedoverexpressedCYP2E1underexpressedoverexpressedDLG7overexpressedunderexpressedDUSP9overexpressedunderexpressedE2F5overexpressedunderexpressedGHRunderexpressedoverexpressedHPDunderexpressedoverexpressedIGSF1overexpressedunderexpressedNLE1overexpressedunderexpressedRPL10Aoverexpressedunderexpressed


Reference samples usually correspond to so-called “robust tumor” for which all the marker genes providing the signature are expressed (either under expressed or overexpressed) as expected i.e., in accordance with the results disclosed in Table 5, when tested in similar conditions, as disclosed in the examples hereafter.


A robust tumor having an overexpression of one or several gene(s) selected among ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, GHR and HPD genes (these genes belong to the so-called group of differentiation-related genes), and/or an underexpression of one or several gene(s) selected among AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE1 and RPL10A genes (these genes belong to the so-called group of proliferation-related genes), is an indicator of a robust liver tumor, in particular of a hepatoblastoma, with a good prognosis. A robust tumor having an overexpression of one or several gene(s) selected among AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE1 and RPL10A genes, and/or an underexpression of one or several gene(s) among ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, GHR and HPD genes, is an indicator of a robust liver tumor, in particular of a hepatoblastoma, with a poor prognosis. In the present application, a gene is said “underexpressed” when its expression is lower than the expression of the same gene in the other tumor grade, and a gene is said “overexpressed” when its expression is higher than the expression of the same gene in the other tumor grade.


In a particular embodiment, Table 5 provides the gene expression profiles of the 16 genes of Table 1 in 13 samples of hepatoblastoma (HB) including 8 samples that have been previously identified as rC1 subtype and 5 samples that have been previously identified as rC2 subtype. This Table can therefore be used for comparison, to determine the gene expression profile of a HB tumor to be classified, with the robust tumors disclosed (constituting reference samples), for a set of genes as defined in the present application. Said comparison involves using the classification algorithms which are disclosed herein, for both the selected reference samples and the assayed sample.

TABLE 5embedded image
Normalized qPCR data of 16 genes in 13 HB samples including 8 samples of the rC1 subtype and 5 samples of the rC2 subtype (in grey). The qpCR values have been obtained by measuring the expression of the 16 genes in 8 samples of the rC1 subtype and 5 samples of the rC2 subtype by the SYBR green method using the primers as disclosed in Table 6 below and in the conditions reported in the examples, and normalized by the ROTH2 gene (primers in Table 7).


The method of the present invention is also suitable to classify new tumor samples, and to use them as new reference samples. Therefore, the gene expression values of these new reference samples may be used in combination or in place of some of the values reported in Table 5.


In another embodiment of the invention, the step of determining the tumor grade comprises performing a method of discretization of continuous values of gene expression obtained on the set of genes the tested patients' samples. Discretization is generally defined as the process of transforming a continuous-valued variable into a discrete one by creating a set of contiguous intervals (or equivalently a set of cutpoints) that spans the range of the variable's values. Discretization has been disclosed for use in classification performance in Lustgarten J. L. et al, 2008.


The inventors have observed that discretization can be effective in determining liver tumor grade, especially for those tumors described in the present application, including Hepatoblastoma (HB) or Hepatocellular carcinoma (HCC).


The discretization method is especially disclosed in the examples where it is illustrated by using data obtained on tumor samples wherein these data are those obtained from profiling the 16 genes providing the large set of genes for expression profiling according to the invention. It is pointed out that the discretization method may however be carried out on a reduced number of profiled genes within this group of 16 genes, starting from a set consisting of 2 genes (or more genes) including one (or more) overexpressed proliferation-related genes chosen among AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE1 and RPL10A and one down-regulated differentiation-related gene chosen among ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, GHR, HPD, said genes being thus classified as a result of gene profiles observed on robust tumors with poor prognosis (according to the classification in Table 4 above). In particular embodiments of the discretization method, the number of assayed gene for expression profiling is 2, 4, 6, 8, 10, 12, 14 or 16 and the same number of genes in each category (either the group of overexpressed proliferation-related genes or the group of downregulated differentiation-related gene) is used to perform the method.


The invention thus relates to a method enabling the determination of the tumor grade on a patient's sample, which comprises a classification of the tumor through discretization according to the following steps:

    • measuring the expression and especially the relative (normalized) expression of each gene in a set of genes defined as the signature of the tumor, for example by quantitative PCR thereby obtaining data as Ct or preferably Delta Ct, wherein said set of genes is divided in two groups, a first group consisting of the proliferation-related genes and a second group consisting of the differentiation-related genes (as disclosed above),
    • comparing the values measured for each gene, to a cut-off value determined for each gene of the set of genes, and assigning a discretized value to each of said measured values with respect to said cut-off value, said discretized value being advantageously a “1” or a “2” value assigned with respect to the cut-off value of the gene and optionally, if two cut-offs values are used for one gene, a further discretized value such as a “1.5” or another value between “1” or “2” may be assigned for the measured values which are intermediate between the cut-offs values,
    • determining the average of the discretized values for the genes, in each group of the set of genes,
    • determining the ratio of the average for the discretized values for the proliferation-related genes on the average for the discretized values for the differentiation-related genes, thereby obtaining a score for the sample,
    • comparing the obtained score for the sample with one or more sample cut-off(s), wherein each cut-off has been assessed for a selected percentile,
    • determining the tumor grade as C1 or C2, as a result of the classification of the sample with respect to said sample cut-off.


The above defined ratio of average values may be alternatively calculated as the ratio of the average for the discresized values for the differentiation-related genes on the average for the discretized values for the proliferation-related genes, to obtain a score. If this calculation made is adopted the cut-offs values are inversed, i.e., are calculated as 1/xxx.


In order to carry out the discretization method of the invention, the data obtained on the assayed genes for profiling a patient's sample are preferably normalized with respect to one or more invariant gene(s) of the present invention, in order to prevent detrimental impact on the results that may arise from possible inaccuracy in the quantification of initial nucleic acid, especially RNA, in the sample.


Normalization with respect to one invariant gene only, especially when said invariant gene is RHOT2 gene has proved to be relevant in the results obtained by the inventors. Similarly normalization with respect to PNN gene would be an advantageous possibility because the gene does also not vary in expression.


In order to design a discretization method for the determination of tumor grade of an individual sample of a patient, according to the invention, cut-offs values have to be determined to allow the determination of the tumor grade. The cut-offs values can be determined experimentally by carrying out the following steps on expression profiling results obtained on a determined number of tumor samples:

    • defining a cut-off (threshold value) for each gene in the set of genes designed for the signature, said cut-off corresponding to the value of the absolute or preferably relative (i.e. normalized) expression of said gene at a selected percentile and said percentile being selected for each of two groups of genes defined in the set of genes. In order to do so, the set of profiled genes comprises the same number of genes within each of the 2 groups of genes consisting of the group of overexpressed proliferation-related genes encompassing AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE1 and RPL10A and the group of down-regulated differentiation-related gene encompassing ALDH2, APCS, APOC4, AQP9, C15, CYP2E1, GHR, HPD (said groups being defined based on gene profiles on robust tumors with poor prognosis),
    • in each tumor sample assigning to each expression value (especially normalized expression value) obtained for each expression profiled gene in the sample, a discretized value which is codified with respect to the cut-off value determined for the same gene and in line with the defined contiguous intervals of continuous values, e.g. a discretized value of “1” or “2” if two intervals (categories) are defined or a discretized value of “1”, “1.5” (or another value between 1 and 2) or “2” if three intervals are defined, said assignment of discretized value being advantageously such that the “1” is assigned for expression values falling below the cut-off found for the differentiation-related genes and for expression values falling below the cut-off found for the proliferation-related genes, the “2” is assigned for expression values falling above the cut-off found for the differentiation-related genes and for expression values falling above the cut-off found for the proliferation-related genes, and optionally if a “1.5” is used it is assigned to values found between the cut-offs;
    • on each tumor sample, determining in each group (proliferation-related genes group or differentiation-related genes group) the average value of said assigned discretized values of profiled genes of the set of profiled genes;
    • determining a score for each sample, as the ratio between the average expression values of said genes in said two groups of genes in the set of profiled genes;
    • determining on the basis of the obtained scores for all the tumor samples, one or more cut-off value(s) for the sample, corresponding to the respective value(s) at one or more (especially 2 or 3) percentile(s), wherein said percentile(s) is (are) either identical or different from the percentiles(s) selected for the genes.


      When the cut-offs values for each gene of the set of genes for profiling have been obtained for a sufficient number of relevant samples and the cut-off value for the sample is determined on the basis of the same samples, these cut-offs can be adopted as reference cut-offs for the user who will be carrying out the analysis of any further patient's tumor sample, especially for the purpose of determining the tumor grade in a patient's sample, if the analysis is performed in identical or similar conditions as the conditions which led to the establishment of the cut-offs values.


Therefore the invention provides cut-offs values as reference cut-offs, in order to carry out the determination of tumor grade in particular testing conditions as those disclosed below and in the examples.


In a particular embodiment of the method of discretization, the cut-off for each gene is the value corresponding to a determined percentile, which can be different for each of the considered two groups of genes (proliferation-related genes on the one hand and differentiation-related genes on the other hand). The selected percentile (or quantile) is determined with respect to the fraction of tumors (such as ⅓ or more) harbouring some chosen features such as overexpression of proliferation-related genes and/or dowregulation of differentiation-related genes, in the two groups of genes of the set of genes. Especially, when one intends to assign more weight to tumors displaying strong overexpression of proliferation-related genes and/or strong downregulation of differentiation-related genes, the cut-off corresponds to a high quantile (above the 50th, preferably the 60th, or even above the 65th, such as the 67th and for example within the range of 55th and 70th) for said proliferation-related genes and the cut-off corresponds to a low quantile (below the 50th, preferably equal to or below the 40th for example the 33rd, and for example within the range of between 20th and 40th) of the differentiation-related genes. The cut-off for each group of genes and the cut-off for the sample may be determined with respect to the same percentile(s) or may be determined with respect to different percentile.


According to a particular embodiment of the invention, for HB tumors, the percentile which is chosen for the overexpressed proliferation-related genes is the 67th and the percentile which is chosen for the downregulated differentiation-related genes is the 33rd. According to a particular embodiment of the invention, for HC tumors, the percentile which is chosen for the overexpressed proliferation-related genes is the 60th and the percentile which is chosen for the downregulated differentiation-related genes is the 40rd.


Each percentile (or cut-off value corresponding to the percentile) defines a cutpoint and the discretized values for each gene are either “1” or “2” below or above said percentile. The values “1” and “2” are distributed with respect to the percentiles so as to create the highest difference in the values of the calculated ratio for the most different tumor grades. This is illustrated in the examples for the selected percentiles.


It has been observed that in a preferred embodiment of the invention, the relative values of the profiled genes are determined by real-time PCR (qPCR).


Conditions to carry out the real-time PCR are disclosed herein, especially in the examples, as conditions applicable to analyzed samples.


PCR primers and probes suitable for the performance of RT-PCR are those disclosed herein for the various genes.


In a particular embodiment of the invention, the analysed tumor is a hepatoblastoma and its grade is determined by discretization as disclosed above and illustrated in the examples, taking into account that:

    • the set of assayed genes for profiling is constituted of the 16 genes disclosed;
    • the invariant gene (of reference) is RHOT2;
    • the cut-offs value for each gene based on −dCt (minus delta Ct) measures) are:


      AFP: 3.96139596; ALDH2: 4.3590482; APCS: 4.4691582; APOC4: 2.03068712; AQP9: 3.38391456; BUB1: −1.41294708; C1S: 4.24839464; CYP2E1: 6.70659644; DLG7: −3.3912188; DUSP9: 2.07022648; E2F5: −0.72728656; GHR: −0.1505569200; HPD: 2.27655628; IGSF1: 0.1075015200; NLE: −0.02343571999; RPL10A: 6.19723876.
    • the cut-off value for the sample is 0.91 (for the 67th) and optionally a further the cut-off value for the sample is 0.615 (for the 33rd). In such a case, a sample with a score above 0.91 is classified into the C2 class and a sample with a score below 0.91 is classified into the C1 class. The reference to the cut-off at 0.615 may be used to refine the results for values between both cut-offs.


In another embodiment of the invention, the tumor is an hepatocellular carcinoma and its grade is determined by discretization as disclosed above and illustrated in the examples, taking into account that:

    • the set of assayed genes for profiling is constituted of the 16 genes disclosed;
    • the invariant gene (of reference) is RHOT2;


the cut-offs value for each gene based on −dCt (minus delta Ct) measures) are:

Gene nameCut-off for TaqmanCut-off for SybrGreenAFP−1.2634010−2.3753035ALDH24.0141435.314302APCS5.61429076.399079APQC4−0.79631584.656336AQP94.28360115.446966BUB1−1.2736579−3.634476C1S6.35146796.240002CYP2E16.95624195.829384DLG7−2.335694−4.614352DUSP9−7.979559−1.8626715E2F5−0.4400218−1.367846GHR1.08326321.169362HPD6.74803286.736329IGSF1−4.84177857.6653982NLE−1.6167268−1.82226RPL10A6.24830565.731897
    • the cut-off value for the score of a sample based on the ration between the average of the discretized values of the “proliferation-related genes” on the “differentiation-related genes” are 0.66 determined as the 30th percentile of the score) and 0.925 (determined as the 67th percentile of the score) In such a case, a sample with a score above 0.925 is classified into the C2 class and a sample with a score below 0.66 is classified into the C1 class. The sample with a score (initial score) between 0.66 and 0.925 can be assigned to an intermediate class. It can alternatively be classified as C1 or C2 using a modified score corresponding to the average of the discretized values of the “proliferation-related genes”. A new cut-off value is determined for said genes, which is the cut-off value for the modified score (in the present case it is 1.3). This cut-off can be determined via a percentile (here the 60th) of the distribution of the modified scores, using the samples of the intermediate class. A sample (initially classified in the intermediate class) with a modified score below 1.3 can be re-classified into the C1 class, and a sample with a modified score above 1.3 can be re-classified into the C2 class.


It is observed that the refinement of the results which are between the cut-offs of the samples is advantageous for hepatocellular carcinoma in order to increase the relevancy of the information on the tumor grade.


Generally said refinement of the classification of the intermediate results in the HCC is obtained by performing the following steps:


a modified score is determined which corresponds to the average of the discretized values of the “proliferation-related genes” only for the sample. A new cut-off value is determined for said genes, which is the cut-off value for the modified score (in the present case it is 1.3). This cut-off can be determined via a percentile (here the 60th) of the distribution of the modified scores, using the samples of the intermediate class. A sample (initially classified in the intermediate class) with a modified score below the “proliferation cut-off” (for example 1.3) can be re-classified into the C1 class, and a sample with a modified score above the “proliferation cut-off” (for example 1.3) can be re-classified into the C2 class.


From the 16 genes expressed in liver cells listed in Table 1, a set comprising from 2 to 16 genes (or more generally a set as defined herein) may be used to assay the grade of tumor cells in a tumor originating from the liver. The results obtained, after determining the expression of each of the genes of the set, are then treated for classification according to the steps disclosed herein. The invention relates to each and any combination of genes disclosed in Table 1, to provide a set comprising from 2 to 16 of these genes, in particular a set comprising or consisting of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16 of these genes. In the designed set, one or many genes of Table 1 may be modified by substitution or by addition of one or several genes as explained above, which also enable to determine the grade of the liver tumor, when assayed in combination with the other genes.


In a preferred embodiment, the liver tumor is a paediatric HB, and the method or process of the invention enables to distinguish a first class, called C1, qualifying as a good prognosis tumor and a second class, called C2, qualifying as a poor prognosis tumor. The C1 grade is predominantly composed of fetal histotype cells (i.e., well differentiated and non proliferative cells). In contrast, the C2 grade presents cells other than the fetal histotype such as embryonic, atypic (crowded fetal), small cell undifferiantiated (SCUD) and/or macrotrabecular cells.


The present invention also relates to a kit suitable to determine the grade of a liver tumor from the sample obtained from a patient. This kit is appropriate to carry out the method or process described in the present application.


In a particular embodiment, the kit comprises a plurality of pairs of primers specific for a set of genes to be assayed, said set comprising from 2 to 16 genes, said 2 to 16 genes being chosen in the group consisting of AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.


By “plurality”, it is mean that the kit comprises at least as many pairs of primers as genes to enable assaying each selected gene, and in particular the nucleotide target of this gene. Accordingly, each gene and in particular its nucleotide target is specifically targeted by a least one of these pairs of primers. In a particular embodiment, the kit comprises the same number of pairs of primers as the number of genes to assay and each primer pair specifically targets one of the genes, and in particular the nucleotide targets of one of these genes, and does not hybridize with the other genes of the set.


The kits of the invention are defined to amplify the nucleotide targets of the sets of genes as described in the present invention. Therefore, the kit of the invention comprises from 2 to 16 pairs of primers which, when taken as a whole, are specific for said from 2 to 16 genes out of the 16 genes of Table 1. In particular, the kit comprises or consists of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 pairs of primers specific for 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 out of the 16 genes of Table 1. In a particular embodiment, the kit comprises or consists of 16 pairs of primers specific for the 16 genes of Table 1 i.e., a primer pair specific for each of the following genes: AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.


When the set of genes has been modified by the addition or substitution of at least one gene as described above, the kit is adapted to contain a pair of primers specific for each added or substituted gene(s). As indicated by the term “comprises”, the kit may, besides the pairs of primers specific for the genes of Table 1, contain additional pair(s) of primers.


In a particular embodiment, the kit comprises at least one pair of primers (preferably one) for at least one invariant gene (preferably one or two) to be assayed for the determination of the expression profile of the genes, by comparison with the expression profile of the invariant gene.


The number of pairs of primers of the kit usually does not exceed 100, particularly 50, 30, 20, more particularly 16, and even more particularly is maximum 5, 6, 7, 8, 9 or 10.


In the kits of the invention, it is understood that, for each gene, at least one pair of primers and preferably exactly one pair, enabling to amplify the nucleotide targets of this gene, is present. When the kits provide several pairs of primers for the same gene, the gene expression level is measured by amplification with only one pair of primers. It is excluded that amplification may be performed using simultaneously several pairs of primers for the same gene.


As defined herein, a pair of primers consists of a forward polynucleotide and a backward polynucleotide, having the capacity to match its nucleotide target and to amplify, when appropriate conditions and reagents are brought, a nucleotide sequence framed by their complementary sequence, in the sequence of their nucleotide target.


The pairs of primers present in the kits of the invention are specific for a gene i.e., each pair of primers amplifies the nucleotide targets of one and only one gene among the set. Therefore, it is excluded that a pair of primers specific for a gene amplifies, in a exponential or even in a linear way, the nucleotide targets of another gene and/or other nucleic acids contained in sample. In this way, the sequence of a primer (whose pair is specific for a gene) is selected to be not found in a sequence found in another gene, is not complementary to a sequence found in this another gene and/or is not able to hybridize in amplification conditions as defined in the present application with the sequence of the nucleotide targets of this another gene.


In a particular embodiment, the forward and/or backward primer(s) may be labelled, either by isotopic (such as radioactive) or non isotopic (such as fluorescent, biotin, fluororochrome) methods. The label of the primer(s) leads to the labelling of the amplicon (product of amplification), since the primers are incorporated in the final product.


The design of a pair of primers is well known in the art and in particular may be carried out by reference to Sambrook et al. (Molecular Cloning, A laboratory Manual, Third Edition; chapter 8 and in particular pages 8.13 to 8.16). Various softwares are available to design pairs of primers, such as Oligo™ or Primer3.


Therefore, each primer of the pair (forward and backward) has, independently from each other, the following features:

    • their size is from 10 and 50 bp, preferably 15 to 30 bp; and
    • they have the capacity to hybridize with the sequence of the nucleotide targets of a gene.


In a particular embodiment, when the pairs of primers are used in a simultaneous amplification reaction carried out on the sample, the various primers have the capacity to hybridize with their respective nucleotide targets at the same temperature and in the same conditions.


Conventional conditions for PCR amplification are well known in the art and in particular in Sambrook et al. An example of common conditions for amplification by PCR is dNTP (200 mM), MgCl2 (0.5-3 mM) and primers (100-200 nM).


In a particular embodiment, the sequence of the primer is 100% identical to one of the strands of the sequence of the nucleotide target to which it must hybridize with, i.e. is 100% complementary to the sequence of the nucleotide target to which it must hybridize. In another embodiment, the identity or complementarity is not 100%, but the similarity is at least 80%, at least 85%, at least 90% or at least 95% with its complementary sequence in the nucleotide target. In a particular embodiment, the primer differs from its counterpart in the sequence of the sequence of the nucleotide target by 1, 2, 3, 4 or 5 mutation(s) (deletion, insertion and/or substitution), preferably by 1, 2, 3, 4 or 5 nucleotide substitutions. In a particular embodiment, the mutations are not located in the last 5 nucleotides of the 3′ end of the primer.


In a particular embodiment, the primer, which is not 100% identical or complementary, keeps the capacity to hybridize with the sequence of the nucleotide target, similarly to the primer that is 100% identical or 100% complementary with the sequence of the nucleotide target (in the hybridization conditions defined herein). In order to be specific, at least one of the primers (having at least 80% similarity as defined above) of the pair specific for a gene can not hybridize with the sequence found in the nucleotide targets of another gene of the set and of another gene of the sample.


In a particular embodiment, the pairs of primers used for amplifying a particular set of genes are designed, besides some or all of the features explained herein, in order that the amplification products (or amplicons) of each gene have approximately the same size. By “approximately” is meant that the difference of size between the longest amplicon and the shortest amplicon of the set is less than 30% (of the size of the longest amplicon), preferably less than 20%, more preferably less than 10%. As particular embodiments, the size of the amplicon is between 100 and 300 bp, such as about 100, 150, 200, 250 or 300 bp.


The nucleotide sequences of the 16 genes of Table 1 are provided in the Figures, and may be used to design specific pairs of primers for amplification, in view of the explanations above.


Examples of primers that may be used to measure the expression of the genes of Table 1, in particular to amplify the nucleotide targets of the genes of Table 1, are the primers having the sequence provided in Table 6 or variant primers having at least 80% similarity (or more as defined above) with the sequences defined in Table 6.

TABLE 6Sequence of forward and backward primers of the 16 genesdefined in Table 1. These primers may be used in any real-time PCR, in particular the SYBR green technique, exceptfor the Taqman ® protocol.ProductsizeTarget(bp)Forward primer (5′-3′)Reverse primer (5′-3′)AFP151AACTATTGGCCTGTGGCGAGTCATCCACCACCAAGCTGCALDH2151GTTTGGAGCCCAGTCACCCTGGGAGGAAGCTTGCATGATTCAPCS151GGCCAGGAATATGAACAAGCCCTTCTCCAGCGGTGTGATCAAPOC4151GGAGCTGCTGGAGACAGTGGTTTGGATTCGAGGAACCAGGAQP9151GCTTCCTCCCTGGGACTGACAACCAAAGGGCCCACTACABUB1152ACCCCTGAAAAAGTGATGCCTTCATCCTGTTCCAAAAATCCGC1S141TTGTTTGGTTCTGTCATCCGCTGGAACACATTTCGGCAGCCYP2E1151CAACCAAGAATTTCCTGATCCAGAAGAAACAACTCCATGCGAGCDLG7151GCAGGAAGAATGTGCTGAAACATCCAAGTCTTTGAGAAGGGCCDUSP9151CGGAGGCCATTGAGTTCATTACCAGGTCATAGGCATCGTTGE2F5151CCATTCAGGCACCTTCTGGTACGGGCTTAGATGAACTCGACTGHR151CTTGGCACTGGCAGGATCAAGGTGAACGGCACTTGGTGHPD151ATCTTCACCAAACCGGTGCACCATGTTGGTGAGGTTACCCCIGSF1152CACTCACACTGAAAAACGCCCGGGTGGAGCAATTGAAAGTCANLE1151ATGTGAAGGCCCAGAAGCTGGAGAACTTCGGGCCGTCTCRPL10A151TATCCCCCACATGGACATCGTGCCTTATTTAAACCTGGGCC


The kit of the invention may further comprise one or many pairs of primers specific for one or many invariant genes, in particular specific for ACTG1, EFF1A1, PNN and/or RHOT2 genes. The pair of primers specific for invariant gene(s) may be designed and selected as explained above for the pair of primers specific for the genes of the set of the invention. In a particular embodiment, the pairs of primers of the invariant genes are designed in order that their amplification product (or amplicon) has approximately the same size as the amplicon of the genes of the set to be assayed (the term approximately being defined as above, with respect to the longest amplicon of the set of genes). Examples of primers that may be used to amplify the particular invariant genes are primers having the sequence provided in Table 7 or primers having at least 80% similarity (or more as defined above) with the sequences defined in Table 7.

TABLE 7Sequence of forward and backward primers specific for theinvariant genes defined in Table 3. These primers may beused in real-time PCR, in particular the SYBR greentechnique, except for the Taqman ® protocol.ProductsizeTarget(bp)Forward primer (5′-3′)Reverse primer (5′-3′)ACTG1151GATGGCCAGGTCATCACCATACAGGTCTTTGCGGATGTCCEFF1A1151TCACCCGTAAGGATGGCAATCGGCCAACAGGAACAGTACCPNN151CCTTTCTGGTCCTGGTGGAGTGATTCTCTTCTGGTCCGACGRHOT2151CTGCGGACTATCTCTCCCCTCAAAAGGCTTTGCAGCTCCAC


The kits of the invention may also further comprise, in association with or independently of the pairs of primers specific for the invariant gene(s), reagents necessary for the amplification of the nucleotide targets of the sets of the invention and if any, of the nucleotide targets of the invariant genes.


The kits of the invention may also comprise probes as disclosed herein in the context of sets of probes, compositions and arrays. In particular, the kits also comprise the four dNTPs (nucleotides), amplification buffer, a polymerase (in particular a DNA polymerase, and more particularly a thermostable DNA polymerase) and/or salts necessary for the activity of the polymerase (such as Mg2+).


Finally, the kits may also comprise one or several control sample(s) i.e., at least one sample(s) representative of tumor with bad (i.e., poor) prognosis (in particular a HB C2 grade), at least one sample(s) representative of tumor with good prognosis (in particular a HB C1 grade), at least one sample of a normal adult liver and/or at least one sample of a fetal liver.


The kits may also comprise instructions to carry out the amplification step or the various steps of the method of the invention.


The invention is also directed to a set of probes suitable to determine the grade of a liver tumor from the sample obtained from a patient. This set of probes is appropriate to carry out the method or process described in the present invention. It may also be part of the kit.


This set of probes comprises a plurality of probes in particular from 2 to 16 probes, these 2 to 16 probes being specific for genes chosen in the group consisting of AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.


By “plurality”, it is mean that the set of probes comprises at least as many probes as genes to assay. In a particular embodiment, the array comprises the same number of probes as the number of genes to assay.


The probes of the sets of the invention are selected for their capacity to hybridize to the nucleotide targets of the sets of genes as described in the present invention. Therefore, the set of probes of the invention comprise from 2 to 16 probes specific for 2 to 16 genes out of the 16 genes of Table 1. In particular, the sets of probes comprise or consist of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 probes specific of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 out of the 16 genes of Table 1. In a particular embodiment, the sets of probes comprise or consist of 16 probes specific for the 16 genes of Table 1 i.e., a probe specific of each of the following genes: AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.


The specificity of the probes is defined according to the same parameters as those applying to define specific primers.


When the set of genes has been modified by the addition or substitution of at least one gene as described above, the set of probes is adapted to contain a probe specific for the added or substituted gene(s). As indicated by the term “comprises”, the set of probes may, besides the probes specific for the genes of Table 1, contain additional probe(s).


The number of probes of the set does usually not exceed 100, particularly 50, 30, 20, more particularly 16, and even more particularly is maximum 5, 6, 7, 8, 9 or 10.


In the set of probes of the invention, it is understood that for each gene corresponds at least one probe to which the nucleotide target of this gene hybridize to. The set of probes may comprise several probes for the same gene, either probes having the same sequence or probes having different sequences.


As defined herein, a probe is a polynucleotide, especially DNA, having the capacity to hybridize to the nucleotide target of a gene. Hybridization is usually carried out at a temperature ranging from 40 to 60° C. in hybridization buffer (see example of buffers below). These probes may be oligonucleotides, PCR products or cDNA vectors or purified inserts. The size of each probe is independently to each other from 15 and 1000 bp, preferably 100 to 500 bp or 15 to 500 bp, more preferably 50 to 200 bp or 15 to 100 bp. The design of probes is well known in the art and in particular may be carried out by reference to Sambrook et al. (Molecular Cloning, A laboratory Manual, Third Edition; chapters 9 and 10 and in particular pages 10.1 to 10.10).


The probes may be optionally labelled, either by isotopic (radioactive) or non isotopic (biotin, fluororochrome) methods. Methods to label probes are disclosed in Sambrook et al. (Molecular Cloning, A laboratory Manual, Third Edition; chapter 8 and in particular page 9.3). In a particular embodiment, the probes are modified to confer them different physicochemical properties (such as by methylation, ethylation). In another particular embodiment, the probes may be modified to add a functional group (such as a thiol group), and optionally immobilized on bead (preferably glass beads).


In a particular embodiment, the sequence of the probe is 100% identical to a part of one strand of the sequence of the nucleotide target to which it must hybridize, i.e. is 100% complementary to a part of the sequence of the nucleotide target to which it must hybridize. In another embodiment, the identity or complementarity is not 100% and the similarity is at least 80%, at least 85%, at least 90% or at least 95% with a part of the sequence of the nucleotide target. In a particular embodiment, the probe differs from a part of one strand of the sequence of the nucleotide target by 1 to 10 mutation(s) (deletion, insertion and/or substitution), preferably by 1 to 10 nucleotide substitutions. By “a part of”, it is meant consecutive nucleotides of the nucleotide target, which correspond to the sequence of the probe.


In a particular embodiment, the probe, which is not 100% identical or complementary, keeps the capacity to hybridize, in particular to specifically hybridize, to the sequence of the nucleotide target, similarly to the probe which is 100% identical or 100% complementary with the sequence of the nucleotide target (in the hybridization conditions defined herein).


In a particular embodiment, the size of the probes used to assay a set of genes is approximately the same for all the probes. By “approximately” is meant that the difference of size between the longest probe and the shortest probe of the set is less than 30% (of the size of the longest probe), preferably less than 20%, more preferably less than 10%.


The set of probes of the invention may further comprise at least one (preferably one) probe specific for at least one invariant gene (preferably one or two), in particular specific for ACTG1, EFF1A1, PNN and/or RHOT2 genes. The probes specific for invariant gene(s) may be designed and selected as explained above for the probes specific for genes of the sets of the invention. In a particular embodiment, the probes specific of the invariant genes have approximately the same size as the probes specific of the genes of the set of be assayed (the term approximately being defined as above, with respect to the longest probes of the set of genes).


The invention is also directed to an array suitable to determine the grade of a liver tumor from the sample obtained from a patient. This array is appropriate to carry out the method or process described in the present application.


An array is defined as a solid support on which probes as defined above, are spotted or immobilized. The solid support may be porous or non-porous, and is usually glass slides, silica, nitrocellulose, acrylamide or nylon membranes or filters.


The arrays of the invention comprise a plurality of probes specific for a set of genes to be assayed. In particular, the array comprises, spotted on it, a set of probes as defined above.


The invention also relates to a composition comprising a set of probes as defined above in solution.


In a first embodiment, the probes (as defined above in the set of probes) may be modified to confer them different physicochemical properties (such as methylation, ethylation). The nucleotide targets (as defined herein and prepared from the sample) are linked to particles, preferably magnetic particles, for example covered with ITO (indium tin oxide) or polyimide. The solution of probes is then put in contact with the target nucleotides linked to the particles. The probe/target complexes are then detected, for example by mass spectrometry.


Alternatively, probes may be modified to add a functional group (such as a thiol group) and immobilized on beads (preferably glass beads). These probes immobilized on beads are put in contact with a sample comprising the nucleotide targets, and the probe/target complexes are detected, for example by capillary reaction.


The invention is also directed to kits comprising the sets of probes, the compositions or the arrays of the invention and preferably the primer pairs disclosed herein. These kits may also further comprise reagents necessary for the hybridization of the nucleotide targets of the sets of genes and/or of the invariant genes, to the probes (as such, in the compositions or on the arrays) and the washing of the array to remove unbound nucleotides targets.


In a particular embodiment, the kits also comprise reagents necessary for the hybridization, such as prehybridization buffer (for example containing 5×SSC, 0.1% SDS and 1% bovine serum albumin), hybridization buffer (for example containing 50% formamide, 10×SSC, and 0.2% SDS), low-stringency wash buffer (for example containing 1×SSC and 0.2% SDS) and/or high-stringency wash buffer (for example containing 0.1×SSC and 0.2% SDS).


The kits may also comprise one or several control sample(s) i.e., at least one sample(s) representative for tumor with poor prognosis, at least one sample(s) representative of tumor with good prognosis, at least one sample of a normal adult liver and/or at least one sample of a fetal liver. Alternatively, it may comprise the representation of a gene expression profile of such tumors.


Finally, the invention provides a kit as described above further comprising instructions to carry out the method or process of the invention.


The arrays and/or kits (either comprising pairs of primers or probes or arrays or compositions of the invention or all the components) according to the invention may be used in various aspects, in particular to determine the grade of a liver tumor from a patient, especially by the method disclosed in the present application.


The arrays and/or kits according to the invention are also useful to determine, depending upon the grade of the liver tumor, the risk for a patient to develop metastasis. Indeed, the classification of a liver tumor in the class with poor prognosis is highly associated with the risk of developing metastasis.


In another embodiment, the arrays and/or kits according to the invention are also useful to define, depending upon the grade of the liver tumor, the therapeutic regimen to apply to the patient.


The invention also relates to a support comprising the data identifying the gene expression profile obtained when carrying out the method of the invention.




BRIEF DESCRIPTION OF THE DRAWINGS

The colour version of the drawings as filed is available upon request to the European Patent Office.



FIG. 1. Identification of Two HB Subclasses by Expression Profiling.


(A) Schematic overview of the approach used to identify robust clusters of samples, including two tumor clusters (rC1 and rC2) and one non-tumor cluster (NL) (B) Expression profiles of 982 probe sets (824 genes) that discriminate rC1 and rC2 samples (p<0.001, two-sample t test). Data are plotted as a heatmap where red and green correspond to high and low expression in log2-transformed scale. (C) Molecular classification of 25 HB samples and status of CTNNB1 gene and β-catenin protein. C1 and C2 classification was based on rC1 and rC2 gene signature by using six different statistical predictive methods (CCP, LDA, 1NN, 3NN, NC and SVM) and the leave-one-out cross-validation. Black and gray squares indicate mutations of the CTNNB1 and AXIN1 genes. Immunohistochemical analysis of β-catenin in representative C1 and C2 cases is shown. (D) Expression of representative Wnt-related and β-catenin target genes (p<0.005, two-sample t test) in HB subclasses and non-tumor livers (NL). (E) Classification of hepatoblastoma by expression profile of a 16-gene signature. (F) Classification of normal human livers of children with HB (from 3 months to 6 years of age) (NT) or fetal livers at 17 to 35 weeks of gestation (FL) by expression profile of a 16-gene signature.



FIG. 2: Molecular HB subclasses are related to liver development stages. (A) Distinctive histologic and immunostaining patterns of HB subclasses C1 and C2. From top to bottom: numbers indicate the ratio of mixed epithelial-mesenchymal tumors and of tumors with predominant fetal histotype in C1 and C2 subtypes; hematoxylin and eosin (H&E) and immunostaining of Ki-67, AFP and GLUL in representative samples. Magnification, ×400. (B) Expression of selected markers of mature hepatocytes and hepatoblast/liver progenitors in HB subclasses and non-tumor livers.



FIG. 3: Validation of the 16-gene signature by qPCR in an independent set of 41 HBs. Expression profiles of the 16 genes forming the HB classifier are shown as a heatmap that indicates high (red) and low (green) expression according to log2-transformed scale. HB tumors, HB biopsies (b) and human fetal livers (FL) at different weeks (w) of gestation were assigned to class 1 or 2 by using the 16-gene expression profile, six different statistical predictive methods (CCP, LDA, 1NN, 3NN, NC and SVM) and leave-one-out cross-validation. Black boxes in the rows indicate from top to bottom: human fetal liver, mixed epithelial-mesenchymal histology, predominant fetal histotype, and β-catenin mutation.



FIG. 4: Gene expression of the 16 genes of the prognostic liver cancer signature assessed by qPCR is presented as box-plot. The boxes represent the 25-75 percentile range, the lines the 10-90 percentile range, and the horizontal bars the median values.



FIG. 5: Expression level of the 16 liver prognostic signature genes shown case by case in 46 hepatoblastomas and 8 normal livers. C1 tumors (green), C2 tumors (red) and normal liver (white).



FIG. 6. Correlation between molecular HB subtypes and clinical outcome in 61 patients. (A) Association of clinical and pathological data with HB classification in the complete set of 61 patients. Only significant correlations (Chi-square test) are shown. PRETEXT IV stage indicates tumorous involvement of all liver sections. (B) Kaplan-Meier plots of overall survival for 48 patients that received preoperative chemotherapy. Profiling via the 16-gene expression signature was used to define C1 and C2 subclasses in tumors resected after chemotherapy, and differences between survival curves were assessed with the log-rank test. (C) Overall survival of 17 HB patients for which pretreatment biopsies or primary surgery specimens were available. The signature was applied exclusively to tumor samples without prior therapy. (D) Multivariate analysis including 3 variables associated to patient's survival. The predominant histotype is defined as either fetal or other (including embryonal, crowed-fetal, macrotrabecular or SCUD types). Tumor stage is defined by PRETEXT stage (Perilongo et al., 2000) and/or distant metastasis at diagnosis and/or vascular invasion. HR, Hazard Ratio; CI, Confidence Interval.



FIG. 7: Clinical, pathological and genetic characteristics of 61 HB cases. SR: standard risk; HR: high risk according to SIOPEL criteria; NA: not available; PRETEXT: pre-treatment extent of disease according to SIOPEL; DOD: dead of disease; *: Vascular invasion was defined by radiological analysis; **: The predominant epithelial histotype variable categorized as “others” included embryonal, crowded fetal, macrotrabecular, and undifferentiated histotypes.



FIG. 8: Clinical, pathological and genetic characteristics of 66 HB samples; Tumor ID number indicates patient number. When more than one sample from the same patient was analyzed, the representative sample used for statistical analysis of clinical correlations is marked by an asterisk; b: biopsy. HB74F: fetal component of HB74; HB74e: embryonal component of HB74. Gender: M, male; F, female; Y, yes; N, no; NA, not available. Multifocality: S, solitary nodules; M, multiple nodules. Histology: E, epithelial; M, mixed; CF, crowded fetal; F, fetal; E, embryonal; M, macrotrabecular; PF, pure fetal; S, SCUD. PRETEXT β-catenin status: wt, wild-type; Δex3, in-frame deletion of part or all exon 3 sequence; FAP, familial polyposis kindred; AXIN1, Axin 1 nonsense mutation (R533stop, CGA to TGA).stage: I to IV according to SIOPEL (Aronson et al., 2005). Treatment protocol: S, standard risk; H, high risk according to SIOPEL. Outcome: A, alive free of disease; DOD, dead of disease; D, death unrelated to cancer; R, alive with recurrence of disease.



FIG. 9: Correlation between molecular HB subtypes and clinical outcome in 86 patients. (A) Association of clinical and pathological data with HB classification in the complete set of 86 patients. Only significant correlations (Chi-square test) are shown. PRETEXT IV stage indicates tumorous involvement of all liver sections. (B) Kaplan-Meier plots of overall survival for 73 patients that received preoperative chemotherapy. Profiling via the 16-gene expression signature was used to define C1 and C2 subclasses in tumors resected after chemotherapy, and differences between survival curves were assessed with the log-rank test. (C) Overall survival of 29 HB patients for which pretreatment biopsies or primary surgery specimens were available. The signature was applied exclusively to tumor samples without prior therapy. (D) Multivariate analysis including 3 variables associated to patient's survival. The predominant histotype is defined as either fetal or other (including embryonal, crowed-fetal, macrotrabecular or SCUD types). Tumor stage is defined by PRETEXT stage (Perilongo et al., 2000) and/or distant metastasis at diagnosis and/or vascular invasion. HR, Hazard Ratio; CI, Confidence Interval.



FIG. 10: Correlation between molecular HCC subtypes and clinical outcome in 64 patients. Kaplan-Meier estimates of overall survival in 64 HCC patients using molecular classification with 16 genes, with the unsupervised clustering (centroid) (A) or unsupervised clustering (average) (B).



FIG. 11: Analysis of the probability of overall survival (OS) of 85 hepatoblastoma patients using Kaplan-Meier estimates. Left pannel: cases were classified by the discretization method into 3 classes using as cut-offs the 33rd percentile and the 67th percentile. Middle pannel: cases were classified into 2 classes using the 33rd percentile. Right pannel: cases were classified into 2 classes using the 67th percentile.



FIG. 12: Analysis of the probability of overall survival (OS) or disease-free survival (DFS) of 113* HCC patients using Kaplan-Meier estimates and log-rank test.
Among the total series of 114 patients, survival data were not available for one case.


Patients were treated either by partial hepatectomy (PH) or by orthotopic liver transplantation (OLT). Unless specified, the follow-up was closed at 146 months.


A: HCC cases were classified into 3 classes by the discretization method using as cut-offs the 33rd and the 67th percentiles.


B: 47 HCC cases previously classified into the intermediate class (33<p<67, see pannel A) were subdivided into 2 new subclasses using the 60th percentile of proliferation-related genes.


C: 92 HCC cases treated by partial hepatectomy (PH) were classified into 3 classes as in pannel A.


D: 21 HCC cases treated by orthotopic liver transplantation (OLT) were classified into 2 classes using as cut-off the 67th percentile.


E: HCC cases were classified into 2 classes using different combinations of scores as described in Table F.


F: HCC cases were classified into 2 classes using as cut-off the 33rd percentile.


G: HCC cases were classified into 2 classes using as cut-off the 50th percentile.


H: HCC cases were classified into 2 classes using as cut-off the 67th percentile.


I: 92 HCC cases treated by partial hepatectomy (PH) were classified into 2 classes using as cut-off the 33rd percentile.


J: 92 HCC cases treated by partial hepatectomy (PH) were classified into 2 classes using as cut-off the 50th percentile.


K: 92 HCC cases treated by partial hepatectomy (PH) were classified into 2 classes using as cut-off the 67th percentile.


L: Disease-free survival of 113 HCC cases after classification into 2 classes using as cut-off the 67th percentile. Follow-up was closed at 48 months. Data were not significant when the follow-up was closed at 146 months.


M: Disease-free survival of 92 HCC cases treated by PH, after classification into 2 classes using as cut-off the 67th percentile. Follow-up was closed at 48 months. Data were not significant when the follow-up was closed at 146 months.



FIG. 13: Analysis of the probability of overall survival (OS) or disease-free survival (DFS) HCC patients using Kaplan-Meier estimates and log-rank test.




EXAMPLES
Experimental Procedures

A. Patients and Tissue Samples.


Sixty-six tumor specimens and biopsies from 61 patients with hepatoblastoma were collected from different hospitals in France (52 cases), Italy (6 cases), United Kingdom (1 case), Switzerland (1 case) and Slovakia (1 case). Forty-eight patients received chemotherapy treatment prior to surgery, most being enrolled in clinical trials of the International Childhood Liver Tumour Strategy Group (SIOPEL) (Perilongo et al., 2000). Samples from fresh tumors avoiding fibrotic and necrotic areas and from adjacent non tumor livers were snap frozen at the time of surgery and stored at −80° C. FIG. 7 describes patient characteristics and clinicopathological parameters.


Patients were children with median age of 2 years, and male:female ratio of 1.5. The median follow-up was 32 months; during this period, 15 patients died from disease. The histology of all tumor specimens was centrally reviewed by expert pathologist according to previously described criteria (Finegold et al., 2007; Zimmermann, 2005). Twenty-five tumors were analyzed on oligonucleotide microarrays and 24 of them, for which DNA was available, were subjected to aCGH analysis, while a second set of 41 tumors was analyzed by qPCR (FIG. 8). No difference was observed in significant clinical and pathological data as well as in the percentage of cases carrying β-catenin mutation between the two sets. This study has been approved by the Ethics Committee of Institut Pasteur, and informed consent of the families was obtained at each Medical Center, in accordance with European Guidelines for biomedical research and with national laws in each country.


B. Oligonucleotide Microarrays and Gene Expression Data Analysis


Twenty-five HB samples and 4 non-tumor samples including a pool of livers from 3 males and a second from 3 females were analyzed using Affymetrix HG-U133A oligonucleotide arrays. Total RNA was prepared using FastPrep® system (Qbiogene, Strasbourg, France) and RNeasy mini Kit (Qiagen, Courtaboeuf, France). RNA quality was checked with the Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, Calif.). Microarray experiments were performed according to the manufacturer's instructions. Affymetrix microarray data were normalized using RMA method (Irizarry et al., 2003). Class discovery was done as described elsewhere (Lamant et al., 2007). Pathway and Gene Ontology enrichment analyses were performed using GSEA method (Subramanian et al., 2005) and hypergeometric tests. For supervised tests and class prediction, we used Biometric Research Branch (BRB) ArrayTools v3.2.2 software, developed by R. Simon and A. Peng. Permutations of the measurements are then used to estimate the FDR (the percentage of genes identified by chance). Additionally, mouse fetal livers at E18.5 and postnatal livers at 8 days of birth were profiled on Affymetrix MG-U74A, B v2 arrays. Data were processed and analyzed as aforementioned.


Except when indicated, transcriptome analysis was carried out using either an assortment of R system software packages (http://www.R-project.org, v2.3.0) including those of Bioconductor v1.8 (Gentleman et al., 2004) or original R code.


B.1. Normalization


Raw data from Affymetrix HG-U133A 2.0 GeneChip™ microarrays were normalized in batch using robust multi-array average method (R package affy, v1.10.0) (Irizarry et al., 2003). Probe sets corresponding to control genes or having a “_x_” annotation were masked yielding a total of 19,787 probe sets available for further analyses.


B.2. Class Discovery


Step 1


Variance Test


The variance of each probe set across samples was tested and compared to the median variance of all the probe sets, using the model: ((n−1)×Var(probe set)/Varmed), where n refers to the number of samples. By using the same filtering tool of BRB ArrayTools software, the P-value for each probe set was obtained by comparison of this model to a percentile of Chi-square distribution with (n−1) degrees of freedom.


Robust Coefficient of Variation (rCV)


The rCV was calculated for each probe set as follows. After ordering the intensity values of n samples from min to max, we eliminated the min and max values and we calculated the coefficient of variation (CV) for the remaining values.


Unsupervised Probe Sets Selection


Unsupervised selection of probe set lists was based on the two following criteria:


(i) variance test at P<0.01,


(ii) rCV less than 10 and superior to a given rCV percentile. We used eight rCV percentile thresholds (60%; 70%; 80%; 90%; 95%; 97.5%; 99%; 99.5%), which yielded 8 probe set lists.


Step 2: Generation of a Series of 24 Dendrograms


Hierarchical clustering was performed by using the 8 rCV-ranked probe sets lists, 3 different linkage methods (average, complete and Ward's), and 1-Pearson correlation as a distance metric (package cluster v1.9.3). This analysis generated 24 dendrograms.


Step 3:


Stability Assessment


The intrinsic stability of each of the 24 dendrograms was assessed by comparing each dendrogram to the dendrograms obtained after data “perturbation” or “resampling” (100 iterations). Perturbation stands for the addition of random gaussian noise (μ=0, σ=1.5×median variance calculated from the data set) to the data matrix, and resampling for the random substitution of 5% of the samples by virtual sample's profiles, generated randomly. The comparison between dendrograms across all iterations yielded a mean ‘similarity score’ (see below). The overall stability was assessed by calculating a mean similarity score, using all pairs of the 24 dendrograms.


Similarity Score


To compare two dendrograms, we compared the two partitions in k clusters (k=2 to 8) obtained from these two dendrograms. To compare a pair of partitions, we used a similarity measure, which corresponds to the symmetric difference distance (Robinson and Foulds, 1981).


Step 4: Identification of Robust Clusters


We identified groups in which any pair of samples was co-classified in at least 22 of the 24 partitions, and considered only groups made of 4 samples or more. Then, for any pair of these groups, we calculated the mean number of co-classification of any sample in the first group with any sample in the second group. We aggregated the groups for which this score was at least 18 (over the 24 partitions).


B.3. Supervised Tests


We compared gene expression between two classes of samples by using the Student's t test with random variance model option (BRB ArrayTools software, version 3.4.0a, developed by Dr. Richard Simon and Amy Peng Lam, http://linus.nci.nih.gov/BRB-ArrayTools.html). False Discovery Rates were assessed by using 1000 random permutations of labels (Monte Carlo approach).


B.4. Classification


To classify samples according to gene expression profile, we used the Class prediction tool of BRB ArrayTools software using all 6 following algorithms: Compound Covariate Predictor (CCP), Linear Discriminant Analysis (LDA), 1-Nearest Neighbor (1NN), 3-Nearest Neighbors (3NN), Nearest Centroid (NC) and Support Vector Machines (SVM). Each sample was classified according to the majority of the 6 algorithms. Samples classified as C2 by at least 3 algorithms were classified accordingly.


B.5. Gene Ontology and Pathway Analysis


We used a hypergeometric test to measure the association between a gene (probe set) list and a gene ontology term (GO term), as in GO stats R package (R. Gentleman). To this end, we mapped the gene list and the GO terms to non-redundant Entrez Gene identifiers by using the annotation file HG-U133_Plus2.annot.csv (http://www.affymetrix.com, Dec. 14, 2006). GO terms and their relationships (parent/child) were downloaded from http://www.geneontology.org (version Dec. 31, 2006). The list of proteins associated to GO terms (table gene_association.goa_human) and mapping the Entrez Gene ids (table human.xrefs) were downloaded from ftp://ftp.ebi.ac.uk/pub/databases/GO/goa.


KEGG pathway annotation was done by Onto-tools software (http://vortex.cs.wayne.edu/ontoexpress/servlet/UserInfo). We designated a significance threshold of each hypergeometric test at P<0.001, and the condition that a GO term or pathway be represented by at least 3 Entrez Gene identifiers.


B.6. Gene Set Enrichment Analysis (gsea)


GSEA (Subramanian et al., 2005) was used to evaluate the correlation of a specific gene list with two different sample groups (phenotypes). Briefly, this method calculates an enrichment score after ranking all genes in the dataset based on their correlation with a chosen phenotype and identifying the rank positions of all the members of a defined gene set. We used the signal2noise ratio as a statistic to compare specific and random phenotypes in order to evaluate statistical differences.


C. Array-Based Comparative Genomic Hybridization (aCGH)


Genomic DNA from 24 HBs and 3 non-tumor liver samples was analyzed using aCGH chips designed by the CIT-CGH consortium. This array contains 3400 sequence-verified PAC/BAC clones spaced at approximately 1 Mb intervals, spotted in triplicate on Ultra Gaps slides (Corning Inc, Corning, N.Y.).


The aCGH chip was designed by CIT-CGH consortium (Olivier Delattre laboratory, Curie Institute, Paris; Charles Theillet laboratory, CRLC Val d'Aurelle, Montpellier; Stanislas du Manoir laboratory, IGBMC, Strasbourg and the company IntegraGen™). DNAs were labeled by the random priming method (Bioprime DNA labelling system; Invitrogen, Cergy-Pontoise, France) with cyanine-5 (Perkin-Elmer, Wellesley, Mass.). Using the same procedure, we labeled control DNAs with cyanine-3. After ethanol-precipitation with 210 μg of Human Cot-1 DNA (Invitrogen), resuspension in hybridization buffer (50% formamide), denaturation at 95° C. for 10 minutes and prehybridization at 37° C. for 90 minutes, probes were cohybridized on aCGH. The aCGH slides were previously preblocked with a buffer containing 2.6 mg succinic anhydride/118 ml N-methyl-2-pyrrolidinone/32 ml sodium tetraborate decahydrate, pH 8.0 (Sigma-Aldrich, Lyon, France). After washing, arrays were scanned using a 4000B scan (Axon, Union City, Calif.). Image analysis was performed with Genepix 5.1 software (Axon) and ratios of Cy5/Cy3 signals were determined. The aCGH data were normalized using lowess per block method (Dudoit et al., 2002). Comparison between groups was done using chi-square test or Fisher's exact test, as appropriate.


Status assignment (Gain/Loss) was performed using R package GLAD v1.6.0. Computation of recurrent minimal genomic alterations was done using slight modification of a previously described method (Rouveirol et al., 2006). For comparison between groups, we used the Fischer exact test. Complete aCGH data will be published elsewhere.


D. Mouse Microarray Analysis


Murine Genome Affymetrix U74v2 A and B arrays were used to investigate liver expression at embryonic day 18.5 (E18.5) and at 8 days after birth (PN8). Each time point consisted of a pool of livers from 3-5 animals analyzed in triplicate. Microarray experiments were performed according to the manufacturer's instructions.


Publicly available Affymetrix Mouse Genome (MG) 430 2.0 array liver expression data at embryonic time points E11.5, E12.5, E13.5, E14.5, and E16.5 days of gestation (Otu et al., 2007), were downloaded from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE6998).


MG-U74v2, MG-430 2.0 and HG-133A 2.0 array intra- and cross-species probeset comparison was achieved by using the Affymetrix NetAffx analysis center and by choosing “Good Match” degree of specificity. Unification of sample replicates, multiple array data standardization and Heatmap visualization was done by using dCHIP v1.6 software. Comparison of fetal liver stages by supervised analysis was performed using BRB ArrayTools software as previously described, by classing E11.5 and E12.5 as “Early” and E14.5 and E16.5 as “Late” fetal liver stage. Supervised signature was applied to HB array data, and intensity cut-off=60 was chosen in order to remove probesets that did not reach such intensity level in at least one sample.


E. Quantitative PCR Analysis (qPCR)


For qPCR analysis, we used RNA from 52 tumor samples (including 11 samples analyzed on microarrays, see FIG. 8), and from 8 non-tumor livers and 5 human fetal livers (RNAs purchased from BioChain Institute, Hayward, Calif.).


RNA was extracted by using either Trizol, RNeasy kit (QIAGEN) or miRvana kit (Ambion), then quantified and quality-checked by Agilent technology. For each cDNA preparation, 1 μg of RNA was diluted at the final concentration of 100 ng/μl, and reverse transcribed with the Superscript RT kit (Invitrogen, Carlsbad, Calif.) following the manufacturer's protocol. Random primers (Promega, Charbonnières-les-Bains, France) were added at the final concentration of 30 ng/μl and the final volume was 20 μl.


The cDNA was diluted 1:25, and 5 μl were used for each qPCR reaction. We added 5 μl of 2×Sybr Green Master mix (Applied Biosystems) and 0.3 μl of each specific primer (final concentration 300 nM). Each reaction was performed in triplicate. qPCR reactions were run on the Applied Biosystems 7900HT Fast Real-Time PCR System with a 384-well thermo-block, in the following conditions: 2 min at 50° C. to activate Uracil-N-glycosylase (UNG)-mediated erase of aspecific reaction; 10 min at 95° C. to activate the polymerase and inactivate the UNG; 40 cycles (15 sec at 95° C. denaturation step and 1 min at 60° C. annealing and extension); and final dissociation step to verify amplicon specificity.


The lists of primers used for qPCR are provided in Table 6 and Table 7 above.


F. Immunohistochemistry (IHC)


IHC was carried out as reported previously (Wei et al., 2000). For antigen retrieval at 95° C., we used 1 mM EDTA (pH 8) for β-catenin and Ki-67 IHC, and 10 mM citrate buffer (pH 6) for AFP and GLUL IHC. We used monoclonal antibodies against β-catenin and GLUL (Cat. Nos. 610154 and 610517; BD Biosciences, Le Pont de Claix, France) and Ki-67 (M7240, Dako, Trappes, France) and polyclonal antibody against AFP (N1501, Dako). Reactions were visualized using the ChemMate Dako Envision Detection kit (Dako) and diaminobenzidine. Subcellular distribution and quantitative evaluation of immunostaining in the different histotypes were assessed by examining at least ten random high-power fields.


G. Clinical Data Analysis


We used the Chi-square test for comparisons between groups. Survival curves were calculated according to the Kaplan-Meier method, using the log-rank test to assess differences between curves. Variables independently related to survival were determined by stepwise forward Cox regression analysis. Follow-up was closed at February 2007 or at time of death. Statistical analysis was done with SPSS software v10.0 (SPSS Inc., Chicago, Ill.).


H. Examples of Other Pairs of Primers and Probes for the 16 Genes of Table 1 and the 4 Invariant Genes (Table 3) that can be Used in the Taqman® Method.

AFP forward primer:GCCAGTGCTGCACTTCTTCAAFP reverse primer:TGTTTCATCCACCACCAAGCTAFP probe:ATGCCAACAGGAGGCCATGCTTCA(for each polynucleotide, the sequence is givenfrom 5′ to 3′)ALDH2 forward primer:TGCAGGATGGCATGACCATALDH2 reverse primer:TCTTGAACTTCAGGATCTGCATCAALDH2 probe:CCAAGGAGGAGATCTTCGGGCCAAPCS forward primer:AGCTGGGAGTCCTCATCAGGTAAPCS reverse primer:CGCAGACCCTTTTTCACCAAAPCS probe:TGCTGAATTTTGGATCAATGGGACACCAPOC4 forward primer:TGAAGGAGCTGCTGGAGACAAPOC4 reverse primer:CGGGCTCCAGAACCATTGAPOC4 probe:TGGTGAACAGGACCAGAGACGGGTGAQP9 forward primer:GCCATCGGCCTCCTGATTAAQP9 reverse primer:GTTCATGGCACAGCCACTGTAQP9 probe:TGTCATTGCTTCCTCCCTGGGACTGBUB1 forward primer:ACATCTGGTTTTCAGTGTGTTGAGABUB1 reverse primer:GTTGCAGCAACCCCAAAGTAABUB1 probe:TCAGCAACAAACCATGGAACTACCAGATCGC1S forward primer:TCCCAATGACAAGACCAAATTCTC1S reverse primer:AGAGCCCATAGGTCCCACACTC1S probe:CGCAGCTGGCCTGGTGTCCTGCYP2E1 forwardCATGAGATTCAGCGGTTCATCAprimer:CYP2E1 reverseGGTGTCTCGGGTTGCTTCAprimer:CYP2E1 probe:CCTCGTGCCCTCCAACCTGCCDLG7 forward primer:GCTGGAGAGGAGACATCAAGAACDLG7 reverse primer:CCTGGTTGTAGAGGTGAAAAAGTAATCDLG7 probe:TGCCAGACACATTTCTTTTGGTGGTAACCDUSP9 forward primer:GGCCTACCTCATGCAGAAGCTDUSP9 reverse primer:GGGAGATGTTAGACTTCTTCCTCTTGDUSP9 probe:CACCTCTCTCTCAACGATGCCTATGACCTGE2F5 forward primer:CCTGTTCCCCCACCTGATGE2F5 reverse primer:TTTCTGTGGAGTCACTGGAGTCAE2F5 probe:CCTCACACAGCCTTCCTCCCAGTCCGHR forward primer:CCCAGGTGAGCGACATTACAGHR reverse primer:CATCCCTGCCTTATTCTTTTGGGHR probe:CAGCAGGTAGTGTGGTCCTTTCCCCGHPD forward primer:CCCACGCTCTTCCTGGAAHPD reverse primer:TTGCCGGCTCCAAAACCHPD probe:TCATCCAGGGCCACAACCACCAIGSF1 forward primer:GACCATTGCCCTTGAAGAGTGTIGSF1 reverse primer:GAGAGGTTGATGAAGGAGAATTGGIGSF1 probe:ACCAAGAAGGAGAACCAGGCACCCCNLE1 forward primer:TGCCTCCTTTGACAAGTCCATNLE1 reverse primer:CGCGTAGGGAAGCCAGGTANLE1 probe:TGGGATGGGAGGAGGGGCARPL10A forward primerTCGGCCCAGGTTTAAATAAGGRPL10A reverse primerCCACTTTGGCCACCATGTTTRPL10A Taqman probeAGTTCCCTTCCCTGCTCACACACAACGACTG1 forward primer:GGCGCCCAGCACCATACTG1 reverse primer:CCGATCCACACCGAGTACTTGACTG1 probe:ATCAAGATCATCGCACCCCCAGAGGEEF1A1 forwardGCGGTGGGTGTCATCAAAGprimer:EEF1A11 reverseTGGGCAGACTTGGTGACCTTprimer:EEF1A11 probe:AGTGGACAAGAAGGCTGCTGGAGCTGPNN forward primer:GAATTCCCGGTCCGACAGAPNN reverse primer:TTTCGGTCTCTTTCACTTCTTGAAPNN probe:AGAGGTCTATATCAGAGAGTAGTCGATCAGGCAAAAGARHQT2 forward primer:CCCAGCACCACGATCTTCACRHOT2 reverse primer:CCAGAAGGAAGAGGGATGCARHOT2 Taqman probe:CAGCTCGCCACCATGGCCG


Results


Identification of Two HB Subclasses by Gene Expression Profiling


For robust unsupervised classification, we generated and screened a series of 24 dendrograms to identify samples that co-clustered whatever the method and the gene list. We obtained two robust subgroups of tumors named robust Cluster 1 (rC1, n=8) and robust Cluster 2 (rC2, n=5) (FIG. 1A). Comparison of rC1 and rC2 expression profiles identified 824 genes (p<0.001, false discovery rate (FDR)=0.02) (FIG. 1B). KEGG pathway analysis pinpointed a strong enrichment of cell cycle related genes (p<10−11), most being up-regulated in rC2 tumors. These: genes were mainly assigned to GO categories including mitosis regulation, spindle checkpoint, nucleotide biosynthesis, RNA helicase activity, ribosome biogenesis, and translational regulation. Evidence that rC2 tumors were faster proliferating than rC1 tumors was further confirmed by Ki-67 immunostaining (see FIG. 2A).


The remaining tumors were classified into C1 (rC1-related) and C2 (rC2-related) subclasses by applying a predictive approach based on the rC1/rC2 gene signature and using robust samples as training set (FIG. 1C). Both groups exhibited similar, high rates of β-catenin mutations, and accordingly, immunohistochemistry (IHC) of β-catenin showed cytoplasmic and nuclear staining of the protein in the majority of HBs. However, β-catenin localization was predominantly membranous and cytoplasmic in C1 tumors, whereas it showed frequent loss of membrane anchoring and intense nuclear accumulation in C2 tumors (FIG. 1C).


We observed differential expression of a number of Wnt members and targets between subclasses. C2 tumors showed increased expression of MYCN, BIRC5 that encodes the anti-apoptotic factor Survivin, NPM1 (encoding nucleophosmin) and HDAC2. By contrast, most C1 tumors prominently expressed the Wnt antagonist DKK3, BMP4, and genes previously found to be activated in liver tumors carrying mutant β-catenin (Boyault et al., 2007; Renard et al., 2007; Stahl et al., 2005). Remarkably, most genes related to liver functions are expressed in the perivenous area of adult livers, such as GLUL, RHBG, and two members of the cytochrome p450 family: CYP2E1 and CYP1A1 (Benhamouche et al., 2006; Braeuning et al., 2006) (FIG. 1D).


Further evidence that the rC1 subclass was enriched in genes assigned to the hepatic perivenous program was provided by Gene Set Enrichment Analysis (GSEA), a computational method for assessing enrichment of a predefined gene list in one class as compared with another (Subramanian et al., 2005). Thus, Wnt/β-catenin signaling appears to activate different transcriptional programs in HB subtypes, likely reflecting different cellular contexts.


HB Subclasses Evoke Distinct Phases of Liver Development


Next, we sought to determine whether HB subclasses were associated with specific histological phenotypes. Mixed epithelial-mesenchymal tumors that represented 20% of cases were not significantly associated with C1 and C2 subclasses. By contrast, a tight association was found with the main epithelial component, which defines the cell type occupying more than 50% of tumor cross-sectional areas. Sixteen out of 18 C1 tumors displayed a predominant fetal phenotype, including 4 ‘pure fetal’ cases, whereas all C2 tumors showed a more immature pattern, with prevailing embryonal or crowded-fetal histotypes associated with high proliferation (Finegold, 1994) (p<0.0001) (FIG. 2A). Further relationship between molecular subclasses and hepatic developmental stages was provided by the finding that a number of mature hepatocyte markers were markedly downregulated in C2 compared to C1 tumors (Tables 1 and 2). Conversely, C2 tumors showed strong overexpression (35-fold) of the oncofetal AFP gene associated to high protein levels in tumor cells by IHC (FIG. 2A) and in patients' sera (r=0.79, p<0.0001). C2 tumors also abundantly expressed hepatic progenitor markers such as KRT19 (encoding cytokeratin 19) and TACSTD1, also known as Ep-CAM (FIG. 2B).


To better define the relationships between HB subclasses and phases of hepatic differentiation, we first generated a liver development-related gene signature by making use of publicly available mouse fetal and adult liver data sets (Otu et al., 2007). When applied to HB samples, this signature was able to distinguish by hierarchical clustering two HB groups closely matching the C1/C2 classification. Next, we integrated HB gene expression data with the orthologous genes expressed in mouse livers at embryonic days (E) 11.5 to 18.5, and at 8 days of birth. In unsupervised clustering, most C2 tumors co-clustered with mouse livers at early stages of embryonic development (E11.5 and E12.5), whereas C1 tumors gathered with mouse livers at late fetal and postnatal stages. Together, these data comfort the notion that tumor cells in C2 and C1 subtypes are arrested at different points of the hepatic differentiation program.


Identification of a 16-Gene Signature as HB Classifier


To investigate the relevance of molecular HB classification in an independent set of tumors, we defined a HB classifier signature derived from the top list of genes differentially expressed between rC1 and rC2 clusters. After qPCR assessment, a list of 16 top genes at p≦10−7 was selected to form a class predictor (Table 1). Most of these genes show drastic variations in expression level during liver development, and among them, BUB1 and DLG7 have been repeatedly identified as hESC markers (Assou et al., 2007). The 16-gene expression profile was first investigated in rC1 and rC2 samples used as training set, and it predicted classification with 100% of accuracy in these samples, using either microarray or qPCR data. The robustness of this signature was confirmed by correct classification into C1 and C2 subclasses of all 13 remaining tumors analyzed by microarray (FIG. 1E). Expression profiles of fetal livers and normal liver for these 16-gene signature were also assayed (FIG. 1F). This signature was therefore employed to classify a new, independent set of 41 HB samples by qPCR (FIGS. 4 and 5 and Table 8), resulting in 21 tumors categorized as C1 and 20 tumors as C2 subtype (FIG. 3).


Extending our previous observation, C1/C2 classification in this new set of tumors was unrelated to CTNNB1 mutation rate. Using qPCR, we also confirmed enhanced expression in C2 tumors of liver progenitor markers such as AFP, Ep-CAM, and KRT19, as well as MYCN (FIG. 3). Moreover, while a similar percentage of C1 and C2 tumors displayed mesenchymal components, a predominant fetal histotype was found in 95% of tumors of the C1 subtype, whereas in 82% of C2 tumors, the major component displayed less differentiated patterns such as embryonal, crowded-fetal, macrotrabecular and SCUD types (p<0.0001) (FIG. 3). To further assess the association of HB subclasses with liver development, 5 human fetal livers at different weeks of gestation were included in the qPCR studies. In unsupervised clustering, fetal livers at late (>35 weeks) and earlier (17 to 26 weeks) developmental stages were classified as C1 and C2 respectively, further supporting that HB subclasses reflect maturation arrest at different developmental phases.

TABLE 8Gene expression of the prognostic signature for liver cancer by quantitative RT-PCR.C1 C2NLFold-changemedianminmaxmedianminmaxmedianminmaxC1/NLC2/NLC2/C1C1/C2AFP0.40.033.330.70.0456.10.20.08.82.338.116.50.1ALDH287.113.2356.715.02.274.4240.4151.6387.60.30.10.25.2APCS61.61.1338.91.90.0276.2158.692.7509.50.20.00.119.8APOC421.34.3122.81.60.124.247.022.3112.40.50.00.116.1AQP960.68.0540.62.50.190.146.638.072.71.30.10.118.9BUB10.00.00.40.90.13.90.00.00.11.216.113.40.1C1S51.114.9277.27.51.396.0223.4129.3565.30.20.00.25.7CYP2E1583.297.73463.019.70.41504.01128.6527.61697.00.70.00.051.6DLG70.00.00.00.10.00.50.00.00.01.712.47.30.1DUSP91.50.445.719.10.0179.00.60.21.34.018.34.60.2E2F50.20.02.01.10.111.70.10.00.51.86.53.50.3GHR5.20.054.00.50.02.435.220.854.50.10.00.18.6HPD22.90.9182.01.20.123.8111.562.6165.70.20.00.114.0IGSF10.10.01.71.70.019.80.10.00.12.222.410.20.1NLE0.40.14.80.80.35.10.40.20.81.22.21.80.5RPL10A73.312.0230.498.211.9432.886.954.1159.90.81.11.50.7
NL, non-tumor liver; C1, good prognosis hepatoblastomas; C2, bad prognosis hepatoblastomas. Shown are the median values of 46 hepatoblastomas from 41 patients, the minimal and maximal values in each class, and the fold changes between classes. Data are presented in arbitrary units after normalization of the raw quantitative PCR values with genes (ACTG1,
# EFF1A1, PNN and RHOT2) that presents highly similar values in all samples. Gene expression of the 16 genes are presented on FIGS. 4 and 5.


The 16-Gene Signature as a Strong Independent Prognostic Factor


In a First Set of 61 Patients


The clinical impact of HB molecular classification was addressed in a first set of 61 patients (FIGS. 7 and 8), comprising 37 (61%) C1 and 24 (39%) C2 cases. Besides strong association with predominant immature histotypes, HBs of the C2 subclass were tightly associated with features of advanced tumor stage, such as vascular invasion and extrahepatic metastasis (FIG. 6A). Accordingly, overall survival of these patients was markedly impaired. Kaplan-Meier estimates of overall survival probability at 2-years were 50% for patients with C2 tumors and 90% for patients with C1 tumors (p=0.0001, log rank test), and similar trends were seen for disease-free survival probabilities (data not shown). Next, we examined whether pre-operative chemotherapy treatment given to 48 patients could affect tumor classification. These cases were evenly distributed among HB subclasses, with no significant association with molecular classification. Of note, available pretreatment biopsies were assigned to the same subclass as matched resected tumors in 3 out of 4 cases (see FIG. 3; HB112 and HB112b have been both classified as C1 grade, and HB114 and HB114b have been both classified as C2 grade). We examined the performance of the 16-gene signature on the 48 tumors resected after chemotherapy, and found significant difference in outcome between patients with C1 and C2 type HBs (p=0.0021, log rank test) (FIG. 6B). Remarkably, Kaplan-Meier analysis confirmed C2 subclass as a poor prognostic group in 17 cases for which pre-treatment biopsies or primary surgery specimens were available (p=0.0318, log rank test) (FIG. 6C).


We further assessed the prognostic validity of the 16-gene signature for all patients in multivariate analysis, using a Cox proportional hazards model with pathological and clinical variables associated to patients' survival. This analysis identified the signature as an independent prognostic factor, with better performance than tumor stage defined by PRETEXT stage, vascular invasion and extrahepatic metastases (FIG. 6D). Thus, this signature demonstrated strong prognostic relevance when compared to current clinical criteria.


In a Second Set of 86 Patients


The clinical impact of HB molecular classification was addressed in a second set of patients (comprising the sample of the first set), comprising 53 (61%) C1 and 33 (39%) C2 cases. Besides strong association with predominant immature histotypes, HBs of the C2 subclass were tightly associated with features of advanced tumor stage, such as vascular invasion and extrahepatic metastasis (FIG. 9A). Accordingly, overall survival of these patients was markedly impaired. Kaplan-Meier estimates of overall survival probability at 2-years were 60% for patients with C2 tumors and 94% for patients with C1 tumors (p=0.00001, log rank test), and similar trends were seen for disease-free survival probabilities (Table 9).

Table 9N. of patients61 C1+25 C2 = 86P valueSurvival (all patients)Alive/DeadC150/3<0.00001C220/13DFS (all Datients)DFS/othersC148/5<0.00001C218/15Survival (non-treated Patients)Alive/DeadC112/00.0164C211/6DES (non-treated patients)DES/othersC112/00.0213C212/6
Survival analysis (Kaplan Mejer, log rank test); DES: disease-free survival; Others: dead or alive with recurrent disease.


Next, we examined whether pre-operative chemotherapy treatment given to 73 patients could affect tumor classification. These cases were evenly distributed among HB subclasses, with no significant association with molecular classification. We examined the performance of the 16-gene signature on the 73 tumors resected after chemotherapy, and found significant difference in outcome between patients with C1 and C2 type HBs (p=0.0002, log rank test) (FIG. 9B). Remarkably, Kaplan-Meier analysis confirmed C2 subclass as a poor prognostic group in 29 cases for which pre-treatment biopsies or primary surgery specimens were available (p=0.0164, log rank test) (FIG. 9C).


We further assessed the prognostic validity of the 16-gene signature for all patients in multivariate analysis, using a Cox proportional hazards model with pathological and clinical variables associated to patients' survival. This analysis identified the signature as an independent prognostic factor, with better performance than tumor stage defined by PRETEXT stage, vascular invasion and extrahepatic metastases (FIG. 9D).


Finally, various clinical elements of 103 HB samples from 86 patients were compared with respect to their classification as C1 or C2 grade using the 16-gene signature (Table 10).

TABLE 10Clinical correlations.N. of patients61 + 25 = 86p-value (chi-square)GendernsChemotherapy treatmentYes/NoC147/6nsC226/7Chemotherapy protocolSTD/HighC130/130.007C2 9/16TUMOR STAGEEarly/AdvancedC132/200.005C210/23MetastasisNo/YesC143/100.004C217/16Vascular InvasionNo/YesC136/150.005C213/20Advanced Pretext stage (IV)No/YesC142/9nsC224/7MultifocalityNo/YesC136/17nsC218/14HistologyEp/MixedC131/21nsC220/13Main EDith ComDFetal/NonFetalC148/4<0.0001C2 6/22
STD: standard risks (cisplatine) - High:high risk (cisplatine/doxorubicine, intensified treatment); Tumor stage (defined as Vasc. Inv and/or metastasis and/or PRETEXT stage IV); metastasis: extrahepatic metastasis (mainly lung); vascular invasion is determined by imagery; Pretext IV (involved an intrahepatic extent of the tumor to all hepatic sections);
# multifocality (more than 2 tumor nodules); Ep: pure epithelial form - Mixed: mesenchymatous and epithelial mixed form; Fetal: well differentiated; non fetal: embryonic, atypic, SCUD and/or macrotrabecular cells.


The above results carried out on a first set of 61 patients, and on a second completed set of 86 patients, demonstrate that the 16-gene signature, identified in the present application, is a strong prognostic relevance when compared to current clinical criteria.


Discussion


The present application demonstrates that, using integrated molecular and genetic studies, hepatoblastoma encompass two major molecular subclasses of tumors that evoke early and late phases of prenatal liver development. Aberrant activation of the canonical Wnt pathway represented a seminal event in both tumor types, with cumulated mutation rates of β-catenin, APC and AXIN over 80%. However, depending on tumor differentiation stage, Wnt signaling activated distinct transcriptional programs involved in tumor growth and invasiveness or in liver metabolism. Further comparisons of immature, embryonal-type HBs with the bulk of more differentiated, fetal-type tumors revealed a tight correlation between stage of hepatic maturation arrest and clinical behavior, notably vascular invasion and metastatic spread, and patients' survival.


Molecular Hb Subclasses are Determined by Liver Differentiation Stages


In this study, expression-based classification of HB was achieved through a highly reliable statistical method combining different unsupervised hierarchical clustering approaches. This method led to the selection of two robust tumor subgroups, and this robustness was confirmed using a new, independent set of samples and 16 relevant genes discriminating these tumor subgroups. These results demonstrated that the most significant differences between HB subclasses can be ascribed to distinct hepatic differentiation stages, as defined by comparison with expression profiles of mouse livers at early (E11.5-E12.5) and late (E14.5-E18.5) embryonic stages. These studies also provide biological relevance to early histologic classification that distinguished fetal and embryonal cells as major HB components (Weinberg and Finegold, 1983). The C1 subclass recapitulates liver features at the latest stage of intrauterine life, both by expression profile and by mostly fetal morphologic patterns, while in the C2 subclass, transcriptional program and predominant embryonal histotype resemble earlier stages of liver development. Thus, despite frequent morphological heterogeneity in HB, these expression-based subclasses closely matched the histologic types found to be prevailing after microscopic examination of the entire tumor mass.


These results, showing that childhood liver tumors recapitulate programs of their developing counterpart, are in line with recent studies using cross-species comparisons. It has been demonstrated that clinically distinct medulloblastoma subtypes can be identified by their similarity with precise stages of murine cerebellar development (Kho et al., 2004). Evidence for conserved mechanisms between development and tumorigenesis was also obtained in Wilms' tumor, the embryonic kidney malignancy, which shares expression of sternness and imprinted genes with murine metanephric blastema (Dekel et al., 2006). It was noticed that HBs, like Wilms' tumors, exhibit robust overexpression of a number of paternally expressed genes like DLK1, IGF2, PEG3, and PEG10 that are involved in growth induction processes and downregulated with differentiation during development.


Previous studies using stem cell markers and markers of hepatocytic and biliary lineages have described differential patterns among HB components that reflect sequential stages of liver development (Schnater et al., 2003). The present data extent these observations, and indicate that immature C2-type tumor cells evoke hepatic cancer progenitor cells, with distinctive overexpression of highly relevant markers such as cytokeratin 19 and Ep-CAM (Roskams, 2006). Recently, embryonic stem/progenitor cells have been isolated from human fetal livers, either by enrichment of blast-like cells in primary hepatoblast cultures or by immunoselection of Ep-CAM-positive epithelial cells (Dan et al., 2006; Schmeizer et al., 2007). These cell lines have self-renewal capacity and can differentiate into mature hepatocytes and cholangiocytes, and one of them also gives rise to various mesenchymal lineages (Dan et al., 2006). Whether HBs arise from transformation of these cell types is presently unknown. As malignant mesenchymal derivatives are frequently admixed with epithelial tissues in HB, it is tempting to speculate that this tumor occurs from a multipotent progenitor harboring characteristics of mesenchymal-epithelial transitional cells. Moreover, since no significant differences in gene expression profiles was noted here between pure epithelial and mixed epithelial-mesenchymal HBs, tumor cells likely kept intrinsic capacities to undergo epithelial-mesenchymal transition.


A salient feature of immature HBs is the characteristic interplay of sternness and proliferation found in aggressive tumors (Glinsky et al., 2005). The C2-type expression profile was significantly enriched in hESC markers, including the mitotic cell cycle and spindle assembly checkpoint regulators cyclin B1, BUB1, BUB1B, and Aurora kinases. These mitotic kinases are centrosomal proteins that ensure proper spindle assembly and faithful chromosome segregation in mitosis. Overexpression of these kinases or other components of the spindle checkpoint induces centrosome amplification and defects in chromosome segregation leading to chromosome number instability and aneuploidy (Marumoto et al., 2005; Zhou et al., 1998). Non-disjunctional events are involved in developmental syndromes (Hassold and Hunt, 2001), and might be responsible for increased rate of chromosomal imbalances evidenced here in C2-type HBs.


Context-Dependent Transcriptional Programs Driven by Wnt Signalling


Mutational activation of β-catenin is a hallmark of HB, and accordingly, we found intracellular accumulation and nuclear localization of the protein in virtually all tumors, albeit with variable frequencies and intensities. Both immature and differentiated tumors overexpressed AXIN2 and DKK1, reflecting an attempt to activate a negative feedback loop aimed at limiting the Wnt signal. However, the two HB subtypes showed significant differences in β-catenin immunoexpression, illustrated by concomitant nuclear accumulation and decreased membranous localization of the protein in poorly differentiated, highly proliferative HBs. Heterogeneous distribution of nuclear β-catenin within colorectal tumors has been linked to different levels of Wnt signaling activity, resulting from differential combinations of autocrine and paracrine factors (Fodde and Brabletz, 2007). Similarly, nuclear β-catenin might be related to the absence of membranous E-cadherin in immature HBs, as we reported previously (Wei et al., 2000), and to cross-talks with growth-stimulating pathways in less differentiated cells. In this context, increased dosage of Wnt signaling might induce migratory and invasive phenotype.


Major differences between the two HB subtypes were found here in expression levels of Wnt targets involved in liver functions. Recent studies have demonstrated that Wnt/β-catenin signaling governs liver metabolic zonation by controlling positively the perivenous gene expression program and negatively the periportal program (Benhamouche et al., 2006). In our study, overexpression of hepatic perivenous markers such as GLUL was prominent in differentiated HBs, while genes encoding periportal functions like GLS2 were downregulated. This profile is highly similar to those of human and murine HCCs expressing mutant β-catenin (Boyault et al., 2007; Stahl et al., 2005), and corresponds to an hepatic signature of Wnt target genes. Accordingly, the zonation-related profile was lessened in poorly differentiated HBs, and mutant β-catenin was found to activate a different, muscle-related expression program in the pediatric Wilms' tumor (Zirn et al., 2006).


Clinical Implications


The clinical behavior of many human solid tumors has been related to their differentiation status and proliferative rate. We show that HB does not depart from this rule, with strong correlation of molecular subclasses linked to hepatic differentiation with clinical tumor stage and patient's outcome. This correlation was mainly determined by differences in invasive and metastatic phenotypes between the two subclasses, but not by differences in tumor localization or tumor extension across liver sections, which defines the preoperative staging (PRETEXT) utilized to evaluate tumor resectability (Perilongo et al., 2000). Major differences in expression profiles of the two molecular HB subtypes led us to elucidate a 16-gene signature that proved highly efficient in stratification of HBs as well as normal livers according to hepatic developmental stage. Most importantly, this classifier also discriminated aggressive tumors, exhibited powerful survival predictor capacities in pre-treatment biopsies and surgical specimens, and demonstrated strong prognostic relevance when confronted to current clinical criteria in multivariate analysis. Although immature HBs have been associated to worse clinical outcome as opposed to differentiated HBs (Weinberg and Finegold, 1983), frequent cellular heterogeneity has hampered the use of histopathologic criteria for defining risk groups, excepted for a minority of cases showing ‘pure fetal’ or SCUD types. The expression signature afforded here enables direct appraisal of the global degree of tumor cell maturation, allowing to bypass these difficulties. Thus, it can improve the outcome prediction and clinical management of hepatoblastoma, by identifying cases with increased risk of developing metastasis, or conversely, by avoiding unnecessary over-treatment.


In conclusion, the present application identifies a 16-gene signature that distinguishes two HB subclasses and that is able to discriminate invasive and metastatic hepatoblastomas, and predicts prognosis with high accuracy. The identification of this expression signature with dual capacities may be used in recognizing liver developmental stage and in predicting disease outcome. This signature can be applied to improve clinical management of pediatric liver cancer and develop novel therapeutic strategies, and is therefore relevant for therapeutic targeting of tumor progenitor populations in liver cancer.


Analysis of 64 Hepatocellular Carcinoma (HCC) from 64 Patients


Real time RT-PCR (Taqman methodology) was performed on 67 HCC samples, as disclosed for HB samples above. The clinical characteristics of the 67 patients diagnosed with HCC as well as the features of the HCC samples are disclosed in Tables 11 and 12 below.


Amplification was carried out with primers of the 16-gene signature disclosed in Table 6. Data were normalized to the expression of the ROTH2 gene (primers disclosed in Table 7) and analyzed by the ΔCt method. Quantitative PCR data are disclosed in Table 13.

TABLE 11features of the HCC samples obtained from 67 patients (pages 60 to 62)Tumorfollow-uptumor gradetumor differentiationtumorvascular invasionrecurrence orIdlength (years)(Edmonson)according to OMSsizemacromicrometastasisHC10.073moderately differentiated120NAabsentno recurrenceHC100.954moderately/poorly differentiated75absentabsentno recurrenceHC1111.10NANA15absentabsentno recurrenceHC120.05NAWell differentiated60NANAno recurrenceHC141.00NAmoderately/poorly differentiated80NANAno recurrenceHC151.223moderately differentiated60presentpresentno recurrenceHC1710.962Well differentiated100absentabsentno recurrenceHC180.393moderately differentiated140presentpresentNAHC2015.40NAWell differentiated40NANAno recurrenceHC210.70NANA100NANANAHC2211.50NAWell differentiated45absentabsentno recurrenceHC2311.932Well differentiated50absentabsentno recurrenceHC2515.872Well differentiated140absentabsentNAHC270.10NAWell differentiated15absentabsentno recurrenceHC280.10NAmoderately differentiated120NApresentno recurrenceHC33.332Well differentiated60absentabsentrecurrenceHC3011.783moderately differentiated16NANAno recurrenceHC320.662Well differentiated60absentNAno recurrenceHC3414.722Well differentiated140absentabsentrecurrenceHC370.20NAmoderately differentiated35presentpresentnonHC381.12NANA50absentNArecurrenceHC411.482Well differentiated100absentabsentno recurrenceHC417.442Well differentiated30NAabsentrecurrenceHC4210.583moderately differentiated130possible;presentno recurrencenon certainHC4310.20NAmoderately differentiated15NANAno recurrenceHC520.253moderately differentiated110absentabsentno recurrenceHC588.302moderately differentiated100absentabsentno recurrenceHC61.252Well differentiated90absentpresentrecurrenceHC645.253moderately differentiated40absentabsentrecurrenceHC668.932-3Well to moderately differentiated75absentabsentno recurrenceHC71.502-3Well differentiated100presentpresentrecurrenceHC88.483moderately differentiated30absentabsentno recurrenceHC90.023-4moderately/poorly differentiated100presentpresentno recurrenceHC1011.002-3Well to moderately differentiated35presentpresentno recurrenceHC1020.10NAPoorly differentiated200presentpresentno recurrenceHC1031.822-3Well to moderately differentiated55absentpresentrecurrenceHC1040.172-3Well to moderately differentiated160Possible;presentno recurrencenon certainHC1050.563moderately differentiated40presentpresentrecurrenceHC1061.703moderately differentiated80presentpresentno recurrenceHC1071.752Well differentiated60absentabsentno recurrenceHC1081.623moderately differentiated26absentpresentno recurrenceHC1091.001-2Well to very well differentiated30absentabsentno recurrenceHC1101.003moderately differentiated30presentpresentno recurrenceHC1110.603moderately differentiated40presentpresentno recurrenceHC11121.482-3Well to moderately differentiated18absentabsentno recurrenceHC1131.002-3Well to moderately differentiated50presentpresentno recurrenceHC1140.442Well differentiated36absentabsentno recurrenceHC1190.751Well differentiated90absentabsentno recurrenceHC1200.693moderately differentiated140absentabsentno recurrenceHC1211.002-3Well to moderately differentiated28absentabsentno recurrenceHC1220.931Very well differentiated40absentabsentno recurrenceHC1230.903moderately differentiated26absentpresentno recurrenceHC1240.822-3Well to moderately differentiated20absentpresentno recurrenceHC1250.603moderately differentiated150Possible;presentno recurrencenon certainHC1260.752Well differentiated20presentpresentrecurrenceHC1270.403moderately differentiated43probableprobableno recurrenceHC1280.523moderately differentiated62absentabsentno recurrenceHC1290.303moderately differentiated25absentpresentno recurrenceHC1310.421-2Well differentiated130presentpresentrecurrenceHC1320.252-3Well to moderately differentiated115presentpresentrecurrenceHC1330.442Well to moderately differentiated110absentpresentno recurrenceHC1340.103moderately differentiated30absentpresentno recurrenceHC1350.143moderately differentiated38absentPossible;no recurrencenon certainHC1360.262-3Well to moderately differentiated120absentpresentno recurrence
N.A: non available;

macro: macrovacular invasion;

micro: microvacular invasion









TABLE 12










features of the HCC samples obtained from 67 patients, and


features of patients (pages 63 and 64)














Chronic


Other


Tumor
Score METAVIR
viral
Viral etiology

etiolo-














ID
Activity
Fibrosis
hepatitis
HBV
HCV
alcohol
gies

















HC1
NA
4
no
no
no
yes



HC10
NA
4
yes
yes
no
no


HC11
NA
NA
yes
yes
yes
no


HC12
NA
NA
yes
yes
no
no


HC14
NA
NA
yes
no
yes
yes


HC15
3
3
no
no
no
yes


HC17
NA
3
yes
yes
no
no


HC18
2
4
no
no
no
yes


HC20
NA
NA
no
no
no
yes


HC21
NA
NA
no
no
no
yes


HC22
NA
NA
no
no
no
yes


HC23
NA
0
no
no
no
no


HC25
0
0
no
no
no
no


HC27
NA
NA
yes
no
yes
no


HC28
0
0
no
no
no
no


HC3
NA
4
yes
no
yes
no


HC30
NA
4
no
no
no
yes


HC32
NA
4
yes
no
yes
no


HC34
NA
0
no
no
no
no


HC37
NA
NA
no
no
no
yes


HC38
NA
4
yes
no
yes
no


HC4
NA
1
no
no
no
no


HC41
NA
4
yes
no
yes
no


HC42
2
1
yes
yes
no
no


HC43
NA
NA
yes
no
yes
no


HC52
NA
4
yes
yes
no
no


HC58
2
3
yes
no
yes
no


HC6
NA
1
no
no
no
yes
Hemochro


HC64
2
2
yes
no
yes
no


HC66
NA
4
yes
yes
no
yes


HC7
2
3
no
no
no
yes


HC8
NA
4
yes
no
yes
no


HC9
1
3
no
no
no
yes


HC101
2
4
yes
yes
yes
yes


HC102
1
1
yes
yes
yes
no


HC103
3
4
yes
yes
no
no


HC104
0
1
no
no
no
no


HC105
2
4
yes
no
yes
no


HC106
1
4
yes
yes
no
no


HC107
0
0-1
no
no
no
yes


HC108
1
1
yes
no
yes
no


HC109
2
4
no
no
no
yes
NASH


HC110
1
4
yes
no
yes
yes


HC111
1
4
no
no
no
yes


HC112
2
2
no
no
no
no
NASH


HC113
1
4
yes
no
yes
no


HC114
2
3
no
no
no
yes


HC119
2
1
no
no
no
no
NASH


HC120
2
3
yes
yes
no
no


HC121
2
4
yes
no
yes
no


HC122
0
1
no
no
no
no


HC123
2
4
yes
no
yes
yes


HC124
1
4
yes
yes
no
no


HC125
2
4
no
no
no
yes
NASH


HC126
1
4
yes
yes
no
no


HC127
2
4
yes
no
yes
no


HC128
1
1
no
no
no
no
NASH


HC129
2
4
no
no
no
yes


HC131
0
1
no
no
no
no


HC132
1
1
yes
yes
no
no


HC133
2
2
no
no
no
yes


HC134
2
3
yes
no
yes
no


HC135
1
2
yes
yes
no
no


HC136
0
1
no
no
no
no







N.A: non available; HBV: hepatitis B virus; HCV; hepatitis C virus; hemochro: hemochromatosis; NASH non alcoholic steatohepatitis.














TABLE 13








Quantitative PCR data of the 16-gene signature normalized to the expression of the ROTH2 gene (pages 65 to 68)

























HC1
HC3
HC4
HC6
HC7
HC8
HC9
HC10
HC11



















AFP
-2.212911
-3.865709
-7.6758115
-7.9469815
5.311541
2.0890815
-70483095
2.3869635
0.6488335


ALDH2
6.2372335
6.230074
2.186358
5.4231035
4.0446765
3.9297005
3.0017225
0.95212
5.958108


AP0C4
0.614689
0.95786
-1.608247
0.9614255
-3.550537
-0.6776965
-9.6721075
NA
1.076151


APCS
7.0721355
7.52919
5.845683
7.3704745
5.1967915
6.567126
-0.017488
-1.0272875
7.7638255


AQP9
6.047695
6.7334475
3.759528
7.006052
6.747103
3.1082155
3.7536735
1.3400495
6.122144


BUB1
-3.841505
-0.147459
-4.221132
-0.5252045
-0.299039
-1.214781
2.980029
-1.864677
-2.362454


C1S
8.163492
8.7963405
5.8997645
8.162856
4.062593
7.2991535
4.830331
2.639902
8.319293


CYP2E1
10.3093235
10.428074
7.1147515
10.1334265
11.024027
7.7910075
0.5825245
3.604805
9.575619


DLG7
-5.30317
-2.057513
-4.4226465
-1.6282005
-1.169221
-2.80866
1.3733475
NA
-2.8432205


DUSP9
-11.616567
-8.8462855
-9.4268185
-10.22051
-6.6521625
-9.6946695
-9.5262655
NA
NA


E2F5
0.05328
-1.909804
-1.7432195
0.024339
-0.2833465
-0.0193165
0.711082
-1.344368
-0.736822


GHR
2.655512
2.069524
-2.0012965
1.887805
-1.7428205
2.342442
-2.3242195
-0.4900285
4.757848


HPD
9.449416
8.549803
9.415253
8.5958965
6.183977
5.329776
-0.011478
2.932809
9.029214


IGSF1
-6.46034
-7.249974
NA
-7.1580385
-3.192514
-2.806768
-4.026769
NA
-7.6390015


NLE1
-1.159417
-1.5801355
-3.1459935
0.6940375
-0.3919565
-1.579419
-0.80375
NA
-1.9328755


RPL10A
6.6225235
6.0562915
4.4121905
6.8637555
7.1381125
6.2574845
6.3016635
9.1966395
7.379063




















HC12
HC15
HC17
HC18
HC20
HC21
HC22
HC23
HC25



















AFP
-6.538312
6.14089
7.1950405
-6.856588
-0.65281
-4.3070475
-4.418018
-5.538438
-3.90298


ALDH2
4.6271565
4.5178635
2.6522585
1.840894
6.287083
2.175112
5.331214
5.853486
6.162477


AP0C4
-1.221393
-5.156026
-2.395651
-3.84764
3.2094885
-6.2591235
0.5455545
0.5708905
1.834891


APCS
6.942673
3.380102
4.5167035
4.916924
8.2117635
5.9159775
6.6835035
6.9009145
8.798759


AQP9
4.1878425
2.373344
2.8711295
3.6093495
7.354605
1.1452535
5.7992305
6.651868
8.758959


BUB1
-3.293346
0.8830545
1.0884485
-0.063545
-1.4635025
0.0802935
-2.173361
-2.5475915
-2.5679685


C1S
6.850023
7.1343975
6.035123
4.263272
8.471663
5.7190985
7.2514145
8.2212235
8.5606875


CYP2E1
7.284587
4.9390935
6.037085
5.811062
10.2536915
1.2878015
8.0876755
9.047509
10.814935


DLG7
-4.7199665
-0.1414205
0.666284
-1.512286
-2.1165725
-0.322455
-3.3904095
-3.848364
-3.34202


DUSP9
NA
-4.4342765
-3.163581
-8.7756845
-9.6208445
-7.8162765
-10.827291
NA
-7.1111525


E2F5
-2.4002515
1.399564
1.206766
-2.426129
-1.1944835
-0.0686475
-0.7133385
-1.4330655
0.049846


GHR
2.2402875
0.2426
-2.353691
-2.9035
4.5756335
0.71981
2.416651
3.7226655
1.9012935


HPD
9.656029
4.473096
0.6808655
5.7101575
10.6864405
4.0108195
9.8859985
9.583194
9.1845675


IGSF1
-7.466951
0.0722075
-6.0490105
-2.4248235
NA
-2.954514
-5.6986975
-7.200325
NA


NLE1
-1.64183
-0.321593
-0.386649
-1.3815525
-1.118745
-1.618369
-1.9449755
-1.823275
-1.770127


RPL10A
5.178571
6.8777395
7.068098
5.9464565
7.542193
6.309556
7.194012
5.9526365
7.4507165




















HC26
HC27
HC28
HC30
HC32
HC34
HC37
HC38
HC41





AFP
-5.69175
-0.626755
NA
6.4370325
0.0037145
-6.6945705
-1.3519745
4.053435
-2.7156435


ALDH2
5.0135775
5.6309605
1.913778
3.8476295
6.802666
5.11617
5.808058
4.596143
6.3503265


AP0C4
0.2581675
1.53158
-6.0251725
0.2797975
2.574347
0.5860455
-0.0768065
-0.129322
2.281983


APCS
7.2072275
7.2809855
1.0475505
7.1142435
7.500133
7.134934
6.755895
5.045701
5.612517


AQP9
3.8645965
5.4736555
0.9613895
5.0250435
7.530391
6.9427395
6.3416265
6.0302545
7.8444565


BUB1
0.545363
-0.8889165
-5.7426525
-0.190936
-5.1317805
-1.2674215
-2.4955985
0.321483
-0.587016


C1S
7.2351705
8.172076
4.910584
7.5279395
7.854502
7.719763
6.921051
6.101331
6.88808


CYP2E1
0.671071
8.6350095
3.6858305
7.5682115
9.4408715
8.545814
10.1686795
8.1123675
9.5090495


DLG7
-0.9710395
-2.3158215
NA
-0.189092
-5.7080765
-2.339621
-2.6534895
-1.4386515
-1.840185


DUSP9
-8.5287915
-10.241011
NA
-9.0027
-9.73163
-9.9728495
NA
-5.2298755
-8.727439


E2F5
-1.1845665
-0.4045835
-4.334386
1.0623035
-0.054818
-1.4281575
-1.2212655
-0.037887
0.466649


GHR
1.964045
2.623084
-1.9788575
2.635437
2.0027475
1.563203
2.9415775
0.2025015
1.428749


HPD
7.6403735
9.597772
3.3142495
7.537
9.0015185
8.3685675
10.367265
7.547286
8.0015745


IGSF1
-5.4960635
-5.588995
NA
-2.651022
NA
-10.112616
-7.5570255
-0.680358
-7.243446


NLE1
-1.851733
-1.851285
-2.4559905
-1.2674865
-1.208576
-1.934745
-1.9881245
-2.1250395
-0.15624


RPL10A
5.9670715
7.6623025
5.521873
7.5046195
8.8437815
6.594006
6.901637
5.1574215
7.7043325






HC42
HC43
KC44
HC52
HC58
HC60
HC64
HC66
HC101





AFP
-5.216493
-1.7983435
-0.564605
10.3337105
1.891958
7.624821
5.0266755
3.156328
-6.873135


ALDH2
4.4086495
5.457548
7.1344115
2.1920375
2.1172735
3.6860195
4.992107
3.8408415
4.339036


AP0C4
-0.627239
-0.7055185
0.499817
-8.124407
-11.8524
-0.545509
0.7860345
-0.6773785
-0.5787185


APCS
4.1054755
7.607914
7.567581
5.9818015
-4.1106695
8.100997
7.4148835
8.2106815
6.288568


AQP9
6.063786
4.7175855
6.058158
-0.4848805
-2.817265
6.8503395
7.0526325
6.2767975
4.6233735


BUB1
-2.224818
-2.8634735
-3.5668895
-1.2986035
1.9395175
-0.576028
-1.367463
-1.1272665
0.081457


C1S
6.3060565
7.9862115
8.547705
5.6337865
3.691331
8.167253
7.1364365
8.026875
7.321092


CYP2E1
9.1411555
8.760714
9.1133175
1.7693015
-4.3317445
9.1875325
9.682147
8.601088
5.806032


DLG7
-3.2531575
-4.2390495
-4.814388
-2.599359
0.1957495
-2.2644225
-2.386875
-2.7680135
-1.3084655


DUSP9
NA
-10.525647
NA
-3.8059605
-3.656912
-6.618755
-7.3184655
-11.5673955
-8.828389


E2F5
-0.3673235
-0.894345
-1.894272
0.4419525
0.804087
-0.432422
-0.2876185
-0.968982
-1.871516


GHR
-1.2545195
3.2916395
4.5598275
-1.843696
-3.7242975
-1.4079225
0.349645
-1.2501855
0.1466275


HPD
8.2669835
8.997825
9.158005
2.481945
1.8257985
8.4643875
8.6027575
8.5231325
5.7252795


IGSF1
-2.899766
-5.5544715
-5.769786
2.254168
1.3471695
-0.7884805
-3.3382005
-9.185554
-4.1394545


NLE1
-0.9401045
-1.8422595
-2.0303285
-1.9474305
-1.209522
-1.9133155
-1.817699
-1.962008
-1.4546305


RPL10A
5.577659
5.480403
5.8488475
5.6154705
6.0601515
5.7041285
6.4617635
5.415169
6.144011






HC102
KC103
HC104
HC105
HC106
HC107
HC108
HC109
HC110





AFP
-4.119697
1.6193685
5.5094265
2.3444245
-3.42054
-4.136209
-4.500336
-4.833024
-3.5240185


ALDH2
2.476355
3.889904
4.936239
4.239726
6.1642895
6.7443095
3.6076385
5.8617665
3.6707715


AP0C4
-5.453696
-0.54698
-0.5059805
-3.577778
-0.7836775
4.4534435
-2.478085
0.729565
-0256479


APCS
-2.3952165
6.014572
5.624234
7.703333
7.8462545
9.2080655
7.275462
6.222909
5.043319


AQP9
0.0196725
7.151639
0.501258
4.2748785
5.85931
8.8878655
4.4353395
6.4504115
4.5999895


BUB1
-0.5553155
-2.086008
-1.311194
0.945674
-4.8909655
-1.7415115
-0.3807995
-2.2918285
-1.449943


C1S
5.939374
5.965432
6.716137
7.774455
8.060072
9.2061165
7.1031155
7.406001
6.9163195


CYP2E1
-2.8566735
8.266311
9.0888685
5.698899
9.9949555
9.3234825
3.889942
8.7101925
7.1 45766


DLG7
-2.1385165
-2.957914
-1.821739
-0.814912
-6.2678815
-1.357756
-2.2445545
-3.222524
-2.333076


DUSP9
-8.6628475
-12.521336
-5.396553
-5.4214725
-11.174152
-6.6136855
-8.0946735
-10.4709205
-11.616244


E2F5
0.830934
-1.8003215
-2.305498
2.0730715
-2.208171
2.78876
0.0923905
-1.9924345
-2.512512


GHR
0.947389
0.636723
1.6860905
0.682142
5.342392
2.935929
1.6363755
2.9233285
1.0803015


HPD
0.568809
6.717282
8.46781
2.288109
9.4440475
10.460972
2.9674235
7.8859205
8.1908235


IGSF1
-2.708733
-9.802921
0.1438735
-1.422332
-7.401009
NA
-7.967992
-10.0122565
-8.1469415


NLE1
-1.1534675
-2.594702
-1.610158
-0.471391
-1.968983
-0.000835
-0.932052
-2.6102395
-2.3529485


RPL10A
5.283399
4.423835
6.21159
6.315756
5.769397
8.6686655
5.818028
5.541229
5.245476






HC111
HC112
HC113
HC114
HC119
HC120
HC121
HC122
HC123





AFP
-1.883473
-2.8803905
1.208649
-5.4433695
1.0580855
-4.0065425
-4.254961
-2.3763095
0.821555


ALDH2
3.8304065
4.8726745
4.407016
4.7113965
6.159706
4.257398
4.556431
6.2844515
4.220769


AP0C4
-1.130067
-0.7777655
-2.366969
-0.833543
1.894453
-3.5241745
-2.167313
1.279577
-0.68167


APCS
5.976754
6.764675
5.197177
6.723142
9.375177
5.6838965
6.2688205
6.9942545
5.778659


AQP9
4.1657805
5.2735435
2.681192
4.445291
7.6266135
6.8239115
4.38702
6.8198535
6.410177


BUB1
0.621548
0.3135015
-3.4825665
-1.7431855
-0.797564
-0.0740105
-2.4486685
-6.0183915
-1.190323


C1S
6.278164
7.455794
6.338901
7.866014
9.1461175
8.5708615
8.118416
7.7653135
5.383781


CYP2E1
4.46942
2.5741475
6.443846
7.3429245
7.095824
7.6044515
7.765037
9.450349
8.528543


DLG7
-0.769283
-0.9196845
-4.5602875
-3.1500875
-1.712686
-1.9563135
-2.852561
-7.228946
-2.929576


DUSP9
-9.137462
-10.105965
-7.8299455
-11.804112
-9.106547
-5.8119685
-9.706684
-9.9054825
-11.584458


E2F5
1.045678
0.0373705
-2.82243
-0.0450475
-0.0248045
1.229768
-0.910943
-3.5033365
-0.646839


GHR
1.1576425
2.5391085
2.16232
2.5053965
3.7649595
3.196589
2.2774645
2.400201
-1.810364


HPD
7.245347
7.714358
6.685692
6.835254
9.220498
8.5127155
7.480725
8.7301975
4.7774665


IGSF1
-1.86965
-3.4428695
-2.045068
-5.1813245
-5.39017
-9.404196
-5.980435
-8.6480295
-5.1400615


NLE1
-1.012752
-1.119237
-2.156348
-1.3170345
-0.400823
-1.1096815
-1.758163
-2.2430545
-1.5951645


RPL10A
5.568205
6.1905075
5.8884625
5.795905
7.954231
6.4517175
6.4042545
5.199782
4.7323885






HC124
HC125
HC126
HC127
HC128
HC129
HC131
HC132
HC133





AFP
3.9525335
-4.806564
-5.899437
-0.0390765
5.8636305
-3.430757
-1.491189
5.4265205
-5.1621395


ALDH2
4.027289
4.5451465
5.02839
2.41699
5.085525
4.6298475
5.425994
3.105643
4.2462915


AP0C4
-0.0499065
2.6326775
0.407895
0.8680995
-0.626498
-1.863955
2.4702
-6.9974515
0.63156


APCS
5.391271
6.5321595
5.2838365
4.846116
5.087517
4.8448705
8.6617295
-3.2748865
7.145861


AQP9
4.463488
8.370224
3.6163545
1.8613935
4.3184915
2.870839
7.4772145
3.9244375
6.05182


BUB1
-1.592563
1.1627945
-2.6943025
-2.048769
-1.3297375
-2.3688215
-0.727709
0.2895395
-4.9277675


C1S
5.151686
8.4244055
7.1365955
6.3641695
6.828468
7.302922
7.525072
4.390082
7.3188145


CYP2E1
9.520436
9.426232
5.226091
6.1813065
7.4344035
2.692798
8.98645
7.0455735
8.1908895


DLG7
-2.03781
0.3286545
-3.944339
-2.96212
-2.6299155
-3.6405185
-1.461713
-1.5572645
-5.5447335


DUSP9
-8.81055
-9.3740615
-8.7174575
-8.672372
-8.499355
-7.0627455
-8.415907
-3.3843145
-8.022457


E2F5
0.574165
-0.028878
-3.271927
-2.162802
-4.393094
-0.470421
0.154573
1.9018925
-2.6341525


GHR
2.2369305
0.697866
1.824385
0.129431
1.9716885
2.332961
4.009655
1.7710325
2.2298335


HPD
7.832169
5.7813
1.865621
3.4481965
5.7052855
5.502918
8.960383
2.3653865
6.1281315


IGSF1
-1.4450915
-10.2234745
-7.659377
-3.1503205
-2.72995
-5.692623
-7.5832005
-1.947055
NA


NLE1
-0.1499775
-0.405397
-2.033278
-2.205965
-1.949352
-1.683808
-1.5313675
0.2035885
-1.4173895


RPL10A
6.691521
7.1196575
5.389272
4.3385115
6.6181545
4.8697295
6.775249
6.7796075
5.762015















AFP
HC134
HC135
HC136
















ALDH2
2.8738695
-0.909107
-0.4105125



AP0C4
4.061101
2.7442165
6.0408575



APCS
-0.1134065
-0.7630605
0.7390785



AQP9
7.5103485
0.959726
7.150737



BUB1
5.550642
4.0595615
5.996196



C1S
1.7425995
-1.2018365
-4.288554



CYP2E1
8.4609335
4.667223
8.243333



DLG7
7.859701
4.30592
9.042865



DUSP9
0.8148735
-2.250305
-5.5267715



E2F5
-4.96739
-5.794605
-10.9307725



GHR
3.1030595
0.986165
-2.4040865



HPD
1.3138565
-0.6955465
4.013948



IGSF1
7.231144
6.7262275
8.223611



NLE1
-0.3848995
-4.394354
-7.4962365



RPL10A
0.794433
-0.9780515
-2.426321



AFP
7.7140665
6.689595
5.5069335









NA: non available







Data were then analyzed by unsupervised clustering (dCHIP software) using 2 methods: average and centroid. Tumors were clustered into 2 groups, C1 and C2. Most of the samples have been attributed the same classification using the 2 methods, except for 6 samples (9%) that have been attributed a different classification (Table 15).


Clinical Parameters Associated to the C1 and C2 Molecular Subclasses


The clinico-pathological parameters of patients and tumors were compared between the two groups C1 and C2, using student's t test and Kaplan-Meier estimates. Since some data are not available for 3 patients, the following statistical studies were performed on 64 tumors.


Survival Analysis


There is a strong correlation of the molecular classification into C1 and C2 with patient's survival by using both classifications (Log rank: Centroid p=0.020 and Average p=0.024) (FIG. 10). In this figure, censored cases indicate the end of the follow-up (the last visit) for individual cases. Probability of survival at two years is 78% for C1 subclass and 39% for C2 subclass (the follow-up may be less than 2 years for some patients).


Association of HCC Classification with Clinical Variables


Table 14 shows the correlation between some clinical variable and the classification of the tumors.

TABLE 14VariableC1C2p-valueTumor grade >2 (Edmonson)13/2921/23<0.0001Moderately-poorly differentiated (OMS)17/3623/25<0.0001Macrovascular Invasion 6/30 9/210.074Microvascular Invasion13/3215/220.043Recurrence7/36 5/25ns
(ns: non-significant)









TABLE 15










Classification of samples by unsupervised clustering


(dCHIP software): average and centroid methods.












Tumor ID
average
centroid
comparison







HC1
C1
C1
Same



HC10
C2
C2
Same



HC11
C1
C1
Same



HC12
C1
C1
Same



HC14
C1
C1
Same



HC15
C2
C2
Same



HC17
C2
C2
Same



HC18
C2
C2
Same



HC20
C1
C1
Same



HC21
C2
C2
Same



HC22
C1
C1
Same



HC23
C1
C1
Same



HC25
C1
C1
Same



HC26
C1
C2
Different



HC27
C1
C1
Same



HC28
C2
C2
Same



HC3
C1
C1
Same



HC30
C2
C2
Same



HC32
C1
C1
Same



HC34
C1
C1
Same



HC37
C1
C1
Same



HC38
C2
C2
Same



HC4
C1
C1
Same



HC41
C1
C1
Same



HC42
C2
C1
Different



HC43
C1
C1
Same



HC44
C1
C1
Same



HC52
C2
C2
Same



HC58
C2
C2
Same



HC6
C1
C1
Same



HC60
C2
C2
Same



HC64
C2
C2
Same



HC66
C1
C1
Same



HC7
C2
C2
Same



HC8
C2
C2
Same



HC9
C2
C2
Same



HC101
C1
C2
Different



HC102
C2
C2
Same



HC103
C1
C1
Same



HC104
C2
C2
Same



HC105
C2
C2
Same



HC106
C1
C1
Same



HC107
C1
C1
Same



HC108
C1
C1
Same



HC109
C1
C1
Same



HC110
C1
C1
Same



HC111
C2
C2
Same



HC112
C1
C2
Different



HC113
C2
C2
Same



HC114
C1
C1
Same



HC119
C1
C1
Same



HC120
C1
C1
Same



HC121
C1
C1
Same



HC122
C1
C1
Same



HC123
C2
C1
Different



HC124
C2
C2
Same



HC125
C1
C1
Same



HC126
C1
C1
Same



HC127
C2
C2
Same



HC128
C2
C2
Same



HC129
C1
C2
Different



HC131
C1
C1
Same



HC132
C2
C2
Same



HC133
C1
C1
Same



HC134
C2
C2
Same



HC135
C2
C2
Same



HC136
C1
C1
Same










In a second analysis, the global set of 64 tumors was analyzed independently of the C1/C2 classification, for parameters associated to survival. Results are presented in Table 16.

TABLE 16VariableLog rankTumor grade >20.108Mod-poor Duff. Degree0.400Macrovasc. mv.0.004Microvasc. mv.0.026recurrencensTumor size 2cm0.397Score METAVIR ActivitynsScore METAVIR Fibrosis0.038<2 vs. ≧ 2 (variable 3)Chronic hepatitis0.948HBV0.093HCV0.352Alcohol0.225
(ns: non-significant)


These results demonstrate that the methods and the signatures of the invention are able to determine the grade not only of HB tumors but also of HCC tumors. The inventors have shown that hierarchical clustering is an efficient method for classification of tumor grade especially for HB. For HCC, this method may be less sufficient (less robust) when the amplitude of variation of expression results of the genes is less important than for HB.


Classification of Hepatoblastomas and Hepatocellular Carcinomas Using the Method of Discretization of Continuous Values.


85 hepatoblastomas (HBs) and 114 hepatocellular carcinomas (HCCs) including to the samples used in the above examples have been analyzed by quantitative PCR using the 16-gene signature and have been classified by the method of discretization of continuous values in order to determine their tumor grade.


Description of the Methodology for Classification


The inventors have designed a methodology for classification based on the principle of discretization of continuous values which refers to the process of converting continuous variables to “discretized” or nominal sets of values.


The major advantage of the discretization method relies on the definition of a cut-off for codification of each qPCR value (either by the Taqman or by the SybrGreen method), which provides an intrinsic score to directly classify an individual sample. There is hence no requirement to compare a sample to a large series of samples. In contrast, in other classification methods, the assigned subclass (such as C1 or C2 disclosed herein) is relative to the values obtained in a large number of cases. Moreover, the use of the average discretized values allows to tolerate missing values when analyzing the qPCR results (i.e. missed amplification of one of the genes for technical reasons).


Using the qPCR data of the 16 genes normalized to the reference RHOT2 gene (−deltaCt values), a cut-off (or threshold) has been defined for each gene. The −deltaCt values are converted into discrete values “1” or “2” depending on an assigned cut-off. In order to privilege the identification of samples that display strong overexpression of proliferation-related genes and/or strong downregulation of differentiation-related genes, the cut-offs have been defined as follows:


for the 8 proliferation-related genes (AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE1, RPL10A), all −DeltaCts with a value above the 67th percentile have been assigned discretized value “2”, otherwise the assigned value was “1”.


for the 8 differentiation-related genes (ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, GHR, HPD), all −deltaCts with a value below the 33rd percentile have been assigned discretized value “1”, otherwise the assigned value was “2”.


Classification of 85 Hepatoblastomas (HB)


RNA Preparation and Quantitative PCR


RNA was extracted by using either Trizol, RNeasy kit (QIAGEN) or miRvana kit (Ambion), then quantified and quality-checked by Agilent technology.


For quantitative PCR analysis, the Sybr Green approach was used as described in point E. above. For each cDNA preparation, 1 μg of RNA was diluted at the final concentration of 100 ng/μl, and reverse transcribed with the Superscript RT kit (Invitrogen) following the manufacturer's protocol. Random primers were added at the final concentration of 30 ng/μl and the final volume was 20 μl. The cDNA was diluted 1:25, and 5 μl were used for each qPCR reaction. We added 5 μl of 2×Sybr Green Master mix (Applied Biosystems) and 0.3 μl of each specific primer (disclosed in point H. above) (final concentration 300 nM). Each reaction was performed in triplicate. qPCR reactions were run on the Applied Biosystems 7900HT Fast Real-Time PCR System with a 384-well thermo-block, and the conditions were the following:


2 min at 50° C. to activate Uracil-N-glycosylase (UNG)-mediated erase of a specific reaction


10 min at 95° C. to activate the polymerase and inactivate the UNG


40 cycles:


15 sec at 95° C. denaturation step


1 min at 60° C. annealing and extension


Final dissociation step to verify amplicon specificity.


The normalized qPCR (deltaCt) values of the 85 HB samples are given in Table A.


Analysis of qPCR Data.


Assignment of a discretized value for the 8 proliferation-related genes (“AFP” “BUB1” “DLG7” “DUSP9” “E2F5” “IGSF1” “NLE” “RPL10A”) was based on the 67th quantile (i.e. percentile), given that around ⅓ of HB cases overexpress proliferation genes, which is correlated with tumor aggressiveness and poor outcome. Assignment of a discretized value for the 8 differentiation-related genes (“ALDH2” “APCS” “APOC4” “AQP9” “C1S” “CYP2E1” “GHR” “HPD”) was based on the 33rd quantile, given that around ⅓ of HB cases underexpress differentiation genes, which is correlated with tumor aggressiveness and poor outcome.


The cut-offs (or thresholds) selected for the −deltaCT value of each gene were determined after considering said chosen percentiles for each group of genes are as follows:


AFP: 3.96139596; ALDH2: 4.3590482; APCS: 4.4691582; APOC4: 2.03068712; AQP9: 3.38391456; BUB1: −1.41294708; C1S: 4.24839464; CYP2E1: 6.70659644; DLG7: −3.3912188; DUSP9: 2.07022648; E2F5: −0.72728656; GHR: −0.1505569200; HPD: 2.27655628; IGSF1: 0.1075015200; NLE: −0.02343571999; RPL10A: 6.19723876


For the sample, the relative expression value is determined for each gene of the set of profiled genes. Each value is compared to the cut-off for the corresponding gene and is then discretized as a result of its position with respect to said cut-off.


The next step consisted in assigning a discretized score to each sample as follows:


1—the average of the “discretized” values of the 8 proliferation-related genes was determined. The 8 proliferation-related genes are the following: AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE, and RPL10A.


2—the average of the “discretized” values of the 8 differentiation-related genes was determined. The 8 differentiation-related genes are the following: ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, GHR, and HPD.


3—The score for each sample was determined as the ratio between the average of proliferation-related genes and the average of differentiation-related genes.


According to this calculation, a score of 2 is the maximal score for highly proliferating and poorly differentiated tumors, whereas well differentiated and slowly proliferating tumors will have a minimal score of 0.5.


Based on the scores assigned to the 85 HB samples analyzed, cut-offs were identified to separate the samples into relevant subclasses. Two different cut-offs that correspond to the 33rd (0.615), and 67th percentile (0.91) have been assessed, leading to the definition of either 2 or 3 subclasses. These data together with the clinical data of 85 HB cases are given in the Table B.


Statistical Analysis of Clinical Correlations


All statistical correlations were analyzed using the discrete classification into 2 subclasses with the 67th percentile (see 3rd column of the table given in Table B).

Samples withSamples withp-valuesscore <67thscore >67th(chi-Characteristicspercentilepercentilesquare test)Previous C1/0252/5  2/261.0739e−14classificationGender Male/Female28/29 7/210.03368PRETEXT.stage30/2511/150.30367I-II/III-IVDistant Metastasis45/1215/130.015808No/YesVascular invasion38/1711/170.0090345No/YesMultifocality No/Yes38/1815/130.20088Histology34/2216/220.75303Epithelial/Mesenchymalβ-catenin mutation 8/45 8/160.067697No/YesMain epithelial49/7 5/212.33206e−9componentFetal/Other*
*Other = embryonal, macrotrabecular, crowded fetal


The best correlation of the discrete classification was observed with the previous classification into C1 and C2 classes, followed by the main epithelial histological component. The correlation with patients' survival is also excellent, as shown by using the Kaplan-Meier estimates and the log-rank test. Illustrative Kaplan-Meier curves are given in FIG. 11 for specific cancer-related survival, using different percentiles to classify the tumors.


In conclusion, this study shows that the discretization method allows to classify hepatoblastoma as efficiently as the previously described method.


A similar approach was therefore applied to the analysis of hepatocellular carcinoma.


Analysis of 114 Hepatocellular Carcinomas (HC)


RNA Preparation


RNA was extracted by using either Trizol, RNeasy kit (QIAGEN) or miRvana kit (Ambion), then quantified and quality-checked by Agilent technology.


For each cDNA preparation, 1 μg of RNA was diluted at the final concentration of 100 ng/μl, and reverse transcribed with the Superscript RT kit (Invitrogen) following the manufacturer's protocol. Random primers were added at the final concentration of 30 ng/μl and the final volume was 20 μl. The cDNA was diluted 1:25, and 5 μl were used for each qPCR reaction. We added 5 μl of 2×Sybr Green Master mix or the Taqman Master mix (Applied Biosystems) and specific primers (and probes when using Taqman chemistry) at the concentration indicated by the manufacturer. Each reaction was performed in triplicate. qPCR reactions were run on the Applied Biosystems 7900HT Fast Real-Time PCR System with a 384-well thermo-block, and the conditions were the following:


2 min at 50° C. to activate Uracil-N-glycosylase (UNG)-mediated erase of aspecific reaction (omit if using the Taqman approach)


10 min at 95° C. to activate the polymerase and inactivate the UNG


40 cycles:


15 sec at 95° C. denaturation step


1 min at 60° C. annealing and extension


Final dissociation step to verify amplicon specificity (omit if using the Taqman approach)


Quantitative PCR


Real time RT-PCR was performed for 16 genes on 114 HCC samples using two different technologies:


Sybr Green as described above for hepatoblastoma (26 samples).


Taqman methodology (88 samples) using primers and probes designed and publicly released by Applied Biosystems company.


Examples












AFP forward primer:
GCCAGTGCTGCACTTCTTCA





AFP reverse primer:
TGTTTCATCCACCACCAAGCT





AFP Taqman probe:
ATGCCAACAGGAGGCCATGCTTCA





RHOT2 forward primer:
CCCAGCACCACCATCTTGAC





RHOT2 reverse primer:
CCAGAAGGAAGAGGGATGCA





RHOT2 Taqman probe:
CAGCTCGCCACCATGGCCG






Each reaction was performed in triplicate for Sybr Green protocol and in duplicate for the taqman protocol. qPCR reactions were run on the Applied Biosystems 7900HT Fast Real-Time PCR System with a 384-well thermo-block.


Raw data for each gene were normalized to the expression of the ROTH2 gene, providing the deltaCt values that were then used for tumor classification into subclasses using the discretization method.


The normalized qPCR values (deltaCt) of the 16 genes in 26 HCC samples analyzed by the Sybr Green approach is given in Table C. The deltaCt values for 88 HCCs analyzed by the Taqman approach are given in Table D.


Analysis of qPCR Data.


The −deltaCt values for each gene in each sample was used. The cut-offs (or thresholds) selected for each gene using the Taqman method or the SybrGreen method are as follows:

Table E of cut-offs for discretization valuesGene nameCut-off for TaqmanCut-off for SybrGreenAFP−1.2634010−2.3753035ALDH24.0141435.314302APCS5.61429076.399079APOC4−0.79631584.656336AQP94.28360115.446966BUB1−1.2736579−3.634476C1S6.35146796.240002CYP2E16.95624195.829384DLG7−2.335694−4.614352DUSP9−7.979559−1.8626715E2F5−0.4400218−1.367846GHR1.08326321.169362HPD6.74803286.736329IGSF1−4.84177857.6653982NLE−1.6167268−1.82226RPL10A6.24830565.731897


For the sample, the relative expression value is determined for each gene of the set of profiled genes. Each value is compared to the cut-off for the corresponding gene and is then discretized as a result of its position with respect to said cut-off.


The next step consisted in assigning a score to each sample as follows:


1—the average of the “discretized” values of the 8 proliferation-related genes was determined. The 8 proliferation-related genes are the following: AFP, BUB1, DLG7, DUSP9, E2F5, IGSF1, NLE, and RPL10A.


2—the average of the “discretized” values of the 8 differentiation-related genes was determined. The 8 differentiation-related genes are the following: ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, GHR, and HPD.


3—The score for each sample was determined as the ratio between the to average of proliferation-related genes and the average of differentiation-related genes.


According to this calculation, a score of 2 is the theoretical maximal score for highly proliferating and poorly differentiated tumors, whereas well differentiated and slowly proliferating tumors will have a theoretical minimal score of 0.5.


Based on the scores assigned to the 114 samples analyzed, cut-offs are identified to separate the samples into relevant subclasses. Three different cut-offs that correspond to the 30rd (0.66), 50th (0.8125) and 67th percentile (0.925) have been assessed, leading to 4 different classification methods.

TABLE Fof discretized values for 114 HCCs using 3 different thresholdsand 4 combinationsMethod 13-class:(1): <q30Method 2Method 3Method 4(2): q302-class:2-class:2-class:Over-Follow-q67;(1): <q30(1): <q67(1): <q50;all.survi-upSamplescore(3): >g67(2): >q30(2): >q67(2): >q50val(years)HC 0010.6875221110.07HC 0030.6875221113.33HC 0040.72727272211011.48HC 0060.8125221211.25HC 0071.4545455322211.5HC 0081.0769231322218.48HC 0091.75322210.02HC 0101.5322210.95HC 0110.64285711111012.2HC 0120.5714286111110.05HC 0140.625111111HC 0151.6322211.22HC 0171.8753222010.96HC 0181.5322210.39HC 0200.78571432211015.4HC 0211.5555556322210.7HC 0220.56251111011.5HC 0230.51111011.93HC 0250.71428572211115.87HC 0260.7142857221110.83HC 0270.8125221210.1HC 0281322210.1HC 03013222112.4HC 0320.7857143221110.66HC 0340.6251111015.7HC 0370.5714286111110.2HC 0381.0769231322211.12HC 0410.8666667221217.44HC 0420.87912092212010.58HC 0430.51111010.9HC 0521.33333333222NA0.25HC 0581.875322208.3HC 06013222NANAHC 0640.8666667221215.25HC 0660.7142857221108.93HC 1010.9230769221202.5HC 1021.625322200.1HC 1030.75221101.82HC 1040.8666667221202.1HC 1051.4545455322200.56HC 1060.5111102HC 1070.8571429221201.75HC 1081322201.62HC 1090.5111101.3HC 1100.6923077221101.95HC 1111.1818182322210.7HC 1120.8666667221201.48HC 1131.1322211HC 1140.6666667221100.44HC 1150.875221200.75HC 1160.9333333322200.69HC 1170.6111101.2HC 1180.5111100.93HC 1190.8461538221201.2HC 1201322200.82HC 1210.9285714322200.6HC 1220.6666667221100.75HC 1231322200.8HC 1240.7857143221100.52HC 1250.8181818221200.9HC 1260.8125221200.42HC 1271.6322200.25HC 1280.6095238111100.44HC 1291322210.15HC 1301.7777778322200.14HC 1310.5625111100.26HC 1371.2222222322205.67HC 1380.75221105.58HC 1391.3333333322206HC 1400.5714286111104.17HC 1410.6153846111105.08HC 1420.8888889221214.08HC 1431.375322202.83HC 1440.6153846111106HC 1450.8221105.58HC 1460.9221204.33HC 1470.6666667221103.83HC 1481.1322203.08HC 1491.2222222322213.42HC 1500.6666667221105.42HC 1510.6153846111102.25HC 1520.6428571111113.67HC 1530.6923077221114.83HC 1541.375322212.21HC 1550.8181818221204.1HC 1561.4322212.31HC 1571322213.59HC 1590.7272727221112.42HC 1610.6111104.47HC 1621.1111111322203.49HC 1630.6111112.21HC 1640.6428571111104.54HC 1650.6428571111104.72HC 1680.6111106HC 1690.6111112.78HC 1700.5625111105.29HC 1710.8181818221204.57HC 1720.8333333221203.9HC 1730.6428571111104.21HC 1760.6428571111104.57HC 1770.6666667221105.42HC 1780.7142857221102.5HC 1790.8181818221205.17HC 1800.8571429221213.58HC 1811322206.83HC 1820.5625111103.5HC 1830.7333333221114.08HC 1840.9230769221212.08HC 1850.7692308221102.25HC 1860.9285714322212.17HC 1870.6428571111107.67HC 1880.7142857221104.67HC 1890.8666667221213.25HC 1900.7619048221105.58


Samples were separated into the corresponding subgroups, and subsequent analysis was carried out using the 4 classification methods. Survival for each group was determined using the Kaplan-Meier estimates and the log-rank test.


Statistical Analysis of Clinical Correlations with the Subclasses for 114 HCCs


A complete table with all clinical and pathological data collected for 114 HCC patients is given in Table G. The different parameters are represented as follows:

TABLE HClinical and pathological parameters and molecularclassification of 114 HB cases.CharacteristicsEtiology*Alcohol  40 (36%)HCV  26 (23%)HBV  23 (20%)Hemochromatosis  6 (5%)NASH  6 (5%)Unknown  23 (20%)Treatment (SR, OLT)93/21Chronic viral hepatitist  46 (41%)Liver cirrhosis  44 (48%)Tumor characteristicsMacrovascular invasion  20 (25%)Microvascular invasion  47 (50%)Mean tumor size, cm (range) 7.9 (1.5-22)Multifocality  46 (48%)Histology:Edmonson Tumor grade(1/2/3/4)7/35/47/5OMS Tumor differentiation (W/M/P)51/55/6Classification with 16-genes by discretization40th Percentile (C1/C2)30/8450th Percentile (C1/C2)55/5967th Percentile (C1/C2)77/37Mean follow-up, months (range)43.6 (0.26-146)Tumor recurrence  43 (40%)Alive/DOD75/38
Abbreviations:

HCV, hepatitis C virus;

HBV, hepatitis B virus;

NASH, Nonalcoholic steatohepatitis;

SR, surgical resection;

OLT, orthotopic liver transplantation;

W, well differentiated;

M, moderately differentiated;

P, poorly differentiated;

NA, not available;

DOD, dead of cancer.

*12 cases have more than one etiological agent and data were not available for 2 Gases.

Data were not available for all cases. Percentages were deduced from available data.


In a second step, the intrinsic parameters of the tumors correlated with patients' survival were analyzed. In this series of tumors, only tumor grade (Edmonson) and vascular invasion were significantly correlated with survival.

TABLE ISummary of the clinical variables associated to overall survival(Kaplan-Meier curves and log-rank test). This Table does nottake into account the molecular classificationN.N. patientsLogVariablepatientsLog rankWith PHrankEdmonson Tumor grade940.028730.032(1-2/3-4)Tumor diff. OMS1110.406900.647(Well/Moderate-poorly duff.)High proliferation: >10450.054340.402Mitosis in 10 fields 40×(N/Y)Macrovascular Invasion790.001590.010(N/Y)Microvascular Invasion920.007720.050(N/Y)Tumor size≧10 cm1130.298920.314


Classification by Discretization of Continuous Values


The clinico-pathological parameters were compared between the tumor groups using student's t test and chi-square test. Survival was analyzed by using Kaplan-Meier curves and log rank test. A special attention was given to the classification with the 67th percentile. Follow-up was closed at 146 months for overall survival (OS) and at 48 months for disease-free survival (DFS).

TABLE JAssociation of 16-gene classification by discretization with clinicaland pathological data (chi-square test). Abbreviations: P33, 33rdpercentile, P50 50th percentile and P67, 67th percentile.p-valueP67VariableP33P50P67C1C2commentsEdmonson Tumor0.006<0.001<0.00138/274/2520 cases withgrade: grade 1missing valuesand 2 (welldifferentiated) vs.3 and 4(moderately andpoorly diff.)Tumor0.0060.001<0.00145/326/292 cases with missingdifferentiationvaluesOMS(Well/Moderate-versus poorlydifferentiated)High proliferation:0.0210.0010.00122/7  4/12>10 mitosis in 10fields 40× (N/Y)Macrovascular0.0970.0330.00844/8 16/12The cases defined asInvasion (N/Y)possible areconsidered negative.Microvascular0.0710.0010.00937/26 9/21The cases defined asInvasion (N/Y)possible areconsidered negative.Tumor sizensns0.01557/2019/18Different cut-offs</≧10 cmassessed:2, 3, 5 and 10 cmMultifocality (N/Y)nsnsns35/3015/16Macronodules ofnsnsns24/9 12/4 regenerationNorm Liver A0F0-nsnsns48/1727/7 A0F1Cirrhosis AXF4nsnsns31/2917/15(N/Y)Score METAVIR0.0530.044ns19/32 5/20Activity >0 (N/Y)Score METAVIRns0.20ns31/2015/10Activity >1 (N/Y)Score METAVIR0.041nsns 5/48 2/27Fibrosis >0 (N/Y)Score METAVIRnsnsns19/35 7/22Fibrosis >1 (N/Y)Score METAVIRnsnsns24/30 8/21Fibrosis >2 (N/Y)Score METAVIRnsnsns26/2815/14Fibrosis >3 (N/Y)Chronic viral0.047nsns48/2918/17hepatitis (N/Y)HBV (N/Y)0.075nsns62/1527/8 HCV (N/Y)nsnsns61/1625/10Alcohol (N/Y)nsnsns47/3025/10Recurrence (N/Y)nsnsns41/3224/11HCC034 andHCC030 censoredSurvival (N/Y)0.0500.0230.03156/2119/17HCC025 andHCC030 censoredDFS (N/Y)nsnsns35/4215/21HCC025 andHCC030 censored


In conclusion, these data show significant correlations between molecular classification using the 3 methods and the following parameters: Tumor grade (Edmonson), tumor differentiation (OMS), proliferation rate, vascular invasion and survival. In contrast, the classifications were not correlated with etiological factors (viral hepatitis, alcohol, etc. . . . ), with the state of the disease in adjacent, non tumoral livers or with tumor recurrence.


The data suggest that classification using the 67th percentile seems to be the most adequate and is strongly recommended to classify HCCs.


Multivariate Analysis


To further determine the efficiency of the molecular classification using the 67th percentile, we performed multivariate analysis with the Cox regression test on two sets of patients for which all data were available:


91 patients that received either surgical resection or orthoptic liver transplantation (OLT)


71 patients that received surgical resection.


Different variables associated to survival in the clinical settings have been included in the multivariate analysis: 1) Edmonson grade, 2) microvascular invasion and 3) Molecular classification using the 67th percentile.

TABLE KMultivariate test (Cox regression).NpatientsvariableHR95% CIp-value91Molec classsif (p67)2.534(1.214-5.289)0.016(surgicalEdmonson Tumor grade1.690(0.747-3.823)0.205resections1-2/3-4)and OLT)Microvascular Invasion2.451(1.105-5435)0.024(N/Y)71Molec classsif (p67)2.646(1.1156.278)0.032(onlyEdmonson Tumor grade (1-2.697(1.103-6.592)0.026surgical213-4)resections)Microvascular Invasion1.681(0.648-4.359)0.282(N/Y)


Correlation of the Molecular Classifications with Survival


For overall survival (OS) and disease-free survival (DFS), we compared the efficiency of the 3 methods of discretization that separate the samples into 2 subclasses. Independent studies were made for patients that received surgical resection and for patients that received orthoptic liver transplantation (OLT). The ability of the 16-gene signature to discriminate between recurrent and non-recurrent tumors was also assessed.

Table LSummary of survival analysis using Kaplan-Meiercurves and log-rank testAnalysisN. patientsClassif. methodLog rankOS113P330.037113P500.005113P670.002DFS113P330.078113P500.019113P670.072recurrence108p33*0.134*108p50*0.115*108P671.000Analysis of 92 cases that received surgical resectionOS92P330.03292P500.00992P670.013DFS92P40ns92P50ns92P67nsrecurrence88P33ns88P50ns88P67ns
Abbreviations: OS, overall survival; DFS, disease free survival

*There is a trend but it is not significant and it is lost in the P60 analysis


The different analyses are illustrated in the Kaplan-Meier plots shown in FIG. 12. The discretization method of classification showed the same efficiency in the analysis of tumors obtained either from surgical resection (also called partial hepatectomy, PH) or from orthotopic liver transplantation (OLT), showing that the clinical management of the tumor had no impact on the classification.


In conclusion, the method described herein is able to classify HCC cases according to tumor grade and patient's survival, and represents a powerful tool at diagnosis to stratify the tumors according to the prognosis, and for further clinical management of HCC. In particular, it may be an excellent tool for the decision of orthotopic liver transplantation, since the criteria used currently are limited and often poorly informative of the outcome.


Protocol for Applying the Method to a New Sample


The following protocol is designed according to the invention:


1—extract total RNA from the tumor specimen using well established technologies.


2—synthesize cDNA synthesis (suggested conditions: 1 μg RNA and 300 ng of random hexamers for a 20 μl-reaction)


3—amplify the selected genes said genes being in equal number of each of the groups defined as overexpressed proliferation-related genes group and downregulated differentiation-related genes group (profiled genes within the group of 2 to 16 genes) and the reference gene (invariant gene) such as for example the RHOT2 gene 1:5 cDNA dilution, using either Taqman or SybrGreen qPCR technology.


4—determine the Delta Ct (DCt) value for each gene


5—compare the value with the threshold of reference (for HB or for HC) in order to assign a discretized value of “1” or “2”.


5—determine the average of discretized values in each group, i.e., for the selected proliferation-related genes (up to 8) separately for and the selected differentiation-related genes (up to 8) and determine the ratio of these 2 average values which is the score of the sample.


6—compare the result with the reference scores corresponding to the following cut-offs:


C1


|30rd=0.6667


|50th=0.8125


|67th=0.925


C2


Example

For patient X having an HC tumor a Taqman qPCR is performed.


Step one: assignment of discretized values to each selected gene among proliferation-related genes and differentiation-related genes.


Example

The DCt of AFP is −4.0523


The cut-off for AFP for qPCR using Taqman technology is −1.2634010 Given that −4.0523 is lower than the cut-off, the assigned discretized value is 2.


Step two: Determination of the average of discretized values for the 2 sets of 8 genes:


AFP=2; BUB1=1; DLG7=2; DUSP9=2; E2F5=2; IGSF1=1; NLE=2; RPL10A=1;


Average of Proliferation-Related Genes: (2+1+2+2+2+1+2+1)/8=1.625


ALDH2=1; APCS=1; APOC4=1; AQP9=1; C1S=2; CYP2E1=2; GHR=1; HPD=2;


Average of Differentiation-Related Genes: (1+1+1+1+2+2+1+2)/8=1.375


Step Three: calculate the ratio proliferation/differentiation score.


In this example: 1.625/1.375=1.18182


Step 4: compare the result with the reference scores:


C1


|30rd percentile=0.6667


|50th percentile=0.8125


|67th percentile=0.925


C2


Classification based on the value of the ratio=1.18182.


As the value is above the 67th percentile, the assigned class is C2.

TABLE AidAFPALDH2APCSAP0C4AQP9BUB1C1SCYP2E1HB1−7.684892−4.592702−0.660189−2.651319-4.194894-1.068025-1.394659-3.334692HB100−7.682724−3.849128−0.3725660.297278−0.3057380.65983−2.572264−7.352142HB1011.801478−7.157316−1.166513−4.924476−8.0678386.222865−5.284734−11.757699HB102−7.761115−5.696697−1.044129−2.374592−3.4470462.724363−3.657616−5.769417HB1032.908026−2.580629−2.748625−2.556351.4806243.891875−2.8193720.454623HB1060.294848−7.534485−1.424535−5.377043−7.8866124.855797−6.80698−11.496242HB1070.719866−6.546079−9.18522−3.425075−6.1896643.901806−5.609115−10.6711555HB111.492805−3.560021−5.094387−1.031623−8.428492.086834−6.166353−9.043371HB1124.155252−6.486961−0.154814−4.48155−5.6345963.762347−7.88579−8.960815HB1146.2971−3.9664565.022660.6042753.0376824.23408−5.29691−0.313326HB1180.318307−4.311795−5.146409−3.787568−5.4284422.329959−5.284827−7.342423HB121−0.971033−6.879043−8.355819−4.679393−6.3614352.329708−6.559457−8.87105HB1222.188721−6.220957−7.7399−3.410743−5.7453063.309004−6.327656−8.906339HB1252.929931−4.053616−4.882212−2.32494−3.3523985.067815−4.255762−7.887455HB1262.458273−5.577951−6.518289−3.182407−5.2433515.270089−5.814672−8.188307HB129−4.930877−2.124281−0.7442621.154663−0.8465720.421372−2.925458−4.708874HB130−4.86199−1.139837−1.3985880.115559−1.3139511.669543−2.372350.175598HB1315.545406−1.714367−1.0456832.6288221.9038531.972112−2.3068180.069456HB1322.654369−3.71955−6.543987−3.876868−4.70994.043489−4.801651−7.725089HB1365.005516−3.234557−4.8272832.471208−0.502385−1.945351−4.324749−4.844765HB1402.835457−7.041546−6.88604−5.561912−5.0896824.140594−6.023758−10.477228HB1425.200474−4.9196162.4168072.058522−3.3961711.380591−5.9651261.196438HB1453.58286−5.186236−5.18731NA−5.1188955.58416−5.786933−7.880334HB146−1.290056−5.422341−5.973879−3.869993−5.9080240.982626−4.124487−8.751883HB147−9.442257−3.655303−0.3621221.179633−2.349782−1.51351−2.7560990.30832HB148−3.566401−5.382548−6.721533−2.380348−6.9513591.183916−4.188648−7.101147HB1502.356994−5.56181−5.496186−4.45536−5.6032475.136577−5.435261−8.522001HB153−2.086302-4.364035-4.049735-1.1908-4.3421862.437297-6.055092-7.522683HB155−1.951256−5.140738−7.17357−0.8013184.5389294.038538−5.9394383.058475HB156−6.523604−4.658012−5.112322−1.499462−1.130311.970226−4.763811−8.138508HB157−8.747252−3.193287−0.9145110.563787−0.1392730.648195−3.089302−2.404646HB1604.40621−0.878277−2.381785−1.95270.7707994.516203−2.895221.197611HB162−1.127062−5.142195−6.564426−2.432348−5.1796013.27157−4.959578−9.351464HB165−1.015428−1.578048−1.612095−1.6774941.921123−0.416058−4.579384−0.458984HB167−7.323435−5.692388−6.461153−2.470512−4.912208−0.369976−4.949694−10.583324HB170−0.980072−5.786627−7.265156−3.690367−5.9529081.548967−6.61768−8.574004HB1712.310988−5.687635−7.127181−3.794631−5.8986352.05689−6.420469−8.856566HB1724.547243−0.385469−1.804453−1.8334782.114424.373205−3.9291511.277285HB1731.889759−5.184791−4.471618−2.235657−5.7430572.116789−4.966413−7.319851HB175−2.0436−6.05152−8.152949−2.996302−3.8292053.036838−5.151913−9.108766HB184−6.561121−2.895788−5.35813−1.6537860.293844−0.082754−3.084271−3.362889HB204.752153−4.811256−5.712608−2.133951−5.3617715.572378−4.283688−8.390209HB28−4.001793−4.719296−7.514733−2.385516−3.8697070.599685−5.187286−9.373678HB30.027392−4.565046−4.462833−2.255273−4.146364.676108−5.373064−6.610781HB33−7.497741−3.066759−5.8812770.2503340.9509660.500246−3.829096−6.510795HB39−8.613403−3.1664273.4217341.699859−0.944463−0.146929−1.480822−0.727464HB48−4.768603−3.632136−4.882397−2.170561−4.9654031.366439−3.944489−9.061667HB491.818606−5.933777−5.948111−4.936781−5.4349314.576628−5.318794−9.381172HB5−2.282703−6.147963−7.059143−4.107155−7.5930992.501017−6.573836−9.813634HB541.132255−4.844075−5.655802−2.937193−4.5954423.040468−4.999207−8.199672HB591.334928−6.792009−7.221196−5.590302−6.3008281.42553−5.648808−9.279234HB6−1.610623−7.099329−7.979286−5.729452−5.26472252.920021−5.482511−10.151809HB60−0.594337−5.206398−6.67766−1.663871−2.8893263.97632−5.504179−6.743858HB61−5.058775−6.113525−5.991888−3.527984−5.3874193.269827−6.119246−8.943929HB62−1.989342−4.487171−6.502588−0.923844−4.7124713.449967−4.22945−7.087853HB63−0.891056−4.153057−5.680458−2.637115−5.7100624.49543−2.939154−9.095241HB653.025127−4.346225−5.338104−1.175748−1.226393−0.613979−5.196916−4.645702HB66-1.861761-4.166485-5.897819-2.09279-3.0032584.774807-4.585607-6.839392HB68−4.313608−6.550704−6.762513−3.66757−5.9826544.060667−5.956246−8.393607HB69−1.820363−9.245314333−8.965648−7.384871667−9.430164667−2.026701667−8.961309−12.31658HB71.334084−4.488213−5.853708−2.13753−5.1429384.894117−4.082335−8.118103HB702.021391−5.678476−7.496267−5.781771−4.3464582.174971−7.066038−8.392057HB72−11.995704673.978023333−1.371737333−2.543168667−6.278723667−5.504070333−7.162789667−8.103601333HB73−10.69629133−8.263771333−4.869197667−2.900671333−5.802080667−5.324255333−8.090371−9.754354333HB74F3.831288−7.73216−4.940396−6.3439−6.3559956.130615−5.584023−10.472842HB750.474553−6.309769−2.777247−4.334006−6.8072994.545387−5.115577−10.418948HB772.915987−5.645872−6.698372−2.284956−5.3923774.544876−5.559466−8.695429HB78−3.945686−2.82555−2.986284−1.790335−0.9387384.523136−2.620165−5.945013HB79−0.781193−5.652768−5.454157−3.953162−5.0514440.254305−5.44242−9.05667HB8−6.696169−3.1089130.4984611.361801−3.3226420.055848−0.348492−1.877119HB80−8.8331005−4.713883−2.9124615−2.810437−0.838727−0.7226515−2.5925445−5.408417HB81−4.851198667−10.55296467−10.55292033−7.621321667−10.19195633−2.962795333−10.17992067−12.72629433HB82−1.942166−5.620028−5.739178−3.972123−6.5204820.934055−3.737063−8.932744HB83−4.169107−9.660034667−9.382586667−8.05219−10.951863−3.521245667−10.12345167−9.850559667HB86−6.283735−5.2876770.896101−1.494853−2.934412−0.46896−2.879366−5.76077HB892.996384−7.323446−7.464817−5.120874−5.8565184.907738−6.676481−9.415603HB9−3.679937−4.761778−6.571455−2.775269−6.2017722.209541−3.895565−8.86438HB902.024206−8.47846−1.33932−6.745716−6.6771225.899195−8.114672−10.459034HB93−4.610162−5.583852−5.277197−1.990982−2.698011−1.085743−4.488914−3.388975HB941.79868−5.621254−7.718202−6.940586−6.673353.551727−6.54809−8.572742HB95−0.444835−5.745006−8.404602−5.637613−6.3960636.671045−5.701559−10.554918HB96−4.775396−6.402052−6.123253−4.340961−5.0666883.365736−6.521753−9.090145HB97−6.841231−6.21691−6.275051−3.638382−3.6175582.362203−6.58495−5.781372HB98−4.911783−2.9469326.4789334.2111470.3959262.311268−2.8278020.584022HB99−4.551378−1.14591−5.549696−1.7968591.629062.600714−2.483835−3.848236HB14.140368−5.212318−0.8124241.2075833.840983−0.715134−0.812792−8.675945HB1004.399124−5.7497060.276981.907294−0.113253−2.8003230.547899−6.153046HB1017.086329−0.6418710.737702−3.913751−4.3402597.0863290.191689−6.757648HB1027.380694−4.3038661.1447780.2784−0.284245−2.5456680.856607−6.803817HB1035.9971430.8804213.6974781.249386−2.7133061.392197−0.453035−4.535615HB1066.79755−1.5407450.77722−4.155098−5.7471642.2743850.291903−6.637275HB1075.239962−1.1842443.145996−1.891404−4.4332713.119114−0.053334−6.319917HB113.688558−1.412987−0.179621−0.149048−1.8976582.297186−0.19686−5.623341HB1126.035002−2.179125−0.998979−3.575994−4.671755−0.776138−2.252113−7.8479HB1146.29712.6158270.8865640.0024871.9193972.508631.785623−7.055851HB1183.9351012.4051052.275962−0.451819−4.8123192.3398130.486307−5.904633HB1213.458157−2.18821.247645−1.155575−5.9382353.7501471.867907−5.131548HB1223.5627771.2297232.386559−1.961029−5.5909192.4066871.976893−5.368023HB1255.7002520.2746422.8648830.118717−3.1552892.138032−0.470879−3.478449HB1266.326020.2741973.089709−1.334371−5.2277052.7265990.54385−4.787822HB1294.474485−3.8297511.1582833.0257281.984295−0.0743541.326073−5.682215HB1305.297728−2.5540082.2511633.3175560.8859620.0393071.389742−4.829542HB1315.8011682.2692722.2269211.2355982.0354525.6211141.777334−4.96776HB1328.180410.4331044.507503−0.157093−2.4414225.8552132.895208−3.579579HB1361.1406860.10165−2.3369470.2612030.1241593.807218−0.676358−7.113232HB1409.015818−0.4012642.325356−3.379816−3.1480683.1564560.80129−7.308986HB1426.2031924.5546313.036612.5988774.1504558.7824611.428955−6.630178HB1456.7342641.9087342.518779−1.358174−5.1816684.6104061.707345−4.6775HB1460.991164−0.6818280.1227−0.510651−4.4714830.7770040.176935−5.992209HB147−1.376061−4.733546−2.5883971.772494−1.944032−2.698708−0.565682−7.527854HB1481.7033−1.806502−0.663069−1.376372−5.121145−0.683001−0.431826−6.201895HB1505.8002330.84362.758596−1.181738−5.4920372.8919370.439392−4.69542HB1533.096912−2.6578620.449197−0.480929−4.2619863.343361.423023−5.963837HB1554.360922−1.232590.752365−3.0624740.657144−1.0910130.911424−5.964497HB1562.483547−1.2142280.687246−1.107338−3.806189−1.1813050.159847−5.65452HB1570.181175−4.14510.2977471.940187−3.850885−1.386230.041349−5.820536HB1606.2245692.9061584.4035452.633949−2.1385693.355814−0.100123−4.568688HB1624.25017−1.4532831.117439−0.163468−4.7338811.809885−0.022627−4.822098HB165−0.0104881.8373050.47467−2.953007−0.655058−1.791164−0.933062−5.535221HB1670.509668−1.7074850.1987420.269552−4.442331−1.197651−0.240385−5.755341HB1702.5672071.1487381.360144−2.397242−4.9444392.424619−0.463297−5.539725HB1712.2783531.674042.062277−1.193735−4.9845522.190980.230044−4.81411HB1726.0604592.3669993.6893412.93017−1.3169212.571021−0.153162−3.812616HB1732.7799991.9214273.05205−0.20919−4.4753760.4188180.678606−4.361307HB1754.414558−1.6232421.49−0.662783−4.6844463.5240491.78088−5.173616HB1841.361379−1.542307−0.5888121.814793−2.048922−0.3263930.097971−4.663763HB209.423325−0.341742.066057−0.975735−3.6958544.3614841.157495−5.27136HB281.922989−2.3048611.222545−0.120436−5.154703−0.1927381.819854−5.824864HB37.2856850.652012.301029−0.0491580.1173734.462211.743745−6.911792HB331.659659−4.338262−0.1482331.134133−4.625204−2.341981.272614−5.63922HB392.485354−4.927491−1.2419311.694781−0.33289−2.652634−0.149609−6.579218HB481.583391−3.620772−0.0890811.342382−2.3302180.6861631.169838−6.508074HB495.6528932.411483.776672−1.220476−5.7467794.7275962.190021−4.286949HB53.674234−2.0824240.98073−1.943451−6.5617911.5921670.449005−6.230808HB543.5562683.9821833.025795−0.158057−4.6383333.6236781.995039−5.061096HB595.1273360.2507533.459226−2.269072−4.7277386.0450931.466312−6.48303HB66.733353−0.2463093.812183−2.459856−3.7289870.8350572.205872−7.208765HB605.1885172.8695443.228365−0.276338−4.0319742.0261162.577353−4.502382HB615.827933−5.514571.00606−3.272672−4.816797−0.2038710.753758−6.140918HB624.3282770.7085121.2189631.021692−3.2651380.7315192.223877−5.334147HB635.003075−1.0820940.9513571.3165532.0006014.9649961.31674−6.741518HB652.978487−0.087486−1.2743880.080222−2.4179461.06702−1.371523−6.195428HB668.039274−0.4233132.141981−1.148424−1.349111−0.3050171.586659−5.393141HB687.010986−0.5305412.5202610.232431−1.779051−0.6031132.342104−4.959414HB693.071106−0.6260596673.421015−5.118794333−6.82405566711.819556−0.603036−2.847600667HB78.076437−0.8330111.354912−0.884629−2.1065922.9787392.384133−5.458546HB704.0835193.8963642.616204−3.614294−6.0630972.0603791.506083−4.669554HB72−1.688566667−8.976227−1.809694−1.750672−3.40203−6.090071333−2.505424−5.054027HB73−2.068555667−9.537516−1.965151−0.544775−5.542041333−7.013002667−3.078154667−5.580986333HB74F8.9860480.4978284.585503−2.916191−3.0419437.7596081.654283−6.380865HB757.231393−2.4118390.378995−1.925637−5.0551062.614561.017432−5.77539HB779.66177−0.1392992.727198−1.675013−4.0799322.7937582.146337−4.964228HB785.293419−0.1856291.7355940.020191−3.984125−2.010153−0.114956−3.94071HB791.903061.1456811.319285−1.978228−5.7573350.01942−0.194167−5.016158HB81.950257−4.043236−1.8146362.2805161.1003530.3146940.29834−7.823095HB802.660644−4.9166885−0.3740310.675995−0.4253495−4.2048885−0.8782055−7.919531HB812.155925333−5.7383630.932455333−5.565798−8.171378−1.999123333−2.092100667−4.795482HB821.47049−3.938165−0.549544−1.023595−3.2674038.0080690.067941−7.635394HB832.492243−4.0039303334.737920667−4.561133333−6.966227667−0.028684333−0.8550546671.789090833HB863.219092−5.894534−0.4966620.35847−0.121981−2.310610.046472−8.510995HB898.2553391.2849163.638735−2.665258−5.1777043.2736491.279167−5.898171HB94.940411−1.9896360.700504−0.698988−3.2556012.6093391.300875−6.54224HB906.548911.1041621.408459−5.754423−7.5074854.450261.52717−6.250036HB933.902565−7.483471−0.4881080.969648−1.415501−1.818147−0.829773−7.824402HB948.669386−1.1323050.4907888.498726−6.8196457.800646−0.149162−5.793072HB956.921267−1.6208692.726241−2.193777−5.4547651.3647380.279802−5.172451HB966.685021−0.5912711.973021−4.924202−4.912831.7225051.829525−5.638435HB976.474525−5.8005371.05047−0.911789−4.571465−4.308964−0.87035−6.60257HB986.837198−2.0654832.4823011.17723−0.98407−0.7010981.175939−5.166874HB996.353711−4.2018281.4675521.703655−0.109186−0.8222661.226265−3.572067



















TABLE B












67th percentile-
percentile-











related
related
previous 16 gene


tumor
score
score
score
based clas-


AFP at diagnosis

text missing or illegible when filed

Treatment
PRETEXT


ID
(ratio)
(2-classes)
(3-classes)
sification
Gender
Age months
ng/mL
treatment
protocol
stage




















HB122
0.5
1
1
C1
M
10
8000
Y
H
I


HB126
0.5
1
1
C1
F
12
153840
Y
S
II


HB145
0.5
1
1
C1
M
7
56000
Y
S
II


HB150
0.5
1
1
C1
F
5
82000
Y
S
III


HB175
0.5
1
1
C1
M
9
220000
Y
S
I


HB20
0.5
1
1
C1
F
50
880
Y
S
II


HB49
0.5
1
1
C1
F
15
11000
Y
S
II


HB54
0.5
1
1
C1
M
10
180
N
N
I


HB70
0.5
1
1
C1
F
42
812
Y
S
II


HB77
0.5
1
1
C1
F
9
204000
Y
S
II


HB89
0.5
1
1
C1
M
13
448
Y
S
I


HB95
0.5
1
1
C1
M
28
1000000
Y
H
IV


HB118
0.53333333
1
1
C1
M
17
14500
Y
S
NA


HB132
0.53333333
1
1
C1
F
23
2078
Y
NA
III


HB121
0.5625
1
1
C1
F
14
296000
Y
S
III


HB140
0.5625
1
1
C1
M
3
22758
Y
S
II


HB162
0.5625
1
1
C1
F
9
960000
Y
S
III


HB171
0.5625
1
1
C1
F
17
300
Y
S
II


HB173
0.5625
1
1
C1
F
27
66810
Y
S
I


HB59
0.5625
1
1
C1
F
24
5643
Y
S
II


HB6
0.5625
1
1
C1
M
24
320000
Y
S
II


HB74F
0.5625
1
1
C1
M
96
325
N
N
I


HB96
0.5625
1
1
C1
M
101
2265000
Y
H
IV


HB60
0.57142857
1
1
C1
F
30
1990800
Y
H
II


HB7
0.57142857
1
1
C1
M
33
45000
Y
S
I


HB101
0.6
1
1
C1
M
42
67747
Y
S
III


HB106
0.6
1
1
C1
F
11
320000
Y
H
IV


HB90
0.6
1
1
C1
F
74
300
N
N
II


HB62
0.61538462
1
2
C1
M
16
1708400
Y
H
IV


HB107
0.625
1
2
C1
M
30
16000
Y
H
IV


HB170
0.625
1
2
C1
M
20
123000
Y
H
III


HB5
0.625
1
2
C1
M
84
300000
Y
H
III


HB125
0.64285714
1
2
C1
F
15
360000
Y
H
IV


HB75
0.66666667
1
2
C1
M
21
131000
Y
S
II


HB9
0.66666667
1
2
C1
F
16
84000
Y
NA
III


HB94
0.66666667
1
2
C1
M
29
1270
Y
S
I


HB61
0.6875
1
2
C1
F
126
346000
Y
NA
IV


HB69
0.6875
1
2
C1
M
25
1163
Y
S
I


HB79
0.6875
1
2
C1
M
144
1200
Y
S
II


HB3
0.69230769
1
2
C1
F
22
3192
Y
S
I


HB66
0.69230769
1
2
C1
M
6
1000000
Y
S
III


HB68
0.71428571
1
2
C1
F
11
119320
Y
S
III


HB146
0.73333333
1
2
C1
F
11
NA
N
N
NA


HB155
0.75
1
2
C2
M
9
849500
Y
S
II


HB63
0.75
1
2
C1
M
204
NA
N
N
III


HB11
0.76923077
1
2
C1
F
18
626100
Y
H
IV


HB153
0.78571429
1
2
C1
F
27
1000000
Y
H
IV


HB28
0.8125
1
2
C1
M
34
172500
Y
NA
II


HB83
0.8125
1
2
C1
M
15
285
Y
S
II


HB156
0.85714286
1
2
C2
F
2
468000
Y
S
III


HB112
0.86666667
1
2
C1
M
36
725
Y
S
II


HB82
0.86666667
1
2
C1
M
120
179000
N
N
II


HB97
0.86666667
1
2
C1
F
42
700000
Y
H
IV


HB81
0.875
1
2
C1
M
22
322197
Y
H
III


HB103
0.9
1
2
C2
F
57
750000
Y
H
IV


HB114
0.9
1
2
C2
F
21
8783
Y
S
II


HB142
0.90909091
1
2
C2
F
48
605000
Y
H
III


HB148
0.93333333
2
3
C1
M
17
200730
Y
S
II


HB167
0.93333333
2
3
C2
M
34
1500000
Y
H
NA


HB73
0.9375
2
3
C2
F
24
667786
Y
H
III


HB131
1
2
3
C2
M
6
7511
Y
H
II


HB65
1
2
3
C2
M
6
1740
N
N
III


HB78
1
2
3
C1
M
126
376000
Y
S
II


HB72
1.07142857
2
3
C2
F
16
1412000
Y
S
III


HB48
1.07692308
2
3
C2
M
72
35558
Y
H
IV


HB102
1.09090909
2
3
C2
M
41
1331000
N
N
II


HB160
1.125
2
3
C2
M
45
342000
Y
H
II


HB172
1.125
2
3
C2
M
50
64170
Y
H
II


HB99
1.22222222
2
3
C2
M
72
277192
N
N
IV


HB130
1.25
2
3
C2
F
19
1980000
Y
H
II


HB98
1.25
2
3
C2
M
60
1285000
Y
H
III


HB136
1.3
2
3
C2
M
6
31828
Y
S
III


HB165
1.3
2
3
C2
M
13
18600
Y
S
II


HB1
1.36363636
2
3
C2
F
43
3000
Y
H
IV


HB93
1.36363636
2
3
C2
M
22
107000
Y
S
III


HB129
1.375
2
3
C2
M
96
14000
N
N
I


HB33
1.4
2
3
C2
M
12
765890
Y
H
IV


HB100
1.44444444
2
3
C2
M
48
576000
N
N
III


HB184
1.44444444
2
3
C2
M
41
912500
Y
H
IV


HB157
1.55555556
2
3
C2
M
7
356000
Y
H
NA


HB80
1.6
2
3
C2
M
180
37000
Y
H
III


HB86
1.66666667
2
3
C2
M
0.08
74000
N
N
III


HB8
1.75
2
3
C2
F
8
44610
Y
NA
II


HB147
2
2
3
C2
F
9
2355000
Y
S
II


HB39
2
2
3
C2
F
11
862067
Y
S
III




























Main Epi-














thelial
beta-



tumor
Distant
Vascular
Multi-
Histol-
com-
catenin
Follow-up

Surgery

Follow-up



ID
Metastasis
invasion
focality
ogy
ponent
status
(months)
Outcome
Type
speOS
(years)
























HB126
N
N
S
Mx
F
mut
18
A
R
0
1.5



HB145
N
N
S
Mx
F
mut
17
A
R
0
1.416666667



HB150
N
N
S
Mx
F
mut
14
A
R
0
1.166666667



HB175
N
N
M
Mx
F
NA
6
A
R
0
0.5



HB20
N
N
M
Mx
F
mut
7
A
R
0
0.583333333



HB49
N
N
S
Ep
F
mut
42
A
R
0
3.5



HB54
N
N
S
Ep
F
mut
6
D
R
0
0.5



HB70
N
N
S
Ep
PF
mut
49
A
R
0
4.083333333



HB77
N
N
S
Ep
PF
mut
53
R
R
0
4.416666667



HB89
N
N
S
Ep
F
mut
37
A
R
0
3.083333333



HB95
N
N
S
Ep
F
mut
33
A
R
0
2.75



HB118
Y
Y
M
Mx
F
mut
32
A
LT
0
2.666666667



HB132
N
N
S
Mx
F
mut
121
A
R
0
10.08333333



HB121
N
N
M
Mx
F
mut
18
A
R
0
1.5



HB140
N
N
S
Mx
F
mut
22
A
R
0
1.833333333



HB162
N
N
S
Mx
F
mut
13
A
R
0
1.083333333



HB171
N
N
S
Ep
F
mut
9
A
R
0
0.75



HB173
N
N
S
Ep
F
NA
11
A
R
0
0.916666667



HB59
N
N
S
Ep
PF
mut
72
A
R
0
6



HB6
N
Y
S
Ep
F
mut
48
A
R
0
4



HB74F
N
Y
S
Ep
F
mut
35
A
R
0
2.916666667



HB96
N
Y
M
Ep
F
mut
23
R
LT
0
1.916666667



HB60
N
Y
S
Ep
F
wt
63
A
R
0
5.25



HB7
N
Y
S
Mx
F
mut
46
A
R
0
3.833333333



HB101
N
N
S
Ep
F
mut
20
A
R
0
1.666666667



HB106
N
N
S
Mx
F
mut
25
A
R
0
2.083333333



HB90
N
N
S
Ep
F
mut
35
A
R
0
2.916666667



HB62
N
N
S
Mx
F
mut
69
A
R
0
5.75



HB107
Y
Y
M
Ep
F
mut
25
A
LT
0
2.083333333



HB170
Y
Y
M
Ep
F
wt (FAP)
15
A
R
0
1.25



HB5
Y
Y
M
Ep
F
mut
24
DOD
R
1
2



HB125
Y
N
M
Mx
F
mut
17
A
LT
0
1.416666667



HB75
N
Y
S
Mx
F
mut
41
A
R
0
3.416666667



HB9
N
N
S
Ep
PF
mut
91
A
R
0
7.583333333



HB94
N
N
S
Ep
PF
wt
29
A
R
0
2.416666667



HB61
Y
Y
M
Mx
F
mut
5
DOD
R
1
0.416666667



HB69
N
N
S
Ep
PF
wt
55
A
R
0
4.583333333



HB79
N
N
M
Ep
M
mut
39
A
LT
0
3.25



HB3
N
N
S
Ep
F
wt
55
A
R
0
4.583333333



HB66
N
N
S
Ep
F
mut
68
A
R
0
5.666666667



HB68
N
N
S
Mx
E
mut
52
A
R
0
4.333333333



HB146
N
NA
S
NA
NA
NA
1
D
R
0
0.083333333



HB155
N
N
S
Mx
CF
mut
8
A
R
0
0.666666667



HB63
N
Y
M
Mx
F
mut
96
A
R
0
8



HB11
Y
Y
M
Mx
F
mut
21
DOD
R
1
1.75



HB153
Y
N
M
Mx
CF
mut
8
A
LT
0
0.666666667



HB28
N
N
S
Ep
F
wt
120
A
R
0
10



HB83
N
N
S
Ep
PF
mut
53
A
R
0
4.416666667



HB156
N
N
NA
Ep
F
NA
6
A
R
0
0.5



HB112
N
N
S
Ep
F
wt
32
A
R
0
2.666666667



HB82
N
N
S
Ep
F
mut
63
A
R
0
5.25



HB97
N
Y
M
Ep
F
mut
30
A
R
0
2.5



HB81
Y
Y
M
Ep
F
mut
36
A
R
0
3



HB103
Y
Y
M
Ep
M
mut
9
DOD
M
1
0.75



HB114
N
N
S
Mx
E
mut
23
A
P
0
1.916666667



HB142
Y
Y
S
Ep
NA
mut
16
A
R
0
1.333333333



HB148
N
N
S
Mx
F
mut
11
A
R
0
0.916666667



HB167
Y
Y
M
Ep
F
mut
2
A
R
0
0.166666667



HB73
Y
Y
S
Ep
E
mut
16
DOD
R
1
1.333333333



HB131
Y
N
S
Ep
E
wt
1
DOD
R
1
0.083333333



HB65
N
N
M
Mx
E
wt
2
DOD
R
1
0.166666667



HB78
N
Y
M
Ep
CF
wt
32
A
R
0
2.666666667



HB72
Y
Y
M
Mx
E
mut
9.5
DOD
R
1
0.791666667



HB48
N
Y
M
Ep
CF
mut
9
DOD
R
1
0.75



HB102
N
N
S
Ep
CF
mut
4
D
B
0
0.333333333



HB160
Y
Y
S
Mx
E
NA
14
R
R
0
1.166666667



HB172
Y
Y
M
Mx
F/E
NA
10
A
R
0
0.833333333



HB99
Y
Y
M
Ep
E
mut
7
DOD
B
1
0.583333333



HB130
Y
N
S
Mx
NA
mut
62
A
R
0
5.166666667



HB98
Y
Y
S
Ep
M
wt (FAP)
30
A
M
0
2.5



HB136
N
N
S
Mx
F
wt
34
A
R
0
2.833333333



HB165
N
N
M
Mx
F/E
mut
4
A
R
0
0.333333333



HB1
Y
Y
M
Ep
E
wt (FAP)
12
DOD
R
1
1



HB93
N
Y
M
Mx
E
mut
33
A
LT
0
2.75



HB129
N
N
S
Mx
E
wt (FAP)
54
DOD
R
1
4.5



HB33
N
Y
M
Ep
CF
wt(AX1N1)
3.5
DOD
R
1
0.291666667



HB100
N
N
S
Ep
F
mut
20
A
B
0
1.666666667



HB184
Y
Y
M
Ep
E
NA
14
DOD
LT
1
1.166666667



HB157
Y
N
M
Ep
CF
mut
5
R
LT
0
0.416666667



HB80
Y
Y
S
Ep
CF
mut
14
DOD
R
1
1.166666667



HB86
N
Y
S
Ep
E
mut
57
A
R
0
4.75



HB8
N
Y
S
Ep
E
mut
135
A
R
0
11.25



HB147
N
N
S
Mx
F
NA
12
A
R
0
1



HB39
N
Y
S
Mx
NA
mut
66
A
R
0
5.5


























TABLE C










Gene











Name
AFP
ALDH2
APCS
AP0C4
AQP9
BUB1
C1S
CYP2E1
DLG7



















HC161
2.079447
−5.920384
−6.086912
−7.366206
−7.320175
4.176845
−6.502865
9.12672475
5.322878


HC162
4.056751
−3.64102
−4.586098
−5.663246
−4.233021
3.559124
−4.64283
−4.136919
5.950173


HC163
3.323238
−6.086663
−6.399079
−4.052853
−6.010302
4.772507
−6.776158
−8.515956
5.551408


HC164
3.075226
−6.146711
−7.241796
−3.371322
−5.446966
3.634476
−7.462807
−5.829384
3.98399


HC165
2.685177
−7.0470725
−6.294538
−7.242275
−6.94561
4.029514
−5.926596
−3.033642
5.723743


HC168
1.501031
−6.016314
−6.696324
−5.130347
−5.64774
3.305894
−6.883263
−4.411302
4.362859


HC169
2.880925
−6.024682
−6.87168
−4.19185
−6.058572
4.09117
−6.767215
−8.63753
4.614352


HC170
2.3753035
−6.6226955
−8.3702955
−5.4072375
−5.6954625
5.5639145
−8.0538815
−9.7948605
6.6275145


HC171
3.001804
−2.573977
−4.213123
−4.040859
−4.992701
3.583809
−5.226561
1.25382
3.874142


HC172
1.164528
−5.314302
−6.094852
−4.127298
−3.890072
3.991173
−6.240002
2.279678
5.651484


HC173
4.694127
−6.373823
−5.51865
−6.056863
−6.314031
4.30288
−4.863168
−8.649852
5.564261


HC176
4.066485
−5.552505
−5.444218
−5.551191
−5.815727
6.073568
−5.850428
−9.402043
6.051409


HC177
2.692613
−5.43842
−3.091896
−4.656336
−5.907612
3.452047
−6.412596
−10.50172
4.083836


HC178
−0.554213
−5.646708
−7.296414
−4.588115
−5.579087
3.125179
−6.556397
−6.591304
4.755443


HC179
1.910595
−4.139932
−8.136252
−6.036987
−2.847761
3.895205
−4.943672
−5.283326
5.054346


HC180
3.212685
−5.831134
−7.519348
−5.962761
−6.611712
1.5179
−6.130592
−9.203789
2.22658


HC181
6.030393
−4.04397
−2.03808
−0.956533
−2.850753
5.430957
−4.712002
−2.555649
5.031845


HC182
3.376941
−7.072651
−7.74873
−5.2003
−5.445893
6.665657
−7.899793
−10.089271
7.487442


HC183
3.149578
−4.684626
−7.045155
−3.800078
−7.042931
2.40337
−6.412624
−9.657513
3.396236


HC184
−0.093476
5.985909
−7.203484
5.482853
−6.208594
1.558788
−6.347367
−9.658434
2.407985


HC185
1.405595
−4.748444
−5.89589
−3.780913
−2.802368
4.37289
−5.800822
−5.410746
4.6459


HC186
1.666457
−5.52819
−7.953401
−3.287374
−3.805233
1.040678
−7.309734
−6.699831
2.197157


HC187
3.652111
−4.151991
−7.459358
−6.247812
−5.346647
4.211928
−6.33068
−8.629261
4.520672


HC188
0.355562
−5.261937
−7.83848
−4.759525
−4.839348
5.111208
−7.787661
−4.575966
5.635841


HC189
1.239891
−4.501697
−8.737075
−6.152778
−6.402122
5.0291015
−6.951675
−5.450079
4.419359


HC190
3.306642
−4.365515
−7.399538
−4.721411
−6.178224
3.016906
−4.970499
−5.850237
9.264351



















Gene










Name
DUSP9
E2F5
GHR
HPD
IGSF1
NLE
RPL10A




















HC161
3.702615
1.025512
−0.817005
−7.653863
14.149408
5.1985405
−5.81852



HC162
1.738977
1.432598
−0.231753
−6.700146
14.781699
1.231146
−5.9665735



HC163
4.00436
1.072797
−2.746621
−6.213082
8.2477055
2.203781
−5.49725



HC164
4.25604
2.567639
−3.606813
−6.079645
12.649441
1.946926
−5.171041



HC165
1.788757
1.157215
−1.197022
−7.969042
14.270796
2.620134
−6.219366



HC168
5.625335
2.2963
−1.169362
−7.52548
8.041574
2.337152
−5.42627



HC169
3.838008
1.60884
−2.921191
−6.51064
8.136143
2.099644
−5.731897



HC170
1.8626715
1.6955475
−3.9034625
−7.4271305
7.756398
2.6917235
−5.8132855



HC171
5.349357
2.074272
−1.437519
−5.297939
6.325863
3.057537
−3.95361



HC172
5.592005
1.291773
−0.040049
−6.989866
6.998259
3.186024
−3.946432



HC173
4.718896
1.367846
−2.3934
−7.781412
9.1259525
1.82226
−4.957916



HC176
2.248373
2.709599
−3.2392
−7.594156
7.5288985
1.817325
−5.042318



HC177
−0.297108
2.149313
−2.166834
−7.847734
5.8240705
1.530536
−5.640103



HC178
4.943904
1.038474
−1.620902
−5.659262
5.416822
1.855914
−4.954215



HC179
1.464274
1.372578
−0.386778
−6.31274
7.244471
1.887378
−5.218281



HC180
0.161194
−0.215954
−0.371454
−6.978048
5.185486
1.004282
−6.187635



HC181
4.322323
2.990459
2.18165
−0.651095
4.292234
4.670446
−2.978533



HC182
2.395117
2.329727
−4.420263
−7.357922
7.932783
2.869667
−5.574881



HC183
3.7002215
−0.85541
0.078707
−7.143723
11.999761
0.63414
−6.105039



HC184
2.266351
6.244093
0.670045
−6.27671
6.935964
1.564672
−6.568913



HC185
1.811225
2.225761
−1.246884
−7.344763
10.1413645
1.39443
−5.015711



HC186
−2.717975
1.183123
−2.657936
−7.680597
8.921477
1.289946
−6.631908



HC187
−0.066629
−2.0378
1.078709
−8.251018
7.478678
1.655093
−5.763416



HC188
1.839584
0.638515
−1.989428
−6.736329
12.8628775
2.27923
−4.743699



HC189
6.509026
−0.7698
−2.238756
−8.600128
11.305903
−0.437812
−7.061492



HC190
0.70722
4.181534
−0.773062
−4.881306

2.422048
−5.53509

















TABLE D








Table of normalized qPCR data (deltaCt values) of 88 HCCs analyzed by the Taqman method























Gene










name
AFP
ALDH2
AP0C4
APCS
AQP9
BUB1
C1S
CYP2E1


















HC 001
2.212911
−6.2372335
−0.614689
−7.0721355
−6.047695
3.841505
−8.163492
−10.3093235


HC 003
3.865709
−6.230074
−0.95786
−7.52919
−6.7334475
0.147459
−8.7963405
−10.428074


HC 004
7.6758115
−2.186358
1.608247
−5.845683
−3.759528
4.221132
−5.8997645
−7.1147515


HC 006
7.9469815
−5.4231035
−0.9614255
−7.3704745
−7.006052
0.5252045
−8.162856
−10.1334265


HC 007
−5.311541
−4.0446765
3.550537
−5.1967915
−6.747103
0.299039
−4.062593
−11.024027


HC 008
−2.0890815
−3.9297005
0.6776965
−6.567126
−3.1082155
1.214781
−7.2991535
−7.791007S


HC 009
7.0483095
−3.0017225
9.6721075
0.017488
−3.7536735
−2.980029
−4.830331
−0.5825245


HC 010
−2.3869635
−0.95212
0
1.0272875
−1.3400495
1.864677
−2.639902
−3.604805


HC 011
−0.6488335
−5.958108
−1.076151
−7.7638255
−6.122144
2.362454
−8.319293
−9.575619


HC 012
6.538312
−4.6271565
1.221393
−6.942673
−4.1878425
3.293346
−6.850023
−7.284587


HC 014
2.987769
−5.194577
−1.3542145
−6.5396565
−6.8623455
1.363697
−6.8939375
−10.7465595


HC 015
−6.14089
−4.5178635
5.156026
−3.380102
−2.373344
−0.8830545
−7.1343975
−4.9390935


HC 017
−7.1950405
−2.6522585
2.395651
−4.5167035
−2.8711295
−1.0884485
−6.035123
−6.037085


HC 018
6.856588
−1.840894
3.84764
−4.916924
−3.6093495
0.063545
−4.263272
−5.811062


HC 020
0.65281
−6.287083
−3.2094885
−8.2117635
−7.354605
1.4635025
−8.471663
−10.2536915


HC 021
4.3070475
−2.175112
6.2591235
−5.9159775
−1.1452535
−0.0802935
−5.7190985
−1.2878015


HC 022
4.418018
−5.331214
−0.5455545
−6.6835035
−5.7992305
2.173361
−7.2514145
−8.0876755


HC23
5.538438
−5.853486
−0.5708905
−6.9009145
−6.651868
2.5475915
−8.2212235
−9.047509


HC 025
3.90298
−6.162477
−1.834891
−8.798759
−8.758959
2.5679685
−8.5606875
−10.814935


HC 026
5.69175
−5.0135775
−0.2581675
−7.2072275
−3.8645965
−0.545363
−7.2351705
−0.671071


HC 027
0.626755
−5.6309605
−1.53158
−7.2809855
−5.4736555
0.8889165
−8.172076
−8.6350095


HC 028
0
−1.913778
6.0251725
−1.0475505
−0.9613895
5.7426525
−4.910584
−3.6858305


HC 030
−6.4370325
−3.8476295
−0.2797975
−7.1142435
−5.0250435
0.190936
−7.5279395
−7.5682115


HC 032
−0.0037145
−6.802666
−2.574347
−7.500133
−7.530391
5.1317805
−7.854502
−9.4408715


HC 034
6.6945705
−5.11617
−0.5860455
−7.134934
−6.9427395
1.2674215
−7.719763
−8.545814


HC 037
1.3519745
−5.808058
0.0768065
−6.755895
−6.3416265
2.4955985
−6.921051
−10.1686795


HC 038
−4.053435
−4.596143
0.129322
−5.045701
−6.0302545
−0.321483
−6.101331
−8.1123675


HC 041
2.7156435
−6.3503265
−2.281983
−5.612517
−7.8444565
0.587016
−6.88808
−9.5090495


HC 042
5.216493
−4.4086495
0.627239
−4.1054755
−6.063786
2.224818
−6.3060565
−9.1411555


HC 043
1.7983435
−5.457548
0.7055185
−7.607914
−4.7175855
2.8634735
−7.9862115
−8.760714


HC 052
−10.3337105
−2.1920375
8.124407
−5.9818015
0.4848805
1.2986035
−5.6337865
−1.7693015


HC 058
−1.891958
−2.1172735
11.8524
4.1106695
2.817265
−1.9395175
−3.691331
4.3317445


HC 060
−7.624821
−3.6860195
0.545509
−8.100997
−6.8503395
0.576028
−8.167253
−9.1875325


HC 064
−5.0266755
−4.992107
−0.7860345
−7.4148835
−7.0526325
1.367463
−7.1364365
−9.682147


HC 066
−3.156328
−3.8408415
0.6773785
−8.2106815
−6.2767975
1.1272665
−8.026875
−8.601088


HC101
6.873135
−4.339036
0.5787185
−6.288568
−4.6233735
−0.081457
−7.321092
−5.806032


HC102
4.119697
−2.476355
5.453696
2.3952165
−0.0196725
0.5553155
−5.939374
2.8566735


HC103
−1.6193685
−3.889904
0.54698
−6.014572
−7.151639
2.086008
−5.965432
−8.266311


HC104
−5.5094265
−4.936239
0.5059805
−5.624234
−0.501258
1.311194
−6.716137
−9.0888685


HC105
−2.3444245
−4.239726
3.577778
−7.703333
−4.2748785
−0.945674
−7.774455
−5.698899


HC106
3.42054
−6.1642895
0.7836775
−7.8462545
−5.85931
4.8909655
−8.060072
−9.9949555


HC107
4.136209
−6.7443095
−4.4534435
−9.2080655
−8.8878655
1.7415115
−9.2061165
−9.3234825


HC108
4.500336
−3.6076385
2.478085
−7.275462
−4.4353395
0.3807995
−7.1031155
−3.889942


HC109
4.833024
−5.8617665
−0.729565
−6.222909
−6.4504115
2.2918285
−7.406001
−8.7101925


HC110
3.5240185
−3.6707715
0.256479
−5.043319
−4.5999895
1.449943
−6.9163195
−7.145766


HC111
1.883473
−3.8304065
1.130067
−5.976754
−4.1657805
−0.621548
−6.278164
−4.46942


HC112
2.8803905
−4.8726745
0.7777655
−6.764675
−5.2735435
−0.3135015
−7.455794
−2.5741475


HC113
−1.208649
−4.407016
2.366969
−5.197177
−2.681192
3.4825665
−6.338901
−6.443846


HC114
5.4433695
−4.7113965
0.833543
−6.723142
−4.445291
1.7431855
−7.866014
−7.3429245


HC119
−1.0580855
−6.159706
−1.894453
−9.375177
−7.6266135
0.797564
−9.1461175
−7.095824


HC120
4.0065425
−4.257398
3.5241745
−5.6838965
−6.8239115
0.0740105
−8.5708615
−7.6044515


HC121
4.254961
−4.556431
2.167313
−6.2688205
−4.38702
2.4486685
−8.118416
−7.765037


HC122
2.3763095
−6.2844515
−1.279577
−6.9942545
−6.8198535
6.0183915
−7.7653135
−9.450349


HC123
−0.821555
−4.220769
0.68167
−5.778659
−6.410177
1.190323
−5.383781
−8.528543


HC124
−3.9525335
−4.027289
0.0499065
−5.391271
−4.463488
1.592563
−5.151686
−9.520436


HC125
4.806564
−4.5451465
−2.6326775
−6.5321595
−8.370224
−1.1627945
−8.4244055
−9.426232


HC126
5.899437
−5.02839
−0.407895
−5.2838365
−3.6163545
2.6943025
−7.1365955
−5.226091


HC127
0.0390765
−2.41699
−0.8680995
−4.846116
−1.8613935
2.048769
−6.3641695
−6.1813065


HC128
−5.8636305
−5.085525
0.626498
−5.087517
−4.3184915
1.3297375
−6.828468
−7.4344035


HC129
3.430757
−4.6298475
1.863955
−4.8448705
−2.870839
2.3688215
−7.302922
−2.692798


HC131
1.491189
−5.425994
−2.4702
−8.6617295
−7.4772145
0.727709
−7.525072
−8.98645


HC132
−5.4265205
−3.105643
6.9974515
3.2748865
−3.9244375
−0.2895395
−4.390082
−7.0455735


HC133
5.1621395
−4.2462915
−0.63156
−7.145861
−6.05182
4.9277675
−7.3188145
−8.1908895


HC134
−2.8738695
−4.061101
0.1134065
−7.5103485
−5.550642
−1.7425995
−8.4609335
−7.859701


HC135
0.909107
−2.7442165
0.7630605
−0.959726
−4.0595615
1.2018365
−4.667223
−4.30592


HC136
0.4105125
−6.0408575
−0.7390785
−7.150737
−5.996196
4.288554
−8.243333
−9.042865


HC137
−4.378388
−3.2913795
3.209294
−4.421328
−0.5225755
4.2185175
−5.647363
−5.532515


HC138
2.4762965
−4.8248625
1.154563
−4.883388
−3.440722
3.408251
−6.459976
−7.2458685


HC139
2.7547595
−2.9782295
3.0252085
−5.3858735
−5.0157665
0.9503045
−6.0281485
−1.1920485


HC140
6.3489955
−4.644452
−1.006979
−2.1507335
−5.3387635
4.075603
−6.7373815
6.646618


HC141
2.4010865
−4.8883675
0.787009
−4.7365085
−4.1224775
4.2728925
−6.8664705
−2.6765195


HC142
4.5984525
−3.7946485
2.8271835
−4.9243665
−3.1411815
4.0713025
−6.3482925
2.654871


HC143
−4.0727165
−2.59764
1.855993
−4.8795135
−2.222047
1.6908025
−4.948264
−3.1057735


HC144
4.7344185
−4.3542505
−1.002913
−0.432856
−5.16696
2.510931
−5.3365195
−4.456082


HC145
8.5175565
−3.375805
0.8672075
−5.0765195
−4.091142
3.9700095
−6.960951
0.8009


HC146
5.741507
−3.5738745
1.2439275
−5.1950135
−3.4305425
2.9843625
−5.666896
−0.913546


HC147
6.0474775
−3.0470955
0.2246755
−5.6213855
−5.257189
2.7534355
−5.349428
−6.933909


HC148
−1.306432
−4.0108565
0.267747
−6.3544915
−3.1846315
1.1995135
−6.2066555
−4.1428355


HC149
−3.9190605
−3.3456535
2.735403
−1.9099995
−1.1810265
2.704253
−5.707004
−5.9300895


HC150
6.1556695
−2.9923905
−1.9485835
−5.821769
−6.3127705
2.452404
−4.984573
−7.3184395


HC151
5.5488065
−4.234966
1.372415
−5.8812085
−4.0297925
3.4239945
−7.2861515
−2.304461


HC152
4.917902
−3.97386
−4.005999
−6.5072455
−7.124415
2.5576145
−5.752235
−9.98327


HC153
5.6708455
−5.004032
−3.204075
−3.8195495
−6.2020215
1.9670395
−5.979251
−7.7421455


HC154
6.699114
−2.0392575
9.6136985
0.885791
−0.68511
1.755108
−0.7395055
2.544628


HC155
6.238831
−3.802053
2.0022335
−6.3105565
−2.974712
4.2276825
−7.058571
−4.1514335


HC156
−1.582839
−3.5688085
0.917505
−3.9333845
−4.163765
1.0763025
−4.6064345
−8.4802835


HC157
3.657864
−4.2315665
2.513598
−7.2096625
−4.573216
−0.284071
−5.856564
−7.9837885


HC159
3.4650565
−2.6801805
2.2596385
−4.0834345
−4.42904
3.44645
−5.923485
−7.778452


















Gene










name
AFP
ALDH2
AP0C4
APCS
AQP9
BUB1
C1S
CYP2E1


















HC 001
5.30317
11.616567
−0.05328
−2.655512
−9.449416
6.46034
1.159417
−6.6225235


HC 003
2.057513
8.8462855
1.909804
−2.069524
−8.549803
7.249974
1.5801355
−6.0562915


HC 004
4.4226465
9.4268185
1.7432195
2.0012965
−9.415253
0
3.1459935
−4.4121905


HC 006
1.6282005
10.22051
−0.024339
−1.887805
−8.5958965
7.1580385
−0.6940375
−6.8637555


HC 007
1.169221
6.6521625
0.2833465
1.7428205
−6.183977
3.192514
0.3919565
−7.1381125


HC 008
2.80866
9.6946695
0.0193165
−2.342442
−5.329776
2.806768
1.579419
−6.2574845


HC 009
−1.3733475
9.5262655
−0.711082
2.3242195
0.011478
4.026769
0.80375
−6.3016635


HC 010
0
0
1.344368
0.4900285
−2.932809
0
0
−9.1966395


HC 011
2.8432205
0
0.736822
−4.757848
−9.029214
7.6390015
1.9328755
−7.379063


HC 012
4.7199665
0
2.4002515
−2.2402875
−9.656029
7.466951
1.64183
−5.178571


HC 014
3.3543285
7.7629895
1.5332515
−1.09511
−9.5837645
8.5836025
1.47219
−5.831244


HC 015
0.1414205
4.4342765
−1.399564
−0.2426
−4.473096
−0.0722075
0.321593
−6.8777395


HC 017
−0.666284
3.163581
−1.206766
2.353691
−0.6808655
6.0490105
0.386649
−7.068098


HC 018
1.512286
8.7756845
2.426129
2.9035
−5.7101575
2.4248235
1.3815525
−5.9464565


HC 020
2.1165725
9.6208445
1.1944835
−4.5756335
−10.6864405
0
1.118745
−7.542193


HC 021
0.322455
7.8162765
0.0686475
−0.71981
−4.0108195
2.954814
1.618369
−6.309556


HC 022
3.3904095
10.82729
10.7133385
−2.416651
−9.8859985
5.6986975
1.9449755
−7.194012


HC23
3.848364
0
1.4330655
−3.7226655
−9.583194
7.200325
1.823275
−5.9526365


HC 025
3.34202
7.1111525
−0.049846
−1.9012935
−9.1845675
0
1.770127
−7.4507165


HC 026
0.9710395
8.5287915
1.1845665
−1.964045
−7.6403735
5.4960635
1.851733
−5.9670715


HC 027
2.3158215
10.241011
0.4045835
−2.623084
−9.597772
5.588995
1.851285
−7.6623025


HC 028
0
0
4.334386
1.9788575
−3.3142495
0
2.4559905
−5.521873


HC 030
0.189092
9.0027
−1.0623035
−2.635437
−7.537
2.651022
1.2674865
−7.5046195


HC 032
5.7080765
9.73163
0.054818
−2.0027475
−9.0015185
0
1.208576
−8.8437815


HC 034
2.339621
9.9728495
1.4281575
−1.563203
−8.3685675
10.112616
1.934745
−6.594006


HC 037
2.6534895
0
1.2212655
−2.9415775
−10.367265
7.5570255
1.9881245
−6.901637


HC 038
1.4386515
5.2298755
0.037887
−0.2025015
−7.547286
0.680358
2.1250395
−5.1574215


HC 041
1.840185
8.727439
−0.466649
−1.428749
−8.0015745
7.243446
0.15624
−7.7043325


HC 042
3.2531575
0
0.3673235
1.2545195
−8.2669835
2.899766
0.9401045
−5.577659


HC 043
4.2390495
10.525647
0.894345
−3.2916395
−8.997825
5.5544715
1.8422595
−5.480403


HC 052
2.599359
3.8059605
−0.4419525
1.843696
−2.481945
−2.254168
1.9474305
−5.6154705


HC 058
−0.1957495
3.656912
−0.804087
3.7242975
−1.8257985
−1.3471695
1.209522
−6.0601515


HC 060
2.2644225
6.618755
0.432422
1.4079225
−8.4643875
0.7884805
1.9133155
−5.7041285


HC 064
2.386875
7.3184655
0.2876185
−0.349645
−8.6027575
3.3382005
1.817699
−6.4617635


HC 066
2.7680135
11.5673955
0.968982
1.2501855
−8.5231325
9.185554
1.962008
−5.415169


HC101
1.3084655
8.828389
1.871516
−0.1466275
−5.7252795
4.1394545
1.4546305
−6.144011


HC102
2.1385165
8.6628475
−0.830934
−0.947389
−0.568809
2.708733
1.1534675
−5.283399


HC103
2.957914
12.521336
1.8003215
−0.636723
−6.717282
9.802921
2.594702
−4.423835


HC104
1.821739
5.396553
2.305498
−1.6860905
−8.46781
−0.1438735
1.610158
−6.21159


HC105
0.814912
5.4214725
−2.0730715
−0.682142
−2.288109
1.422332
0.471391
−6.315756


HC106
6.2678815
11.174152
2.208171
−5.342392
−9.4440475
7.401009
1.968983
−5.769397


HC107
1.357756
6.6136855
−2.78876
−2.935929
−10.460972
0
0.000835
−8.6686655


HC105
2.2445545
8.0946735
−0.0923905
−1.6363755
−2.9674235
7.967992
0.932052
−5.818028


HC109
3.222524
10.4709205
1.9924345
−2.9233285
−7.8859205
10.0122565
2.6102395
−5.541229


HC110
2.333076
11.616244
2.512512
−1.0803015
−8.1908235
8.1469415
2.3529485
−5.245476


HC111
0.769283
9.137462
−1.045678
−1.1576425
−7.245347
1.86965
1.012752
−5.568205


HC112
0.9196845
10.105965
−0.0373705
−2.5391085
−7.714358
3.4428695
1.119237
−6.1905075


HC113
4.5602875
7.8299455
2.82243
−2.16232
−6.685692
2.045068
2.156348
−5.8884625


HC114
3.1500875
11.804112
0.0450475
−2.5053965
−6.835254
5.1813245
1.3170345
−5.795905


HC119
1.712686
9.106547
0.0248045
−3.7649595
−9.220498
5.39017
0.400823
−7.954231


HC120
1.9563135
5.8119685
−1.229768
−3.196589
−8.5127155
9.404196
1.1096815
−6.4517175


HC121
2.852561
9.706684
0.910943
−2.2774645
−7.480725
5.980435
1.758163
−6.4042545


HC122
7.228946
9.9054825
3.5033365
−2.400201
−8.7301975
8.6480295
2.2430545
−5.199782


HC123
2.929576
11.584458
0.646839
1.810364
−4.7774665
5.1400615
1.5951645
−4.7323885


HC124
2.03781
8.81055
−0.574165
−2.2369305
−7.832169
1.4450915
0.1499775
−6.691521


HC125
−0.3286545
9.3740615
0.028878
−0.697866
−5.7813
10.2234745
0.405397
−7.1196575


HC128
3.944339
8.7174575
3.271927
−1.824385
−1.865621
7.659377
2.033278
−5.389272


HC127
2.96212
8.672372
2.162602
−0.129431
−3.4481965
3.1503205
2.205965
−4.3385115


HC128
2.6299155
8.499355
4.393094
−1.9716885
−5.7052855
2.72995
1.949352
−6.6181545


HC129
3.6405185
7.0627455
0.470421
−2.332961
−5.502918
5.692623
1.683808
−4.8697295


HC131
1.461713
8.415907
−0.154573
−4.009655
−8.960383
7.5832005
1.5313675
−6.775249


HC132
1.5572645
3.3843145
−1.9018925
−1.7710325
−2.3653865
1.947055
−0.2035885
−6.7796075


HC133
5.5447335
8.022457
2.6341825
−2.2298335
−6.1281315
0
1.4173895
−5.762015


HC134
−0.8148735
4.96739
−3.1030595
−1.3138565
−7.231144
0.3848995
−0.794433
−7.7140665


HC135
2.250305
5.794605
−0.986165
0.6955465
−6.7262275
4.394354
0.9780515
−6.689595


HC136
5.5267715
10.9307725
2.4040865
−4.013948
−8.223611
7.4962365
2.426321
−5.5069335


HC137
5.2105355
4.767228
5.62451
−1.6355645
−5.8875425
1.0556075
3.7311615
−5.2271275


HC 138
5.028429
5.576937
4.1601375
−1.738341
−6.019837
7.169314
4.19882
−4.2322595


HC 139
2.940447
4.3133685
0.685194
1.632571
−4.6240035
3.333358
1.7913325
−6.6866335


HC 140
5.1767035
10.874029
2.488357
−3.1717235
−7.5439415
9.276635
5.0732625
−4.266519


HC 141
6.1148255
7.979559
2.66802
−1.687093
−7.2596615
#DIV/0!
3.5973445
−4.952551


HC 142
5.8031125
8.2104255
2.0983905
−1.5934495
−5.8074755
9.442329
3.4164995
4.6520795


HC 143
3.470906
3.981805
1.474377
0.695168
−2.049901
3.754627
3.058019
−4.7443975


HC 144
3.844786
10.7187705
3.540563
−1.6857605
−6.869217
11.9441575
4.417722
−4.817306


HC 145
5.482263
9.313039
2.112409
−1.525041
−6.669204
10.0458615
3.0082705
−5.7677005


HC 148
5.1824885
7.611916
2.8802325
−1.791636
−6.9831945
5.450716
3.884913
−4.427413


HC 147
4.5366875
9.358894
3.2373475
−2.0156545
−6.053345
8.7065355
3.732017
−4.317148


HC 148
2.490156
5.4985645
8.523611
−0.773246
−3.7206575
5.663583
3.295068
−6.0532135


HC 149
3.4454215
6.8563245
2.4724295
−0.9357605
−7.337568
−0.063395
4.267075
−5.7767065


HC 150
3.585447
7.980274
3.118546
0.5916635
−5.762837
9.1651835
2.811495
−5.7495535


HC 151
4.613043
8.9062765
2.2090065
−2.8000785
−7.251033
9.44137
3.5959505
−4.6972005


HC 152
4.17552
10.736246
4.56538
−1.578246
−8.106859
12.118351
2.6658355
−6.944767


HC 153
3.133394
7.298329
3.85894
−0.616143
−7.947464
11.674272
2.670245
−5.0796695


HC 154
3.2541115
3.139705
−0.3936805
−1.070278
−4.611328
1.5925535
2.2396475
−6.2090535


HC 155
5.7341595
6.4585135
2.4375015
−0.254649
−7.297162
10.0981895
3.3878795
−5.37231


HC 156
2.1302465
4.4056075
1.070339
0.42868
−6.890963
2.0124875
2.225275
−7.037827


HC 157
1.3778545
2.0950385
−0.56173
−0.8411435
−8.474893
7.2842685
1.6720135
−6.6310375


HC 159
5.727853
8.8523415
2.7886015
−1.0442865
−7.268645
8.8204775
2.861685
−5.4777465


























TABLE G











Date of



follow-

Date of 1st
secondary
date of
tumor


HC 000 tumor
surgery or
(PH) or
Date of
Date of
up
recur-
recurrence or
OLT after
secondary
grade


identification
transplantation
(OLT)
last visit
death
(years)
rence
metastasis
hepatectomy
OLT
Edmondson




















HC 001
12/12/1996
PH

07/01/1997
0.07
N



3


HC 003
21/02/1997
PH

20/06/2000
3.33
Y
 4/11/1998
N

2


HC 004
28/02/1997
PH
20/08/2008

11.48
N



2


HC 006
07/10/1996
PH

06/01/1998
1.25
N
28/11/1997
N

2


HC 007
02/07/1996
PH

31/12/1997
1.50
Y
 4/11/1997
N

2-3


HC 008
05/06/1996
PH

24/01/2005
8.48
N



3


HC 009
28/08/1996
PH

05/09/1996
0.02
N



3-4


HC 010
10/10/1996
PH

20/09/1997
0.95
N



4


HC 011
10/10/1996
OLT
14/12/2008

12.20
N



2


HC 012
24/10/1995
OLT

14/11/1995
0.05
N



2


HC 014
10/06/1995
OLT

27/07/1995
1.00
N



3-4


HC 015
21/07/1995
PH

10/10/1996
1.22
Y
10/10/1996
N

3


HC 017
05/05/1997
PH
16/04/2008

10.96
N



2


HC 018
07/05/1997
PH

28/09/1997
0.39
NA



3


HC 020
13/05/1993
OLT
20/10/2008

15.40
N



2


HC 021
15/01/1992
PH

28/09/1992
0.70
Y
15/06/1992
N

NA


HC 022
15/03/1997
OLT
02/09/2008

11.50
N



2


HC 023
20/07/1995
PH
20/06/2007

11.93
N



2


HC 025
05/10/1992
PH

13/08/2008
15.87
N



2


HC 026
04/06/1993
OLT

18/04/1994
0.83
NA



2


HC 027
20/01/1993
OLT

15/02/1993
0.10
N



2


HC 028
16/02/1996
OLT

13/03/1996
0.10
N



3


HC 030
10/04/1996
PH

07/09/2008
12.40
Y
15/10/1996
Y
17/12/1993
3


HC 032
17/02/1993
PH

17/10/1993
0.66
N



2


HC 034
10/03/1993
PH
05/11/2008

15.70
Y
15/11/1995
Y
20/06/1996
2


HC 037
08/06/1997
OLT

13/08/1997
0.20
N



3


HC 038
16/07/1997
PH

28/08/1998
1.12
Y
 1/01/1998
N

NA


HC 041
24/11/1997
PH

01/05/2005
7.44
Y
29/06/1999
Y
 9/3/2000
2











2nd











recurrence











15/1/2005


HC 042
05/11/1997
PH
03/06/2008

10.58
N



3


HC 043
19/11/1997
OLT
22/10/2008

10.90
N



3


HC 052
17/02/1999
PH
18/05/1999
PDV
0.25
N



3


HC 058
14/10/1999
PH
30/01/2008

8.30
N



2


HC 060
15/05/1925
PH



NA



NA


HC 064
10/04/2000
PH

09/07/2005
5.25
Y
15/10/2001
N

3


HC 066
15/09/1999
PH
18/08/2008

8.93
N



2-3


HC 101
03/05/2006
0LT
27/10/2008

2.50
N



2-3


HC 102
12/07/2006
PH
18/08/2006

0.10
N



4


HC 103
16/08/2006
PH
11/06/2008

1.82
Y
15/1/2007 
N

2-3


HC 104
20/09/2006
PH
05/11/2008

2.10
N



2-3


HC 105
11/12/2006
PH
04/07/2007

0.56
Y
15/04/2007
N

3


HC 106
22/01/2007
OLT
16/01/2009

2.00
Y



3


HC 107
25/01/2007
PH
23/10/2008

1.75
N



2


HC 108
12/02/2007
PH
24/09/2008

1.62
N



3


HC 109
19/02/2007
OLT
26/05/2008

1.30
N



2-3


HC 110
 6/02/2007
OLT
04/02/2009

1.95
N



2-3


HC 111
07/03/2007
OLT

03/10/2007
0.70
N



2-3


HC 112
19/03/2007
PH
08/09/2008

1.48
N



2-3


HC 113
23/03/2007
OLT

15/03/2008
1.00
N



2-3


HC 114
03/04/2007
PH
11/09/2007

0.44
N



2


HC 115
01/08/2007
PH
29/04/2008

0.75
N



1


HC 116
09/08/2008
PH
18/04/2008

0.69
N



3


HC 117
25/10/2007
OLT
23/12/2008

1.20
N



2-3


HC 118
25/10/2007
PH
28/09/2008

0.93
N



1


HC 119
03/12/2007
OLT
08/01/2009

1.20
N



2-3


HC 120
18/12/2007
PH
14/10/2008

0.82
N

Y
12/05/2008
2-3


HC 121
02/01/2008
PH
08/08/2008

0.60
N



3


HC 122
16/01/2008
PH
17/10/2008

0.75
Y
10/10/2008
N

2


HC 123
11/02/2008
OLT
01/12/2008

0.80
N



3


HC 124
20/02/2008
PH
26/08/2008

0.52
N



3


HC 125
22/02/2008
OLT
08/01/2009

0.90
N



3


HC 126
12/03/2008
PH
14/08/2008

0.42
Y
6/8/2008
N

1-2


HC 127
19/03/2008
PH
20/06/2008

0.25
Y
4/6/2008
N

2-3


HC 128
20/03/2008
PH
29/08/2008

0.44
N



2


HC 129
01/04/2008
0LT

31/05/2008
0.15
N



3


HC 130
07/04/2008
PH
27/05/2008

0.14
N



3


HC 131
10/04/2008
PH
15/07/2008

0.26
N



2-3


HC 137
19/07/2002
PH
31/03/2008
.
5.67
N
.


NA


HC 138
25/04/2003
PH
03/12/2008
.
5.58
Y
03/10/2003


NA


HC 139
15/05/2002
PH
09/05/2008
.
6.00
N
.


NA


HC 140
03/06/2004
PH
 5/08/2008
.
4.17
Y
30/06/2005


NA


HC 141
06/02/2004
PH
12/03/2009
.
5.08
Y
Dec. 2005


NA


HC 142
14/05/2002
PH
21/06/2006
21/06/2006
4.08
Y
24/03/2006


NA


HC 143
04/03/2002
PH
26/01/2007
.
2.83
Y
2005


NA


HC 144
27/06/2002
PH
17/06/2008
.
6.00
Y
16/03/2004


NA


HC 145
14/11/2002
PH
30/07/2008
.
5.58
Y
09/06/2005


NA


HC 146
30/07/2004
PH
11/12/2008
.
4.33
Y
June 2005


NA


HC 147
23/11/2004
PH
22/09/2008
.
3.83
Y
12/06/2008


NA


HC 148
12/09/2003
PH
15/10/2006
.
3.08
N



NA


HC 149
26/08/2003
PH
16/01/2007
16/01/2007
3.42
N



NA


HC 150
31/01/2003
PH
23/06/2008
.
5.42
N



NA


HC 151
10/12/2004
PH
15/03/2007
.
2.25
N



NA


HC 152
14/05/2003
PH
17/01/2007
17/01/2007
3.67
Y
mars-09


NA


HC 153
25/02/2003
PH
24/12/2007
24/12/2007
4.83
Y
06/05/2005


NA


HC 154
06/09/2004
PH

23/11/2006
2.21
Y
01/01/2005
N

2-3


HC 155
18/10/2004
PH
09/12/2008

4.10
Y
18/10/2004
Y
31/05/2005
2


HC 156
03/02/2005
PH

28/05/2007
2.31
Y
15/06/2006


3


HC 157
24/02/2003
PH

26/10/2006
3.59
Y
15/08/2004


2


HC 159
16/10/2002
PH

18/03/2005
2.42
Y
03/05/2004


2


HC 161
20/08/2003
PH
06/02/2008

4.47
Y



2


HC 162
30/10/2003
PH
25/04/2007

3.49
N



3


HC 163
20/09/2004
PH

07/12/2006
2.21
Y
01/09/2006
N

3


HC 164
05/09/2002
PH
21/03/2007

4.54
N



1


HC 165
08/08/2003
PH
29/05/2008

4.72
N



2


HC 168
10/02/2003
PH
04/02/2009

6.00
Y
15/07/2004
Y
18/02/2008
2


HC 169
10/06/2002
PH
22/03/2005
22/03/2005
2.78
Y
15/03/2003
N

2


HC 170
14/03/2002
PH
28/06/2007

5.29
N



1


HC 171
25/03/2004
PH
17/10/2008

4.57
Y
15/11/2004
N

4


HC 172
10/01/2005
PH
25/11/2008

3.90
Y
25/11/2005
N

3


HC 173
18/12/2003
PH
03/03/2008

4.21
N



1


HC 176
13/03/2002
PH
05/10/2006

4.57
N



2


HC 177
29/10/2003
PH
mars-09

5.42
Y
01/2009


2


HC 178
19/03/2003
PH
19/09/2005

2.50
N



2


HC 179
27/10/2000
PH
06/12/2005

5.17
Y
10/2002


2-3


HC 180
9/4/2002
PH
03/11/2005
03/11/2005
3.58
Y
05/2005


3


HC 181
27/05/2002
PH
mars-09

6.83
Y
04/2008


2


HC 182
30/03/2004
PH
October

3.50
N



1





2007


HC 183
21/07/2003
PH
02/09/2007
02/09/2007
4.08
Y
July 2007


3


HC 184
18/01/2002
PH
08/02/2004
08/02/2004
2.08
Y
April 2002


2


HC 185
19/11/2002
PH
03/03/2005

2.25
N



3


HC 186
31/08/2004
PH
06/11/2006
06/11/2006
2.17
N



3


HC 187
7/06/2001
PH
févr-09

7.67
Y
March 2003


1


HC 188
29/07/2004
PH
avr-09

4.67
Y
July 2004


2


HC 189
30/04/2002
PH
13/08/2005
13/08/2005
3.25
Y
January


2









2005


HC 190
29/07/2003
PH
mars-09

5.58
N



3


























number













max of

Macro-










mitosis

Ndules
Nrmal

Score
Score



Tumor

vascular
vascular
per 10

of
liver
Cir-
META-
META-


HC 000 tumor
differenti-
tumor
invasion
invasion
fields ×
multiple
regen-
A0F0 or
rhosis
VIR
VIR


identification
ation (OMS)
size (mm)
macro
micro
40
Ndules
eration
A0F1
AXF4
Activity
Fibrosis





















HC 001
moderately
120
N
N
NA
N

N
Y
NA
4



differentiated


HC 003
well
60
N
N
NA
N

N
Y
NA
4



differentiated


HC 004
well
100
N
N
NA
N

Y
N
0
1



differentiated


HC 006
well
90
N
Y
NA
N

Y
N
0
1



differentiated


HC 007
well
100
Y
Y
NA
Y

N
N
2
3



differentiated


HC 008
moderately
30
N
N
NA
N

N
Y
N
4



differentiated


HC 009
Moderately
100
Y
Y
NA
Y

N
N
1
3



poorly


HC 010
moderately-
75
N
N
NA
N

N
Y
NA
4



poorly


HC 011
well
15
N
N
NA
Y

N
Y
NA
4



differentiated


HC 012
well
60
N
N
NA
Y

N
Y
NA
4



differentiated


HC 014
Moderate
80
Y
Y
NA
Y

N
Y
NA
4



poor


HC 015
moderately
60
Y
Y
NA
Y

N
N
3
3



differentiated


HC 017
well
100
N
N
NA
N

N
N
NA
3



differentiated


HC 018
moderately
140
Y
Y
NA
N

N
Y
2
4



differentiated


HC 020
well
40
NA
NA
NA
Y

N
Y
NA
4



differentiated


HC 021
NA
100
NA
NA
NA
Y

N
Y
NA
4


HC 022
well
45
N
N
NA
Y

N
Y
NA
4



differentiated


HC 023
well
50
N
N
NA
N

Y
N
NA
0



differentiated


HC 025
well
140
N
N
NA
N

Y
N
0
0



differentiated


HC 026
well
30
Y
Y
NA
Y

N
Y
NA
4



differentiated


HC 027
well
15
N
N
NA
Y
Y
N
Y
NA
4



differentiated


HC 028
moderately
120
N
Y
NA
Y

Y
N
0
0



differentiated


HC 030
moderately
16
NA
NA
NA
N

N
Y
NA
4



differentiated


HC 032
well
60
N
NA
NA
Y

N
Y
NA
4



differentiated


HC 034
well
140
N
N
NA

Y
Y
N
NA
0



differentiated


HC 037
moderately
35
Y
Y
NA
Y
Y
N
Y
NA
4



differentiated


HC 038
moderately
50
N
N
NA
Y

N
Y
NA
4



differentiated


HC 041
well
30
N
N
NA
N

N
Y
NA
4



differentiated


HC 042
moderately
130
prob-
Y
NA
N

N
N
2
1



differentiated

able


HC 043
moderately
15
N
N
NA
Y

N
Y
N
4



differentiated


HC 052
moderately
110
N
Y
NA
Y

N
Y
N
4



differentiated


HC 058
moderately
100
N
N
NA
N

N
N
2
3



differentiated


HC 060
well
55
N
N
NA



differentiated


HC 064
moderately
40
N
N
NA
N

N
N
2
2



differentiated


HC 066
well
75
N
N
NA
Y

N
Y
NA
4



moderately


HC 101
well
35
Y
Y
18
Y
Y
N
Y
2
4



moderately


HC 102
Peu
200
Y
Y
7
N
N
N
N
1
1



différencié


HC 103
well
55
N
Y
8
N
Y
N
Y
3
4



moderately


HC 104
well
160
prob-
Y
10
Y
N
Y
N
0
1



moderately

able


HC 105
moderately
40
Y
Y
20
Y
Y
N
Y
2
4



differentiated


HC 106
moderately
80
Y
Y
32
Y
N
N
Y
1
4



differentiated


HC 107
well
60
N
N
1
N
N
Y
N
0
0-1



differentiated


HC 108
moderately
26
N
Y
18
N
N
N
N
1
1



differentiated


HC 109
well
30
N
N
<1
Y
Y
N
Y
2
4



moderately


HC 110
well
30
N
Y
1á5
Y
Y
N
Y
1
4



moderately


HC 111
well
40
Y
Y
45
Y
Y
N
Y
1
4



moderately


HC 112
well
18
N
N
0
N
N
N
N
2
2



moderately


HC 113
well
50
Y
Y
25
Y
Y
N
Y
1
4



moderately


HC 114
well
36
N
N
<1
N
N
N
N
2
3



differentiated


HC 115
well
90
N
N
0
N
N
N
N
2
1



differentiated


HC 116
moderately
140
N
N
12
N
N
N
N
2
3



differentiated


HC 117
well
28
N
N
4
Y
Y
N
Y
2
4



moderately


HC 118
well
40
N
N
<1
N
N
Y
N
0
1



differentiated


HC 119
well
26
N
Y
15
Y
Y
N
Y
2
4



moderately


HC 120
well
20
N
Y
3
Y
N
N
Y
1
4



moderately


HC 121
moderately
150
prob-
Y
8á30
Y
Y
N
Y
2
4



differentiated

able


HC 122
well
20
Y
Y
8
Y
?
N
Y
1
4



differentiated


HC 123
moderately
43
prob-
prob-
4
Y
N
N
Y
2
4



differentiated

able
able


HC 124
moderately
62
N
N
4
N
N
N
N
1
1



differentiated


HC 125
moderately
33
N
Y
2
Y
N
N
Y
2
4



differentiated


HC 126
well
130
Y
Y
2
Y
N
Y
N
0
1



differentiated


HC 127
well
115
Y
Y
>100
N
N
N
N
1
1



moderately


HC 128
well
110
N
Y
5
N
N
N
N
2
2



moderately


HC 129
moderately
30
N
Y
40
Y
N
N
N
2
3



differentiated


HC 130
moderately
38
N
prob-
12
N
N
N
N
1
2



differentiated


able


HC 131
well
120
N
Y
20á25
N
N
Y
N
0
1



moderately


HC 137
moderately
10
NA
NA
NA


Y
.
.
.



differentiated


HC 138
well
5.5
NA
NA
NA


Y
N
.
.



differentiated


HC 139
moderately
16
NA
NA
NA


Y
.
.
.



differentiated


HC 140
well
15
NA
NA
NA


N
N
0
1



differentiated


HC 141
well
3.5
NA
NA
NA


N
N
.
.



differentiated


HC 142
well
8
NA
NA
NA


Y
.
.
.



differentiated


HC 143
well
3
NA
NA
NA


N
Y
1
4



differentiated


HC 144
well
15
NA
NA
NA


Y .
.
.



differentiated


HC 145
well
6
NA
NA
NA


N
.
0
3



differentiated


HC 146
well
7.5
NA
NA
NA


N
N
.
2



differentiated


HC 147
moderately
15
NA
NA
NA


N
N
0
3



differentiated


HC 148
moderately
21
NA
NA
NA


Y
N
.
.



differentiated


HC 149
NA
8
NA
NA
NA


N
N
0
0


HC 150
moderately
13
NA
NA
NA


N
.
0
3



differentiated


HC 151
well
6.5
NA
NA
NA


N
Y
2
4



differentiated


HC 152
well
3.5
NA
NA
NA


N
N
0
2



differentiated


HC 153
well
5
NA
NA
NA


N
.
0
3



differentiated


HC 154
well
45
Y
Y
25
N
N
Y
N
0
1



differentiated


HC 155
well
24
N
N
1
N
N
N
Y
2
4



differentiated


HC 156
moderately
70
N
Y
16
Y
N
N
Y
2
4



differentiated


HC 157
well
140
Y
Y
2
N
N
Y
N
0
1



differentiated


HC 159
well
35
N
N
NA
N
N
N
Y
2
4



differentiated


HC 161
well
210
N
Y
2
N
N
N
N
1
1



differentiated


HC 162
moderately
130
Y
Y
77
N
N
Y
N
0
0



differentiated


HC 163
moderately
80
N
Y
4
N
N
N
Y
1
4



differentiated


HC 164
well
90
N
N
1
N
N
Y
N
0
1



differentiated


HC 165
well
30
N
Y
4
N
N
N
N
0
2



differentiated


HC 168
well
25
N
N
1
Y
Y
N
Y
2
4



differentiated


HC 169
well
35
N
N
NA
N
N
N
Y
2
4



differentiated


HC 170
well
220
N
N
0
N
N
Y
N
0
0



differentiated


HC 171
Peu
70
Y
Y
10
Y
N
N
N
1
2



différencié


HC 172
moderately
40
N
Y
28
N
N
N
N
2
3



differentiated


HC 173
well
40
N
N
0
N
N
Y
N
0
0



differentiated


HC 176
well
75
N
N
NA
N
N
Y
N
0
0



differentiated


HC 177
moderately
2.3
NA
N
NA
Y



A1
F4



differentiated


HC 178
well
6.5
NA
N
NA
Y



A1
F4



differentiated


HC 179
well-moder-
9
NA
Y
NA
Y



A2
F1



ate-poor


HC 180
moderately
15
NA
Y
NA
Y



A2
F2



differentiated


HC 181
well
3.5
NA
Y
NA
Y



A1
F4



moderately


HC 182
well
11
NA
N
NA
N




F1



differentiated


HC 183
well
8
NA
Y
NA
N



A1
F3



differentiated


HC 184
well
6.5
NA
N
NA
N




F1



differentiated


HC 185
moderately
3.5
NA
N
NA
N



A1
F4



differentiated


HC 186
well
17
NA
Y
NA
N




F0



moderately


HC 187
well
8
NA
Y
NA
N




F4



differentiated


HC 188
well
13
NA
N
NA
N




F0



differentiated


HC 189
well
22
NA
Y
NA
Y




F1



differentiated


HC 190
moderately
15
NA
N
NA
Y



A1
F3



differentiated



















chronic








HC 000 tumor
viral
Etiology
Etiology





identification
hepatitis
HBV
HCV
alcool
Hemochromatos
—NASH







HC 001
N
N
N
Y
N
N



HC 003
Y
N
Y
N
N
N



HC 004
N
N
N
N
N
N



HC 006
N
N
N
Y
Y
N



HC 007
N
N
N
Y
N
N



HC 008
Y
N
Y
N
N
N



HC 009
N
N
N
Y
N
N



HC 010
Y
Y
N
N
N
N



HC 011
Y
Y
Y
N
N
N



HC 012
Y
Y
N
N
N
N



HC 014
Y
N
Y
Y
N
N



HC 015
N
N
N
Y
N
N



HC 017
Y
Y
N
N
N
N



HC 018
N
N
N
Y
N
N



HC 020
N
N
N
Y
N
N



HC 021
N
N
N
Y
N
N



HC 022
N
N
N
Y
N
N



HC 023
N
N
N
N
N
N



HC 025
N
N
N
N
N
N



HC 026
Y
Y
N
N
N
N



HC 027
Y
N
Y
N
N
N



HC 028
N
N
N
N
N
N



HC 030
N
N
N
Y
N
N



HC 032
Y
N
Y
N
N
N



HC 034
N
N
N
N
N
N



HC 037
N
N
N
Y
N
N



HC 038
Y
N
Y
N
N
N



HC 041
Y
N
Y
N
N
N



HC 042
Y
Y
N
N
N
N



HC 043
Y
N
Y
N
N
N



HC 052
Y
Y
N
N
N
N



HC 058
Y
N
Y
N
N
N



HC 060



HC 064
Y
N
Y
N
N
N



HC 066
Y
Y
N
Y
N
N



HC 101
Y
Y
Y
Y
N
N



HC 102
Y
Y
Y
N
N
N



HC 103
Y
Y
N
N
N
N



HC 104
N
N
N
N
N
N



HC 105
Y
N
Y
N
N
N



HC 106
Y
Y
N
N
N
N



HC 107
N
N
N
Y
N
N



HC 108
Y
N
Y
N
N
N



HC 109
N
N
N
Y
N
Y



HC 110
Y
N
Y
Y
N
N



HC 111
N
N
N
Y
N
N



HC 112
N
N
N
N
N
Y



HC 113
Y
N
Y
N
N
N



HC 114
N
N
N
Y
N
N



HC 115
N
N
N
N
N
Y



HC 116
Y
Y
N
N
N
N



HC 117
Y
N
Y
N
N
N



HC 118
N
N
N
N
N
N



HC 119
Y
N
Y
Y
N
N



HC 120
Y
Y
N
N
N
N



HC 121
N
N
N
Y
N
Y



HC 122
Y
Y
N
N
N
N



HC 123
Y
N
Y
N
N
N



HC 124
N
N
N
N
N
Y



HC 125
N
N
N
Y
N
N



HC 126
N
N
N
N
N
N



HC 127
Y
Y
N
N
N
N



HC 128
N
N
N
Y
N
N



HC 129
Y
N
Y
N
N
N



HC 130
Y
Y
N
N
N
N



HC 131
N
N
N
N
N
N



HC 137
N
N
N
Y
N
N



HC 138
N
N
N
N
N
N



HC 139
N
N
N
N
N
N



HC 140
N
N
N
N
N
N



HC 141
N
N
N
N
N
N



HC 142
N
N
N
Y
N
N



HC 143
N
N
N
N
Y
N



HC 144
N
N
N
N
N
N



HC 145
N
N
N
Y
N
N



HC 146
Y
Y
N
Y
N
N



HC 147
N
N
N
N
Y
N



HC 148
N
N
N
N
N
N



HC 149
N
N
N
N
N
N



HC 150
N
N
N
Y
N
N



HC 151
N
N
N
N
N
Y



HC 152
N
N
N
N
Y
N



HC 153
Y
Y
N
Y
N
N



HC 154
N
N
N
N
N
N



HC 155
N
N
N
Y
N
N



HC 156
Y
N
Y
N
N
N



HC 157
N
N
N
N
N
N



HC 159
N
N
N
Y
N
N



HC 161
N
N
N
N
N
N



HC 162
N
N
N
N
N
N



HC 163
N
N
N
Y
N
N



HC 164
N
N
N
N
N
N



HC 165
N
N
N
N
Y
N



HC 168
Y
N
Y
N
N
N



HC 169
N
N
N
Y
N
N



HC 170
N
N
N
N
N
N



HC 171
N
N
N
Y
N
N



HC 172
N
N
N
N
Y
N



HC 173
N
N
N
N
N
N



HC 176
N
N
N
N
N
N



HC 177
Y
Y
N
N
N
N



HC 178
N
N
N
Y
N
N



HC 179
Y
N
Y
N
N
N



HC 180
Y
Y
N
N
N
N



HC 181
N
N
N
Y
N
N



HC 182
Y
N
Y
N
N
N



HC 183
Y
Y
N
N
N
N



HC 184
N
N
N
Y
N
N



HC 185
Y
N
Y
N
N
N



HC 186
NA
NA
NA
NA
NA
NA



HC 187
N
N
N
Y
N
N



HC 188
N
N
N
Y
N
N



HC 189
N
N
N
Y
N
N



HC 190
Y
Y
N
N
N
N










REFERENCES



  • Assou, S., Le Carrour, T., Tondeur, S., Strom, S., Gabelle, A., Marty, S., Nadal, L., Pantesco, V., Reme, T., Hugnot, J. P., et al. (2007). A meta-analysis of human embryonic stem cells transcriptome integrated into a web-based expression atlas. Stem Cells 25, 961-973.

  • Boyault, S., Rickman, D. S., de Reynies, A., Balabaud, C., Rebouissou, S., Jeannot, E., Herault, A., Saric, J., Belghiti, J., Franco, D., et al. (2007). Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 45, 42-52.

  • Finegold, M. J., Lopez-Terrada, D. H., Bowen, J., Washington, M. K., and Qualman, S. J. (2007). Protocol for the examination of specimens from pediatric patients with hepatoblastoma. Arch Pathol Lab Med 131, 520-529.

  • Fodde, R., and Brabletz, T. (2007). Wnt/beta-catenin signaling in cancer sternness and malignant behavior. Curr Opin Cell Biol 19, 150-158.

  • Glinsky, G. V., Berezovska, O., and Glinskii, A. B. (2005). Microarray analysis identifies a death-from-cancer signature predicting therapy failure in patients with multiple types of cancer. J Clin Invest 115, 1503-1521.

  • Hirschman, B. A., Pollock, B. H., and Tomlinson, G. E. (2005). The spectrum of APC mutations in children with hepatoblastoma from familial adenomatous polyposis kindreds. J Pediatr 147, 263-266.

  • Irizarry, R. A., Hobbs, B., Collin, F., Beazer-Barclay, Y. D., Antonellis, K. J., Scherf, U., and Speed, T. P. (2003). Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249-264.

  • Lee, J. S., Heo, J., Libbrecht, L., Chu, I. S., Kaposi-Novak, P., Calvisi, D. F., Mikaelyan, A., Roberts, L. R., Demetris, A. J., Sun, Z., et al., (2006). A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells. Nat Med 12, 410-416.

  • McLin, V. A., Rankin, S. A., and Zorn, A. M. (2007). Repression of Wnt/β-catenin signaling in the anterior endoderm is essential for liver and pancreas development. Development 134, 2207-2217.

  • Perilongo, G., Shafford, E., and Plaschkes, J. (2000). SIOPEL trials using preoperative chemotherapy in hepatoblastoma. Lancet Oncol 1, 94-100.

  • Rowland, J. M. (2002). Hepatoblastoma: assessment of criteria for histologic classification. Med Pediatr Oncol 39, 478-483.

  • Schnater, J. M., Kohler, S. E., Lamers, W. H., von Schweinitz, D., and Aronson, D. C. (2003). Where do we stand with hepatoblastoma? A review. Cancer 98, 668-678.

  • Taniguchi, K., Roberts, L. R., Aderca, I. N., Dong, X., Qian, C., Murphy, L. M., Nagorney, D. M., Burgart, L. J., Roche, P. C., Smith, D. I., et al. (2002). Mutational spectrum of beta-catenin, AXIN1, and AXIN2 in hepatocellular carcinomas and hepatoblastomas. Oncogene 21, 4863-4871.

  • Wei, Y., Fabre, M., Branchereau, S., Gauthier, F., Perilongo, G., and Buendia, M. A. (2000). Activation of beta-catenin in epithelial and mesenchymal hepatoblastomas. Oncogene 19, 498-504.

  • Lustgarten, J. L. et al (2008)—Improving classification performance with discretization on biomedical datasets. AMIA 2008 Symposium Proceedings,


Claims
  • 1. Method to determine the gene expression profile on a biological sample, comprising: a. assaying the expression of a set of genes in a sample previously obtained from a patient diagnosed for a liver tumor, wherein said set comprises from 2 to 16 genes or consists of 2 to 16 genes, said 2 to 16 genes being chosen in the group consisting in the alpha-fetoprotein (AFP), aldehyde dehydrogenase 2 (ALDH2), amyloid P component serum (APCS), apolipoprotein C-IV (APOC4), aquaporin 9 (AQP9), budding uninhibited by benzimidazoles 1 (BUB1), complement componant 1 (C1S), cytochrome p450 2E1 (CYP2E1), discs large homolog 7 (DLG7), dual specificity phosphatase 9 (DUSP9), E2F5 transcription factor (E2F5), growth hormone receptor (GHR), 4-hydroxyphenylpyruvase dioxygenase (DHP), immunoglogulin superfamily member 1 (IGSF1), Notchless homolog 1 (NLE1) and the ribosomal protein L10a (RPL10A) genes; and b. determining the gene expression profile of said sample.
  • 2. Method according to claim 1, which further comprises determining the grade of the liver tumor providing the sample, for example by comparing the obtained gene expression profile of said sample to the gene expression profile of a reference sample or to the gene expression profiles of a collection of reference samples or by applying a discretization method for classification.
  • 3. Method according to claim 1 or 2, wherein the assay of the expression of said set of genes comprises a step of detecting nucleotide targets, wherein each nucleotide target is a product resulting from the expression of one of the genes in said set.
  • 4. Method according to claim 2, wherein said nucleotide targets are mRNA.
  • 5. Method according to any one of claims 1 to 4, wherein the assay of the expression of said set of genes comprises an amplification step, such as performed by qualitative polymerase chain reaction prior to a step of detecting the mRNA of each gene of said set.
  • 6. Method according to any one of claims 1 to 5, wherein the assay of the expression of said set of genes comprises a hybridization step, such as one performed by hybridization on a solid or liquid support, especially on an array, prior to a step of detecting the mRNA of each gene of said set.
  • 7. Method according to any one of claims 2 to 5, wherein said detected nucleotide targets are quantified with respect to at least one nucleotide target, expression product of an invariant gene, such as ACTG1, EFF1A1, PNN and RHOT2 genes.
  • 8. Method according to any one of claims 1 to 7, wherein said liver tumor is a hepatoblastoma (HB) or a hepatocellular carcinoma (HCC).
  • 9. Method according to any one of claims 2 to 8, wherein said method comprises, before step a., the preparation of said nucleotide targets from the sample.
  • 10. Method according to any one of claims 1 to 9, wherein said set of genes comprises or consists in a set chosen in the group consisting of: (a) E2F5 and HPD genes; (b) APCS, BUB1, E2F5, GHR and HPD genes; (c) ALDH2, APCS, APOC4, BUB1, C1S, CYP2E1, E2F5, GHR and HPD genes; (d) ALDH2, APCS, APOC4, AQP9, BUB1, C1S, DUSP9, E2F5 and RPL10A genes; (e) ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, E2F5, GHR, IGSF1 and RPL10A genes; and (f) AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.
  • 11. Method enabling the determination of the tumor grade on a patient's biological sample, which comprises a classification of the tumor through discretization according the following steps: In a method according to any of claims 1 to 10, measuring the expression and especially the relative (normalized) expression of each gene in a set of genes defined as the signature of the tumor, for example by quantitative PCR thereby obtaining data as Ct or preferably Delta Ct in said biological sample wherein said set of genes is divided in two groups, a first group consisting of the proliferation-related genes and a second group consisting of the differentiation-related genes, comparing the values measured for each gene, to a cut-off value determined for each gene of the set of genes, and assigning a discretized value to each of said measured expression values with respect to said cut-off value, said discretized value being advantageously a “1” or a “2” and optionally a “1.5” value with respect to the cut-off value, determining the average of the discretized values for the genes, in each group of the set of genes, determining a score calculated as a ratio the average for the discretized values for the proliferating-related genes on the average for the discretized values for the differentiation-related genes, comparing the obtained score for the biological sample with one or more sample cut-off(s) value(s), wherein each cut-off value corresponds to a selected percentile, determining the tumor grade as C1 or C2, as a result of the classification of the biological sample with respect to said sample cut-off.
  • 12. Method according to claim 11, wherein the relative expression determined for the profiled gene is obtained by normalizing with respect to the invariant RHOT2 gene.
  • 13. Method according to claim 11, wherein the determination of the tumor grade on a biological sample comprises applying the following conditions: a) for a hepatoblastoma: the set of assayed genes for profiling is constituted of the 16 genes disclosed; the invariant gene (of reference) is RHOT2; the cut-offs value for each gene are: AFP: 3.96139596; ALDH2: 4.3590482; APCS: 4.4691582; APOC4: 2.03068712; AQP9: 3.38391456; BUB1: −1.41294708; C1S: 4.24839464; CYP2E1: 6.70659644; DLG7: −3.3912188; DUSP9: 2.07022648; E2F5: −0.72728656; GHR: −0.1505569200; HPD: 2.27655628; IGSF1: 0.1075015200; NLE: −0.02343571999; RPL10A: 6.19723876. the cut-off value for the sample is 0.91 and a sample with a score above 0.91 is classified into the C2 class and a sample with a score below 0.91 is classified into the C1 class. b) for a hepatocellular carcinoma: the set of assayed genes for profiling is constituted of the 16 genes disclosed; the invariant gene (of reference) is RHOT2; the cut-offs value for each gene is: Gene nameCut-off for TaqmanCut-off for SybrGreenAEP−1.2634010−2.3753035ALDH24.0141435.314302APCS5.61429076.399079APOC4−0.79631584.656336AQP94.28360115.446966BUB1−1.2736579−3.634476C1S6.35146796.240002CYP2E16.95624195.829384DLG7−2.335694−4.614352DUSP9−7.979559−1.8626715E2F5−0.4400218−1.367846GHR1.08326321.169362HPD6.4803286.736329IGSF1−4.84177857.6653982NLE−1.6167268−1.82226RPL10A6.24830565.731897the cut-off value for the sample corresponding to the 67th percentile is 0.925 and the cut-off value corresponding to the 33th percentile is 0.66 and a sample with a score above 0.925 is classified into the C2 class and a sample with a score below 0.66 is classified into the C1 class.
  • 14. Method according to claim 13 wherein in the case of a hepatocellular carcinoma, a sample with a score (initial score) between 0.66 and 0.925 is refined to obtain a modified score, the modified score being either “1” or “2” depending on the calculated average of the discretized values for the proliferation-related genes only, said average being discretized at a determined percentile (the 60th for example) and “1” is assigned if the sample has an average below the value at the percentile of reference and “2” is assigned if the sample has an average above the value at the percentile of reference.
  • 15. Kit, suitable to carry out the method as defined in any one of claims 1 to 13, comprising a. a plurality of pairs of primers specific for a set of genes to be assayed, said set comprising 2 to 16 genes or consisting of 2 to 16 genes, said 2 to 16 genes being chosen in the group consisting of AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes; and b. optionally reagents necessary for the amplification of the nucleotide targets of these genes by said primers, and optionally reagents for detecting the amplification products.
  • 16. Kit according to claim 14, wherein each primer is 10 to 30 bp in length and has at least 80% similarity with its complementary sequence in the nucleotide target, preferably 100%.
  • 17. Kit according to claim 14 or 15, wherein said pairs of primers are chosen in the group consisting of:
  • 18. A set of probes, suitable to carry out the method as defined in any one of claims 1 to 13, comprising a plurality of probes specific for a set of genes to assay, said set comprising or having from 2 to 16 genes, said 2 to 16 genes being chosen in the group consisting of AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.
  • 19. A set of probes according to claim 17, wherein said probes are 50 to 200 bp in length and have at least 80% similarity to the complementary sequence of the nucleotide target of the gene, preferably 100%.
  • 20. A solid support, especially an array comprising a set of probes as defined in claims 17 or 18 linked to a support.
  • 21. A composition comprising a set of probes as defined in claim 17 or 18, in solution.
  • 22. A kit comprising a set of probes as defined in claim 17 or 18, a solid support as defined in claim 19 or a composition as defined in claim 20, and optionally reagents necessary for the hybridization of said nucleotide targets to said probes.
  • 23. Set of probes, solid support, arrays, compositions or kits according to any one of claims 14 to 21, suitable for assaying a set of genes which comprises or consists in a set chosen in the group consisting of: (a) E2F5 and HPD genes; (b) APCS, BUB1, E2F5, GHR and HPD genes; (c) ALDH2, APCS, APOC4, BUB1, C1S, CYP2E1, E2F5, GHR and HPD genes; (d) ALDH2, APCS, APOC4, AQP9, BUB1, CIS, DUSP9, E2F5 and RPL10A genes; (e) ALDH2, APCS, APOC4, AQP9, C1S, CYP2E1, E2F5, GHR, IGSF1 and RPL10A genes; and (f) AFP, ALDH2, APCS, APOC4, AQP9, BUB1, C1S, CYP2E1, DLG7, DUSP9, E2F5, GHR, HPD, IGSF1, NLE1 and RPL10A genes.
  • 24. Set of probes, solid support, arrays, compositions or kits according to any one of claims 14 to 23, wherein the invariant gene is the RHOT2 gene or the PNN gene.
  • 25. Use of a set of probes, solid support, arrays, compositions or kits according to any one of claims 14 to 24, to determine the grade of a liver tumor in a sample obtained from a patient.
  • 26. Use according to claim 25 or method of claim 11, wherein for a hepatoblastoma or for a hepatocellular carcinoma the cut-off value of the profiled genes are determined for the overexpressed proliferation-related genes at a percentile within the range of the 60th to the 80th percentile, especially at the 67th percentile and the cut-off value of the profiled genes are determined for the downregulated differentiation-related genes at a percentile within the range of the 30rd to 45th percentile, especially at the 33rd or 40th percentile and the cut-off value of the sample is determined within the same range of the 60th to the 80th percentile.
  • 27. Use of a set of probes, arrays, compositions or kits according to any one of claims 14 to 21, to determine, in a patient, the risk of developing metastasis.
  • 28. Use of a set of probes, arrays, compositions or kits according to any one of claims 14 to 21, to define the therapeutic regimen to apply to said patient.
Priority Claims (2)
Number Date Country Kind
08290628.0 Jun 2008 EP regional
09151808.4 Jan 2009 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/IB2009/006450 6/26/2009 WO 4/7/2011
Related Publications (1)
Number Date Country
20110183862 A1 Jul 2011 US