WO2008021163A2 - Kits and methods for determining risk for primary liver cancer - Google Patents

Kits and methods for determining risk for primary liver cancer Download PDF

Info

Publication number
WO2008021163A2
WO2008021163A2 PCT/US2007/017690 US2007017690W WO2008021163A2 WO 2008021163 A2 WO2008021163 A2 WO 2008021163A2 US 2007017690 W US2007017690 W US 2007017690W WO 2008021163 A2 WO2008021163 A2 WO 2008021163A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
hepatocellular carcinoma
serum
kit
hcc
Prior art date
Application number
PCT/US2007/017690
Other languages
French (fr)
Other versions
WO2008021163A3 (en
Inventor
Claus Fimmel
Yie-Hwa Chang
Original Assignee
Saint Louis University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Saint Louis University filed Critical Saint Louis University
Publication of WO2008021163A2 publication Critical patent/WO2008021163A2/en
Publication of WO2008021163A3 publication Critical patent/WO2008021163A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney

Definitions

  • the invention is directed generally to medical diagnostics and specifically to diagnostic screens for primary liver cancer.
  • Hepatocellular carcinoma is the fifth most common cancer in the world today, and the overall 5-year survival rate remains less than 5% (1). Although the incidence rate will likely fall with the institution of mass vaccination against the hepatitis B virus (2), major impacts will not be felt immediately as the age of presentation in most areas of the world is over 50 years long. More importantly, there is no immediate prospect of a vaccine against the hepatitis C virus on the market, the major aetiological factor for HCC in the US, Japan and Southern Europe (3, 4). Despite the absence of randomized clinical trials, there is strong evidence that surgical resection, liver transplantation or ablative therapies significantly improve survival (5, 6).
  • Plasma/serum proteomics Traditionally, serum proteomics studies have been based on two-dimensional gel electrophoresis, though this method does not appear to be sufficiently sensitive for the detection of low-abundance marker proteins. A number of such studies have been performed in HCC patients. Steel and coworkers performed 2-dimensional gel electrophoresis of sera from HBV-infected patients with and without HCC (16). Two protein signals were consistently less abundant in HCC patients as compared to HBV-negative controls and HBV-carriers (17). They were identified by mass spectroscopy as fragments of C3 complement and apolipoprotein A1, respectively.
  • Serum qlvcoproteomics Cancer-specific changes in the glycosylation of serum proteins have been previously reported in several types of cancer. Mehta and colleagues recently reported significant increases in the core- fucosylation of several serum proteins in woodchucks with HBV-related HCC, and introduced lectin affinity purification methods to identify and quantitate such proteins (26). The authors subsequently extended their analysis to patients with HBV-related liver disease, and documented similar changes in a number of highly-abundant proteins (including hemopexin, alpha-acid glycoprotein, alpha-1- antichymotrypsin, alpha-1 -antitrypsin, and haptoglobin) as well as AFP and GP73, two previously described HCC markers.
  • highly-abundant proteins including hemopexin, alpha-acid glycoprotein, alpha-1- antichymotrypsin, alpha-1 -antitrypsin, and haptoglobin
  • Tissue proteomics Proteomics studies on liver tissues have traditionally utilized a combination of two-dimensional gel electrophoresis and mass spectrometry analysis. Li and colleagues performed a comprehensive liver proteomics analysis of patients with HBV-related HCC, comparing the profiles of cancer tissue with that of surrounding non-cancerous tissue (27). They identified 80 proteins with differential expression in HCC (43 upregulated in HCC, 37 downregulated) by peptide mass spectrometry, and assigned them to several functional categories. A few candidates, including proliferating cell antigen and stathmin 1, were confirmed by Western blotting analysis. Um identified 21 proteins with significant changes in expression in cirrhotic livers with HCC (28).
  • TAA tumor-derived autoantibodies
  • P62 nucleophosmin
  • P62 nuclear protein that had previously been found to be present at elevated levels in the serum of HCC patients.
  • HCC-TAAs Increased cellular nucleophosmin expression and abnormal protein processing may trigger the formation of a cell- or antibody-mediated autoimmune response (35).
  • a common feature of HCC-TAAs is their low apparent sensitivity - positive serum reactivities are typically found in less than one fifth of all cases (36). Furthermore, the reported sensitivities were typically obtained in comparing HCC patients and healthy subjects, whereas there are few studies on cirrhotic patients who represent a more relevant target population.
  • current data suggest that the "cancer specificity" of certain TAAs may be quite high (approaching 90% in some cases) - a feature that would compare favorably to many of the conventional serum markers (37).
  • One potential limitation may be the lack of liver- specificity, since the same TAAs may occur in different malignancies. At present, none of the individual TAAs has been subjected to a rigorous prospective analysis in HCC cohorts (see table 1).
  • Diamandis EP Mass spectrometry as a diagnostic and a cancer biomarker discovery tool: opportunities and potential limitations. MoI Cell Proteomics 2004; 3: 367-378.
  • Nerve injury stimulates the secretion of apolipoprotein E by nonneuronal cells, Proc. Natl. Acad. Sci. U.S.A. 83, 1130-1134.
  • HCC hepatocellular carcinoma
  • the present teachings include methods for diagnosing hepatocellular carcinoma in a subject.
  • these methods include a) obtaining a serum sample from a subject, and b) quantifying, in the serum sample, at least one protein that is differentially upregulated in patients diagnosed with HCC.
  • a subject can be diagnosed with HCC if the quantity of at least one of the proteins is determined to be elevated relative to control levels.
  • serum proteins that can be differentially upregulated in a subject with HCC can be, without limitation, apolipoprotein E 1 Trypsin precursor, KIAA0284, Apolipoprotein A-I precursor, v-kit (c-kit homologue), c-kit and Apolipoprotein C-III precursor.
  • an assay for measuring the amount of a protein which is upregulated in HCC can be an aptamer-based assay, an antibody-based assay, a mass spectroscopy assay, or a high performance liquid chromatography assay.
  • an antibody-based assay can be an ELISA, a radioimmunoassay or a Western blot.
  • the methods can further include removing at least one high-abundance serum protein from the serum sample prior to analyzing a serum sample for the quantity of a protein diagnostic for HCC.
  • high-abundance proteins include albumin, IgG 1 antitrypsin, IgA 1 transferrin and haptoglobin. In some aspects, all six of these proteins can be removed from the serum sample prior to the analysis.
  • kits for diagnosing hepatocellular carcinoma can include, without limitation, a capture antibody directed against a first epitope of an HCC serum biomarker, a solid support upon which the capture antibody is immobilized, a detection antibody directed against a second epitope of the HCC serum biomarker; and a labeled secondary antibody directed against the detection antibody.
  • the solid support can comprise at least one plastic bead or a multiwell plate such as an ELISA plate well known to skilled artisans.
  • an HCC serum biomarker can be, without limitation, a protein such as an apolipoprotein E 1 a Trypsin precursor, a KIAA0284, a Apolipoprotein A-I precursor, a v-kit (c-kit homologue), a c-kit or an Apolipoprotein C-III precursor.
  • a labeled secondary antibody comprised by a kit can include a label such as a fluorophore, an enzyme or a radioisotope.
  • Figure 1 illustrates the 2D gel image of c-kit protein from a cirrhosis serum sample and an HCC serum sample.
  • Figure 2 illustrates the 2D gel image of Apo E protein from a cirrhosis serum sample and an HCC serum sample.
  • Figure 3 illustrates a Western blot analysis of serum samples using anti-kit antibodies.
  • Figure 4 illustrates a Western blot analysis of serum samples using anti-ApoE antibodies
  • FIG. 5 illustrates fractionation of serum proteins using the PF-2D LC system.
  • HCC hepatocellular carcinoma
  • the invention is directed to a method for determining the risk of a patient having or acquiring hepatocellular carcinoma ("HCC"), the steps comprising obtaining a serum sample from a patient, detecting and/or quantifying any one or more of the biomarkers listed in Table 2, alone or in combination with one or more known HCC serum biomarkers, and determining the patient's risk for having or acquiring HCC.
  • HCC hepatocellular carcinoma
  • the detecting step may be any one of myriad well-known methods in the art, including for example aptamer- based assays, antibody-based assays (e.g., ELISA, RIA, western), mass spec, HPLC, and the like.
  • the detection method is an antibody-based ELISA, as is currently the preference in the medical diagnostics art.
  • the preferred determining step is quantifying the signal generated by the detection of the one of more the biomarkers of Table 2 in the test sample (e.g., serum of an individual suspected of having risk factors for HCC) and comparing that signal quantity to a control sample. A difference between the test and control values for the biomarkers can be interpreted to assess whether the test individual is at risk for HCC and/or needs additional testing and/or therapy.
  • the invention is directed to a diagnostic kit for determining the risk of an individual for having or getting HCC.
  • the preferred kit is based upon the ELISA immunodetection method, but the skilled artisan in the practice of this invention would recognize that other methods to which the kit can be based upon are available and applicable.
  • the kit comprises a substrate to which a capture antibody is fixed, a detection antibody, and a detectable moiety antibody.
  • the preferred substrate for example may be a plastic bead or surface of a multiwell plate.
  • the preferred capture antibody recognizes a first epitope on a HCC serum biomarker.
  • the preferred detection antibody recognizes a second epitope on the HCC serum biomarker.
  • the preferred detectable moiety antibody recognizes the detection antibody and has a detection moiety such as for example a fluorophore, chemiluminescence molecule, enzyme, or the like.
  • HCV-related chronic liver disease Patients with HCV-related chronic liver disease were enrolled at the Johns Cochran VA Hospital and Saint Louis University Health Sciences Center.
  • two groups of male patients with proven, compensated liver cirrhosis were studied, one without evidence of hepatocellular cancer, the other with biopsy-proven small HCC ( ⁇ 3 cm) that were discovered by surveillance imaging studies. Serum samples were obtained from patients after an overnight fast. None of the patients were receiving treatment for their HCC at the time of the blood draw.
  • the clinical characteristics of the two groups are provided in Table 3.
  • the Agilent spin cartridge was used to remove six high-abundant proteins from the serum pooled from three patients. These six proteins include albumin, IgG, antitrypsin, IgA, transferrin, and haptoglobin according to manufacturer's procedures. Specific removal of six high- abundant proteins can deplete approximately 85%-88% of total protein mass from human serum.
  • Gel images are acquired with a Typhoon 9410 imager (Amersham Biosciences). Gel images are then analyzed using Phoretix 2D Evolution gel analysis software (Nonlinear Dynamics Ltd.). Automatic spot detection was performed with default parameters; spot editing and removal of artifacts were performed manually. Subsequent data analyses were performed on the MS- identified spots and manually matched among the gels. The normalized spot volume relative to the total volume of all detected spots in a gel (%vol) was used to calculate averages and CV between gels of replicate samples for individual spots. Microsoft Excel's STDEV function was used to calculate the SD for a given spot. A propagation of errors calculation was used to calculate the SD of expression ratios, which was subsequently used to calculate a CV for expression ratios.
  • Searches were restricted to 50 ppm MS mass tolerance, and 0.3 Da MS/MS mass tolerance. Search results were ranked by protein score confidence interval% (Cl%), a GPS calculation based on Mascot score with the significance threshold removed to normalize results across different databases. Protein hits with protein score Cl% between 95 and 100 were accepted for identifications.
  • serum protein with six abundant proteins depleted as described was used and the buffer was exchanged to the starting buffer provided by the ProteomeLab PF 2D kit.
  • 1 mg of the normal serum protein were resolved using the default method, and proteins were collected by interval of 0.3 pH unit from the first dimension separation using the ProteomeLab PF 2D fractionation system (Beckman Coulter). Twenty fractions were generated and each fraction was further separated by the C18 column with a linear gradient of 0%-100% ACN. The results are analyzed by proteovue program from Beckman Coulter Fractionation of serum proteins using PF-2D LC system from Beckman Coulter as shown in Figure 5.

Abstract

Methods and kits for diagnosing hepatocellular carcinoma are disclosed. The methods comprise detecting in a serum sample from a subject the quantity of at least one serum protein which is upregulated in patients with hepatocellular carcinoma. Kits of the present teachings include a capture antibody directed against a first epitope of a serum protein which is upregulated in patients with hepatocellular carcinoma, a detection antibody against a second epitope of the protein, and a labeled secondary protein directed against the detection antibody. The capture antibody can be immobilized on a solid support, such as a multiwell plate or a plastic bead.

Description

Kits and Methods for Determining Risk for Primary Liver
Cancer
CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a non-provisional of Ser. No. 60/821,921 , filed August 9, 2006 which is incorporated herein by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT Not Applicable
Background of the Invention
Filed of the invention
The invention is directed generally to medical diagnostics and specifically to diagnostic screens for primary liver cancer.
Brief Description of the Related Art
Hepatocellular carcinoma (HCC) is the fifth most common cancer in the world today, and the overall 5-year survival rate remains less than 5% (1). Although the incidence rate will likely fall with the institution of mass vaccination against the hepatitis B virus (2), major impacts will not be felt immediately as the age of presentation in most areas of the world is over 50 years long. More importantly, there is no immediate prospect of a vaccine against the hepatitis C virus on the market, the major aetiological factor for HCC in the US, Japan and Southern Europe (3, 4). Despite the absence of randomized clinical trials, there is strong evidence that surgical resection, liver transplantation or ablative therapies significantly improve survival (5, 6). Such approaches, however, are only applicable to those in whom the tumor is detected at an early stage — typically less than 3 cm in diameter without vascular involvement -- and tumors at these stages only rarely present symptoms (5-7). Early diagnosis has therefore become a priority. Surveillance of those with cirrhosis has been aided in the detection of small tumors, and there is emerging evidence that this early detection is correlated to improved survival rates (8). Currently, surveillance usually occurs at intervals of 6 month and involves both testing with serum alpha-fetoprotein (AFP) estimation and ultrasound scanning (7, 9). Although AFP may be a useful diagnostic serum marker in patients with advanced symptomatic disease, the technique's low sensitivity has rendered it much less useful in patients with early stage or small tumors (7-10). While ultrasound is far more sensitive (in the order of 80%), it is highly operator dependent (8, 9). More specific and sensitive serological tests, to complement ultrasound scanning, would therefore be of great clinical value.
A number of novel candidate markers have recently been introduced. Their discovery has been based on the identification of single proteins, complex proteomics features, or tumor-specific autoantibodies. The excitement about new candidate markers is tempered by the realization that none of them have yet met the most stringent criteria defined by the Early Detection Research Network (EDRN). Studies meeting the above criteria were systematically evaluated according to the guidelines established by EDRN (11). The guidelines are based on a 5-step process that reflects an increasing burden of proof, beginning with preclinical exploration of markers (11), the development and validation of a noninvasive clinical assay (12), a retrospective longitudinal testing of repository materials (13), the prospective screening of populations at risk (14), and studies to determine whether screening reduces the burden of cancer in the population (15). Markers meeting the latter goals would be considered the most valuable. Part of the results are shown in Table 1.
Table 1: E f HCC candidate serum markers
Figure imgf000004_0001
commerciablly available and FDA-approved.
2 available in Japan and other Asian countries.
Plasma/serum proteomics: Traditionally, serum proteomics studies have been based on two-dimensional gel electrophoresis, though this method does not appear to be sufficiently sensitive for the detection of low-abundance marker proteins. A number of such studies have been performed in HCC patients. Steel and coworkers performed 2-dimensional gel electrophoresis of sera from HBV-infected patients with and without HCC (16). Two protein signals were consistently less abundant in HCC patients as compared to HBV-negative controls and HBV-carriers (17). They were identified by mass spectroscopy as fragments of C3 complement and apolipoprotein A1, respectively. However, similar decreases in the serum levels of the two proteins were also observed in HCC-negative patients with chronic hepatitis, suggesting that the observed changes were not cancer-specific. A recent study by the same group suggests that a decrease in a serum pre-amyloid P fragment may correlate with HCC (18). Similar reports based on this method have been published by several authors. It is doubtful that subtle changes in the levels of highly abundant proteins will be sufficiently sensitive and specific for diagnostic purposes.
More recently, surface-enhanced laser desorption/ionization - time of flight spectrometry (SELDI-TOF) has been advocated as a superior method for the identification of unique serum protein fragments. Poon and colleagues used this approach to study 38 HCC patients and 20 control patients with chronic hepatitis (19). The patients' sera were processed by anion-exchange fractionation, followed by absorption to protein chip arrays, and time-of-flight mass spectrometry. The data were analyzed using mathematical algorithms previously validated for gene array data, including SAM ("significant analysis of microarray") filtering. Using artificial neural networks, the authors identified 250 differential "serum proteomic features" which reportedly allowed the detection of HCC with sensitivity and specificity in the 90% range. This level of performance exceeds that of any known single serum marker reported to date. Since the publication of this study, four groups have reported their findings using SELDI- TOF-based analyses: Ward and colleagues reported their data obtained on 182 patients with HCV-related liver disease. Their neural network approach resulted in a set of protein peaks with 94% sensitivity and 86% specificity (20). Of note, the authors identified the two most informative SELDI peaks, and determined that they represented k and I immunoglobulin light chains. This result was confirmed by Western blotting, and indicated a modest (approximately 50%) increase. This study was noteworthy for its careful documentation of the reproducibility of the analyses and description of the SELDI methods.
In a study of Chinese patients with poorly characterized chronic liver disease and HCC, Wang and colleagues reported two sets of distinct SELDI peaks that allowed differentiation between HCC and normal subjects or patients with cirrhosis, respectively (21). The identity of the four protein peaks was not determined, however, and the sensitivity and specificity of the informative peaks were not fully reported. Drake and colleagues analyzed SELDI-TOF data alone or in combination with measurements of AFP, DCP, and GP73 in 170 patients with HCV-related liver disease. They identified 38 proteomic "features" that distinguished cirrhotics with and without cancer with a sensitivity and specificity of 61% and 76%, respectively. Combining the proteomics features with the serum levels of AFP1 DCP, and GP73 improved the sensitivity and specificity to 75% and 92%, respectively (22). These results are less impressive than those obtained by Poon, but still represent a significant improvement over single marker measurements. Paradis and colleagues studied 82 French patients with liver cirrhosis from a variety of causes, and identified six SELDI peaks that distinguished HCC and non-HCC patients in 90% of the cases (23). The most informative protein peak was identified as a C-terminal vitronectin fragment. The authors provided in vitro evidence suggesting that this fragment may be generated by cleavage of the intact molecule by a metalloprotease activity, suggesting a biologically plausible mechanism for the appearance of the fragment in serum. Of note, the hepatic expression of vitronectin was not increased in HCC, supporting the notion that the serum peak was not simply due to a transcriptional upregulation of its expression. This study demonstrates the potential of the proteomics approach to reveal candidate markers that would be missed by tissue proteomics or gene array analysis.
The quality of serum SELDI data has recently come into question. Drawbacks of the method include the limited dynamic range of measurable protein fragments, the lack of identification of protein fragments of interest, and its unproven reproducibility (24, 25). It is highly likely that the improved accuracy of immunoassays over SELDI 'quantitation1 could improve the performance of biomarkers originally identified by SELDI. The use of multiple carefully chosen markers should enhance both the sensitivity and specificity of HCC screening.
Serum qlvcoproteomics: Cancer-specific changes in the glycosylation of serum proteins have been previously reported in several types of cancer. Mehta and colleagues recently reported significant increases in the core- fucosylation of several serum proteins in woodchucks with HBV-related HCC, and introduced lectin affinity purification methods to identify and quantitate such proteins (26). The authors subsequently extended their analysis to patients with HBV-related liver disease, and documented similar changes in a number of highly-abundant proteins (including hemopexin, alpha-acid glycoprotein, alpha-1- antichymotrypsin, alpha-1 -antitrypsin, and haptoglobin) as well as AFP and GP73, two previously described HCC markers. Strikingly, the differences in fucosylation were observed even when the overall abundance of the marker proteins were similar in cancer patients and controls (26). Another interesting feature of these studies was that the HCC-specific fucosylation changes were observed in patients with normal and increased AFP levels, suggesting that the two marker categories might be complementary.
Tissue proteomics: Proteomics studies on liver tissues have traditionally utilized a combination of two-dimensional gel electrophoresis and mass spectrometry analysis. Li and colleagues performed a comprehensive liver proteomics analysis of patients with HBV-related HCC, comparing the profiles of cancer tissue with that of surrounding non-cancerous tissue (27). They identified 80 proteins with differential expression in HCC (43 upregulated in HCC, 37 downregulated) by peptide mass spectrometry, and assigned them to several functional categories. A few candidates, including proliferating cell antigen and stathmin 1, were confirmed by Western blotting analysis. Um identified 21 proteins with significant changes in expression in cirrhotic livers with HCC (28). Several previously identified candidate markers were upregulated, including the heat shock protein HSP70RY and nucleophosmin, whereas protein disulfide isomerase (PDI) levels were reduced. In a similar study, Kim identified 12 consistently up- and 2 downregulated proteins in HCC as compared to adjacent non-cancerous tissues (29), and confirmed the increase in nucleophosmin serum levels previously described by Lim. In contrast, a similar study by Kim identified the A3 isoform of PDI as consistently upregulated in HCC (30). The reason for the discrepancies between published data are unclear, although variability in serum composition is likely to account for at least some of the differences. The sensitivity and specificity of the reported alterations in tissue protein profiles have not been formally evaluated, and it remains to be seen whether the tissue data can be exploited for the purpose of serum diagnostics. More recently, MeIIe and coworkers performed laser capture microdissection studies of hepatocellular cancer cells and adjacent, noncancerous hepatocytes (31), and identified proteins with different abundance by proteomics analysis. The only distinguishing peak was identified as hemoglobin, most likely a reflection of the hypovascularity of hepatocellular cancers as compared to surrounding, cirrhotic tissues. No other candidate peaks were identified. The unimpressive results of this study, despite its use of the most sophisticated cell isolation and proteomics techniques, highlight the potential pitfalls of HCC tissue proteomics.
HCC-specific auto-antibodies: An extensive body of literature has documented the presence of tumor-derived autoantibodies (TAA) in the sera of HCC patients. They include some of the earliest TAAs, such as anti-p53 (32), as well as newer members (such as anti-telomerase (33)). In some cases, TAAs may be markers of the malignant transformation, since their occurrence appears to be triggered by the abnormal processing of antigenic cellular proteins during the process of hepatic carcinogenesis (34). Evidence for such a sequence of events has recently been presented for P62 (nucleophosmin), a nuclear protein that had previously been found to be present at elevated levels in the serum of HCC patients. Increased cellular nucleophosmin expression and abnormal protein processing may trigger the formation of a cell- or antibody-mediated autoimmune response (35). A common feature of HCC-TAAs is their low apparent sensitivity - positive serum reactivities are typically found in less than one fifth of all cases (36). Furthermore, the reported sensitivities were typically obtained in comparing HCC patients and healthy subjects, whereas there are few studies on cirrhotic patients who represent a more relevant target population. On the other hand, current data suggest that the "cancer specificity" of certain TAAs may be quite high (approaching 90% in some cases) - a feature that would compare favorably to many of the conventional serum markers (37). One potential limitation may be the lack of liver- specificity, since the same TAAs may occur in different malignancies. At present, none of the individual TAAs has been subjected to a rigorous prospective analysis in HCC cohorts (see table 1).
The following references are cited parenthetically as numbers throughout this disclosure. The references are incorporated herein by reference. Applicant reserves the right to challenge the veracity of any statements made therein.
1. Parkin DM, Bray F, Ferlay J, Pisani P (2001) Estimating the world cancer burden: Globocan 2000. lnt J Cancer 94: 153-156
2. Chang MH1 Chen CJ, Lai MS, Hsu HM, Wu TC1 Kong MS, Liang DC, Shau WY, Chen CS (1997) Universal hepatitis B vaccination in Taiwan and the incidence of hepatocellular carcinoma in children. Taiwan childhood hepatoma study group. N Engl J Med 336: 1855-1859 3. Di Bisceglie AM, Order SE, Klein JL, Waggoner JG, Sjogren MH1 Kuo G, Houghton M, Choo Q-L, Hoofnagle JH (1991) The role of chronic viral hepatitis in hepatocellular carcinoma in the United States. Am J
Gastroenterol 86: 335-338
4. El-Sarag HB, Hashem B (2004) Hepatocellular carcinoma: an epidemiologic view. J Clin Gastroenterol 35: S72-S78
5. Bruix J, Sherman M, Llovet JM, Beaugrand M, Lencioni R, Burroughs AK, Christensen E, Pagliaro L, Colombo M, Rodes J (2001) EASL panel of experts on HCC. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. J Hepatol 35: 421-430
6. Beaugrand M, N'kontchou G, Seror O, Ganne N, Trinchet J-C (2005) Local regional and systemic treatments of hepatocellular carcinoma. Semin Liver Dis 205: 201-211
7. Mazzaferro V, Regalia E, Doci R, Andreola S1 Pulvirenti A (1996) Liver transplantation for treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med 334: 693-699
8. Sherman M (2005) Screening for hepatocellular carcinoma. Best Practice Res Clin Gastroenterol 19:101- 118
9. Trevisani F, De NS1 Rappaccini G, Farinati F, Benvegnu L, ZoIi M, Grazi GL, Del PP, Di N, Bernardi M (2002) Semiannual and annual surveillance of cirrhotic patients for hepatocellular carcinoma: effects on cancer stage and patient survival (Italian experience). Am J Gastroenterol 97: 734-744
10. Johnson PJ (2001) The role of serum alpha-fetoprotein estimation in the diagnosis and management of hepatocellular carcinoma. In: Clinics in Liver Disease, Di Bisceglie A (ed) Vol. 5, pp 145-159. Philadelphia: WB Saunders.
11. Sullivan Pepe M, Etzioni R, Feng Z, Potter JD1 Thompson ML, Thornquist M, Winget M, et al. Phases of biomarker development for early detection of cancer. J Natl Cancer Inst 2001 ;93: 1054-1061.
12. Law SW1 Dugaiczyk A. Homology between the primary structure of alpha-fetoprotein, deduced from a complete cDNA sequence, and serum albumin. Nature 1981;291:201-205.
13. Peters EH, Nishi S1 Miura K, Lorscheider FL, Dixon GH, Tamaoki T. In vitro synthesis of murine pre-alpha-fetoprotein. Cancer Res 1979;39:3702- 3706. 14. Abelev Gl, Eraiser TL. Cellular aspects of alpha-fetoprotein reexpression in tumors. Semin Cancer Biol 1999;9:95-107.
15. Torres JM, Darracq N, Uriel J. Membrane proteins from lymphoblastoid cells showing cross-affinity for alpha-fetoprotein and albumin. Isolation and characterization. Biochim Biophys Acta 1992; 1159:60-66.
16. Chignard N, Beretta L. Proteomics for hepatocellular carcinoma marker discovery. Gastroenterology 2004; 127:S120-125.
17. Steel LF, Shumpert D, Trotter M1 Seeholzer SH, Evans AA, London WT1 Dwek R, et al. A strategy for the comparative analysis of serum proteomes for the discovery of biomarkers for hepatocellular carcinoma. Proteomics 2003:3:601-609.
18. Comunale MA, Mattu TS, Lowman MA, Evans AA, London WT1 Semmes OJ, Ward M, et al. Comparative proteomic analysis of de-N- glycosylated serum from hepatitis B carriers reveals polypeptides that correlate with disease status. Proteomics 2004;4:826-838.
19. Poon TC, Yip TT, Chan AT, Yip C, Yip V1 Mok TS, Lee CC, et al. Comprehensive proteomic profiling identifies serum proteomic signatures for detection of hepatocellular carcinoma and its subtypes. Clin Chem 2003;49:752- 760.
20. Ward DG, Cheng Y, N'Kontchou G1 Thar TT, Barget N1 Wei W, Billingham LJ, et al. Changes in the serum proteome associated with the development of hepatocellular carcinoma in hepatitis C-related cirrhosis. Br J Cancer 2006;94:287-292.
21. Wang JX, Zhang B, Yu JK, Liu J, Yang MQ, Zheng S. Application of serum protein fingerprinting coupled with artificial neural network model in diagnosis of hepatocellular carcinoma. Chin Med J (Engl) 2005; 118: 1278-1284.
22. Schwegler EE, Cazares L1 Steel LF, Adam BL, Johnson DA, Semmes OJ, Block TM, et al. SELDI-TOF MS profiling of serum for detection of the progression of chronic hepatitis C to hepatocellular carcinoma. Hepatology 2005;41 :634-642.
23. Paradis V, Degos F, Dargere D, Pham N1 Belghiti J1 Degott C, Janeau JL, et al. Identification of a new marker of hepatocellular carcinoma by serum protein profiling of patients with chronic liver diseases. Hepatology 2005;41:40-47. 24. Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. MoI Cell Proteomics 2002; 1:845-867.
25. Diamandis EP. Mass spectrometry as a diagnostic and a cancer biomarker discovery tool: opportunities and potential limitations. MoI Cell Proteomics 2004; 3: 367-378.
26. Comunale MA, Lowman M, Long RE1 Krakover J, Philip R, Seeholzer S, Evans AA, et al. Proteomic analysis of serum associated fucosylated glycoproteins in the development of primary hepatocellular carcinoma. J Proteome Res 2006;5:308-315.
27. Li C, Tan YX1 Zhou H, Ding SJ, Li SJ, Ma DJ, Man XB, et al. Proteomic analysis of hepatitis B virus-associated hepatocellular carcinoma: Identification of potential tumor markers. Proteomics 2005;5:1125-1139.
28. Lim SO, Park SJ, Kim W1 Park SG, Kim HJ, Kim Yl, Sohn TS1 et al. Proteome analysis of hepatocellular carcinoma. Biochem Biophys Res Commun 2002;291:1031-1037.
29. Kim W, Oe Lim S1 Kim JS, Ryu YH, Byeon JY, Kim HJ1 Kim Yl, et al. Comparison of proteome between hepatitis B virus- and hepatitis C virus- associated hepatocellular carcinoma. Clin Cancer Res 2003;9:5493-5500.
30. Kim J1 Kim SH, Lee SU1 Ha GH, Kang DG1 Ha NY1 Ahn JS1 et al. Proteome analysis of human liver tumor tissue by two-dimensional gel electrophoresis and matrix assisted laser desorption/ionization-mass spectrometry for identification of disease-related proteins. Electrophoresis 2002;23:4142-4156.
31 MeIIe C1 Kaufmann R, Hommann M, Bleul A1 Driesch D1 Ernst G1 von Eggeling F. Proteomic profiling in microdissected hepatocellular carcinoma tissue using ProteinChip technology, lnt J Oncol 2004;24:885-891.
32. Soussi T. p53 Antibodies in the sera of patients with various types of cancer: a review. Cancer Res 2000;60: 1777-1788.
33. Masutomi K1 Kaneko S, Yasukawa M, Arai K, Murakami S, Kobayashi K. Identification of serum anti-human telomerase reverse transcriptase (hTERT) auto-antibodies during progression to hepatocellular carcinoma. Oncogene 2002;21:5946-5950.
34. Zhang JY1 Zhu W, lmai H, Kiyosawa K, Chan EK, Tan EM. De- novo humoral immune responses to cancer-associated autoantigens during transition from chronic liver disease to hepatocellular carcinoma. Clin Exp Immunol 2001 ;125:3-9.
35. Ulanet DB, Torbenson M, Dang CV1 Casciola-Rosen L1 Rosen A. Unique conformation of cancer autoantigen B23 in hepatoma: a mechanism for specificity in the autoimmune response. Proc Natl Acad Sci U S A 2003;100:12361-12366.
36. Wang Y, Han KJ1 Pang XW, Vaughan HA, Qu W, Dong XY, Peng JR, et al. Large scale identification of human hepatocellular carcinoma- associated antigens by autoantibodies. J Immunol 2002;169:1102-1109.
37. Raedle J, Oremek G, Truschnowitsch M, Lorenz M, Roth WK1 Caspary WF, Zeuzem S. Clinical evaluation of autoantibodies to p53 protein in patients with chronic liver disease and hepatocellular carcinoma. Eur J Cancer 1998:34:1198-1203.
38. Roskoski, R. 2005. Signaling by kit protein-tyrosine kinase-the stem cell factor receptor. BBRC 337:1-13.
39. Akin C, Metcalfe DD. 2004. The biology of kit in disease and the application of pharmacogenetics. J Allergy Clin Immunol 114:13-19.
40. Sette C, Dolci S, Geremia R, Rossi P. 2000. The role of stem cell factor and of alternative c-kit gene products in the establishment, maintenance and function of germ cells, lnt J Dev Biol 44:599-608.
41. Tian YW, Smith PG, Yeoh GC. The oval-shaped cell as a candidate for a liver stem cell in embryonic, neonatal and precancerous liver: identification based on morphology and immunohistochemical staining for albumin and pyruvate kinase isoenzyme expression. Histochem Cell Biol. 1997, 107:243-250.
42. Tee LB1 Kirilak Y, Huang WH, et al. Differentiation of oval cells into duct-like cells in preneoplastic liver of rats placed on a choline-deficient diet supplemented with ethionine. Carcinogenesis. 1994, 15:2747-2756.
43. Rosenberg D1 Hie Z1 Yin L, et al. Proliferation of hepatic lineage cells of normal C57BL and interleukin-6 knockout mice after cocaine-induced periportal injury. Hepatology. 2000, 31:948-955.
44. Overturf K, al-Dhalimy M1 Ou CN1 et al. Serial transplantation reveals the stem cell-like regenerative potential of adult mouse hepatocytes. Am J Pathol. 1997, 151:1273-1280. 45. Gordon GJ, Coleman WB, Grisham JW. Temporal analysis of hepatocyte differentiation by small hepatocyte-like progenitor cells during liver regeneration in retrorsine exposed rats. Am J Pathol. 2000, 157:771 -786.
46. Alison M, Sarraf C. Hepatic stem cells. J Hepatol. 1998, 29:676- 682.
47. Slack JMW. Stem cells in epithelial tissues. Science. 2000, 287:1431-1433.
48. Petersen BE, Zajac VF, Michalopoulos GK. Bile ductular damage induced by methylene diamine inhibits oval cell activation. Am J Pathol. 1997, 151:905-909.
49. Hsia CC, Evarts RP, Nakatsukasa H, et al. Occurrence of ovaltype cells in hepatitis B virus-associated human hepatocarcinogenesis. Hepatology. 1992, 16:1327-1333.
50. Yamaguchi.M., Nakazawa, T., Hiroki Kuyama, H., Obama, T., Ando E., Okamura, T., Ueyama, N., and Norioka, S. High-Throughput Method for N- Terminal Sequencing of Proteins by MALDI Mass Spectrometry Anal. Chem., 2005, 77:645-651.
51. Sechi, S and Chait, BT. A Method To Define the Carboxyl Terminal of Proteins. Anal. Chem., 2000, 72:3374-3378.
52. Miyazaki, K., Tsugita, A. C-terminal sequencing method for proteins in polyacrylamide gel by the reaction of acetic anhydride Proteomics 2006, 6:2026-2033.
53. Rail, S. C, Jr., Weisgraber, K. H., Innerarity, T. L., and Mahley, R. W. (1982) Structural basis for receptor binding heterogeneity of apolipoprotein E from type III hyperlipoproteinemic subjects, Proc. Natl. Acad. Sci. U.S.A. 79, 4696-4700.
54. Wilson, C, Wardell, M. R., Weisgraber, K. H., Mahley, R. W., and Agard, D. A. (1991) Three-dimensional structure of the LDL receptor-binding domain of human apolipoprotein E, Science 252, 1817-1822.
55. Segrest, J. P., Jones, M. K., De Loof, H., Brouillette, C. G., Venkatachalapathi, Y. V., and Anantharamaiah, G. M. (1992) The amphipathic helix in the exchangeable apolipoproteins: a review of secondary structure and function, J. Lipid Res. 33, 141-166. 56. Wernette-Hammond, M. E., Lauer, S. J., Corsini, A., Walker, D., Taylor, J. M., and Rail, S. C, Jr. (1989) Glycosylation of human apolipoprotein E. The carbohydrate attachment site is threonine 194, J. Biol. Chem. 264, 9094- 9101.
57. Cardin, A. D., Hirose, N., Blankenship, D. T., Jackson, R. L1 Harmony, J. A., Sparrow, D. A., and Sparrow, J. T. (1986) Binding of a high reactive heparin to human apolipoprotein E: identification of two heparin-binding domains, Biochem. Biophys. Res. Commun. 134, 783-789.
58. Mahley, R. W. (1988) Apolipoprotein E: cholesterol transport protein with expanding role in cell biology, Science 240, 622-630.
59. Choy, N., Raussens, V., and Narayanaswami, V. (2003) lntermolecular coiled-coil formation in human apolipoprotein E C-terminal domain, J. MoI. Biol. 334, 527-539.
60. Fan, D., Li1 Q., Korando, L., Jerome, W. G., and Wang, J. (2004) A monomeric human apolipoprotein E carboxyl-terminal domain, Biochemistry 43, 5055-5064.
61. Snipes, G. J., McGuire, C. B., Norden, J. J., and Freeman, J. A. (1986) Nerve injury stimulates the secretion of apolipoprotein E by nonneuronal cells, Proc. Natl. Acad. Sci. U.S.A. 83, 1130-1134.
62. A new prognostic system for hepatocellular carcinoma: a retrospective study of 435 patients: the Cancer of the Liver Italian Program (CLIP) investigators. Hepatology 1998;28:751-755.
63. Omenn GS, States DJ, Adamski M, Blackwell TW, Menon R1 Hermjakob H, Apweiler R, et al. Overview of the HUPO Plasma Proteome Project: results from the pilot phase with 35 collaborating laboratories and multiple analytical groups, generating a core dataset of 3020 proteins and a publicly-available database. Proteomics 2005;5:3226-3245.
64. Lin SM, Kibbe WA. Irrational exuberance in clinical proteomics. Clin Cancer Res 2005; 11.7963-7964.
65. Ransohoff DF. Bias as a threat to the validity of cancer molecular- marker research. Nat Rev Cancer 2005; 5: 142-149.
66. Baggerly KA1 Morris JS1 Edmonson SR1 Coombes KR. Signal in noise: evaluating reported reproducibility of serum proteomic tests for ovarian cancer. J Natl Cancer Inst 2005;97:307-309. 67. Molloy MP1 Brzezinski EE, Hang J1 McDowell MT, VanBogelen RA. Overcoming technical variation and biological variation in quantitative proteomics. Proteomics 2003;3:1912-1919.
68. Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. MoI Cell Proteomics 2002; 1:845-867.
69. Rai AJ, Gelfand CA, Haywood BC, Warunek DJ1 Yi J, Schuchard MD1 Mehigh RJ1 et al. HUPO Plasma Proteome Project specimen collection and handling: towards the standardization of parameters for plasma proteome samples. Proteomics 2005;5:3262-3277.
Summary of the Invention
In view of the need for alternative methods for the diagnosis and/or prediction of risk of hepatocellular carcinoma (HCC), the present inventors have developed methods and kits .
In various embodiments, the present teachings include methods for diagnosing hepatocellular carcinoma in a subject. In various aspects, these methods include a) obtaining a serum sample from a subject, and b) quantifying, in the serum sample, at least one protein that is differentially upregulated in patients diagnosed with HCC. A subject can be diagnosed with HCC if the quantity of at least one of the proteins is determined to be elevated relative to control levels. Examples of serum proteins that can be differentially upregulated in a subject with HCC can be, without limitation, apolipoprotein E1 Trypsin precursor, KIAA0284, Apolipoprotein A-I precursor, v-kit (c-kit homologue), c-kit and Apolipoprotein C-III precursor. In various aspects of the present teachings, an assay for measuring the amount of a protein which is upregulated in HCC can be an aptamer-based assay, an antibody-based assay, a mass spectroscopy assay, or a high performance liquid chromatography assay. In some configurations, an antibody-based assay can be an ELISA, a radioimmunoassay or a Western blot.
In some aspects or the present teachings, the methods can further include removing at least one high-abundance serum protein from the serum sample prior to analyzing a serum sample for the quantity of a protein diagnostic for HCC. Such high-abundance proteins include albumin, IgG1 antitrypsin, IgA1 transferrin and haptoglobin. In some aspects, all six of these proteins can be removed from the serum sample prior to the analysis.
In various embodiments of the present teachings, the inventors disclose kits for diagnosing hepatocellular carcinoma. A kit of these embodiments can include, without limitation, a capture antibody directed against a first epitope of an HCC serum biomarker, a solid support upon which the capture antibody is immobilized, a detection antibody directed against a second epitope of the HCC serum biomarker; and a labeled secondary antibody directed against the detection antibody. In some aspects, the solid support can comprise at least one plastic bead or a multiwell plate such as an ELISA plate well known to skilled artisans. In various aspects, an HCC serum biomarker can be, without limitation, a protein such as an apolipoprotein E1 a Trypsin precursor, a KIAA0284, a Apolipoprotein A-I precursor, a v-kit (c-kit homologue), a c-kit or an Apolipoprotein C-III precursor. In some additional aspects, a labeled secondary antibody comprised by a kit can include a label such as a fluorophore, an enzyme or a radioisotope.
Brief Description of the Drawings
Figure 1 illustrates the 2D gel image of c-kit protein from a cirrhosis serum sample and an HCC serum sample.
Figure 2 illustrates the 2D gel image of Apo E protein from a cirrhosis serum sample and an HCC serum sample.
Figure 3 illustrates a Western blot analysis of serum samples using anti-kit antibodies.
Figure 4 illustrates a Western blot analysis of serum samples using anti-ApoE antibodies
Figure 5 illustrates fractionation of serum proteins using the PF-2D LC system. Detailed Description
Early diagnosis of hepatocellular carcinoma (HCC) is the key to the delivery of effective therapies. There is a long felt need to identify novel serum markers of hepatocellular cancer (HCC). Applicants have established serum banks from patients with hepatitis C virus infection and HCV-related HCC at Saint Louis University Medical Center and the Saint Louis VA. In a recent pilot study using agilent affinity-column to remove 6 abundant proteins followed by 2 dimensional gel electrophoresis and mass spectroscopy (MS), applicants identified several new candidate serum biomarkers.
The inventors made the surprising discovery that various proteins are differentially found in the serum of patients at risk for or having HCC. Those proteins and their exemplary GenBank accession numbers are listed in Table 2. Thus, in one embodiment, the invention is directed to a method for determining the risk of a patient having or acquiring hepatocellular carcinoma ("HCC"), the steps comprising obtaining a serum sample from a patient, detecting and/or quantifying any one or more of the biomarkers listed in Table 2, alone or in combination with one or more known HCC serum biomarkers, and determining the patient's risk for having or acquiring HCC. The detecting step may be any one of myriad well-known methods in the art, including for example aptamer- based assays, antibody-based assays (e.g., ELISA, RIA, western), mass spec, HPLC, and the like. Preferably, the detection method is an antibody-based ELISA, as is currently the preference in the medical diagnostics art. The preferred determining step is quantifying the signal generated by the detection of the one of more the biomarkers of Table 2 in the test sample (e.g., serum of an individual suspected of having risk factors for HCC) and comparing that signal quantity to a control sample. A difference between the test and control values for the biomarkers can be interpreted to assess whether the test individual is at risk for HCC and/or needs additional testing and/or therapy.
In another embodiment, the invention is directed to a diagnostic kit for determining the risk of an individual for having or getting HCC. The preferred kit is based upon the ELISA immunodetection method, but the skilled artisan in the practice of this invention would recognize that other methods to which the kit can be based upon are available and applicable. In the ELISA-based kit, the kit comprises a substrate to which a capture antibody is fixed, a detection antibody, and a detectable moiety antibody. The preferred substrate for example may be a plastic bead or surface of a multiwell plate. The preferred capture antibody recognizes a first epitope on a HCC serum biomarker. The preferred detection antibody recognizes a second epitope on the HCC serum biomarker. The preferred detectable moiety antibody recognizes the detection antibody and has a detection moiety such as for example a fluorophore, chemiluminescence molecule, enzyme, or the like.
Table 2: Proteins Differentially Found in Serum of HCC Patients
Figure imgf000018_0001
Examples
The methods described herein utilize laboratory techniques well known to skilled artisans, and guidance can be found in laboratory manuals such as Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2001; Spector, D. L. et al., Cells: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY1 1998; and Harlow, E., Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY1 1999.
Example 1. Identification of Biomaker candidates using 2-D gel approaches followed by MS analysis
Patients with HCV-related chronic liver disease were enrolled at the Johns Cochran VA Hospital and Saint Louis University Health Sciences Center. For the initial pilot study using two-dimensional gel electrophoresis methods, two groups of male patients with proven, compensated liver cirrhosis were studied, one without evidence of hepatocellular cancer, the other with biopsy-proven small HCC (<3 cm) that were discovered by surveillance imaging studies. Serum samples were obtained from patients after an overnight fast. None of the patients were receiving treatment for their HCC at the time of the blood draw. The clinical characteristics of the two groups are provided in Table 3.
Table 3: Clinical Characteristics
Figure imgf000019_0001
Patients in the two groups were comparable with regard to their age, MELD score, and serum AFP values. Of note, none of the HCC patients had AFP levels above the upper limit of normal (20mg/L), the most commonly used cutoff of HCC screening studies (12).
To analyze the low-abundant serum proteins, the Agilent spin cartridge was used to remove six high-abundant proteins from the serum pooled from three patients. These six proteins include albumin, IgG, antitrypsin, IgA, transferrin, and haptoglobin according to manufacturer's procedures. Specific removal of six high- abundant proteins can deplete approximately 85%-88% of total protein mass from human serum.
To remove the six abundant serum proteins, we first diluted 10 μL of pooled serum with Buffer A to a final volume of 200 μL and load it onto the spin cartridge that has been equilibrated with 4 ml of Buffer A into syringe. We then centrifuge for 1.5 minutes at 100xg to collect the flow through fraction (F1), followed by washing the column with 400 μL of Buffer A by spin for 2.5 minutes at 100xg. The flow-through fraction was collected into the same F1 collection tube. This procedure was repeated once and this fraction was collected into the F2 collection tube. Next, we used 2 ml of Buffer B to elute the bound abundant proteins. The column was re-equilibrated with Buffer A and the procedure was repeated with pooled serum samples. Each fraction was then analyzed separately by 1D SDS-PAGE to ensure the reproducibility of the depletion procedure. After the confirmation, all F1 and F2 fractions obtained from the same pooled sera were pooled and concentrated by 5KDa MWCO spin concentrators. The buffer was exchanged to 0.1 M ammonium bicarbonate separated into two aliquots and lyophilized. Protein pellet from one aliquot was re-suspended in PBS and the protein concentration was determined with Bradford method using BSA as standards.
An important consideration in the measurement of quantitative changes in protein expression is the consistency of the observations for a given technique as well as the reproducibility of the experiment. Several techniques are available for measuring changes in protein expression including gel-based approaches and shotgun proteomic methods. High-resolution 2-DE based separation of protein mixtures followed by image analysis for relative protein quantitation and mass spectrometric analysis for identification is a hallmark method in proteomics [1 ,2]. One of the benefits of the 2-DE approach is that the same separation technology is used both to measure protein expression changes as well as to purify proteins for subsequent MS analysis. The development of pre-and post separation fluorescent stains has facilitated reliable measures of changes in spot intensity [3,4 ].
We used 200 mg of each sample for the 2D gel analysis. Protein pellets were resuspended in a mixture containing 80 ul of 8M urea, 4% CHAPS, 65 mM DTT in 66.7 mM Tris and 300 ul of 8M urea, 4%CHAPS,65 mM DTT,1% ampholytes for in-gel rehydration. IEF and SDS-PAGE were performed by standard procedures. IEF was performed on 18 cm, 3-10 nonlinear IPG strips (GE Amersham Biosciences, Piscataway, NJ1 USA) using the following program: 1 250V 20 min linear, 2 8000V 2.5 h linear and 3 8000V to 20,000V Rapid. SDS- PAGE was performed on a 12%T slab gels with an agarose overlay. Gels were fixed, stained with SYPRO Ruby (Molecular Probes, Eugene, OR1USA) and destained according to the manufacturer's protocol.
Gel images are acquired with a Typhoon 9410 imager (Amersham Biosciences). Gel images are then analyzed using Phoretix 2D Evolution gel analysis software (Nonlinear Dynamics Ltd.). Automatic spot detection was performed with default parameters; spot editing and removal of artifacts were performed manually. Subsequent data analyses were performed on the MS- identified spots and manually matched among the gels. The normalized spot volume relative to the total volume of all detected spots in a gel (%vol) was used to calculate averages and CV between gels of replicate samples for individual spots. Microsoft Excel's STDEV function was used to calculate the SD for a given spot. A propagation of errors calculation was used to calculate the SD of expression ratios, which was subsequently used to calculate a CV for expression ratios.
Following image analysis, interesting proteins were excised from gels by GelPix (Genetix Inc.) and digested by an automated digester with trypsin, the MultiProbe Il workstation (PerkinElmer Inc.). Digests were de-salted and concentrated with C18 ZipTips (Millipore, Bedford, MA, USA) following the manufacturer's instructions and spotted onto a MALDI target plate and analyzed by a QSTAR XL mass spectrometer (Applied Biosystems). MS peak lists were submitted for automatic precursor ion selection excluding common trypsin autolysis peaks. Up to four of the most intense peaks were selected from each MS spectrum for MS/MS The acquired data are automatically submitted to database searching and protein identification by MASCOT v1.9 database searching engine. Searches were restricted to 50 ppm MS mass tolerance, and 0.3 Da MS/MS mass tolerance. Search results were ranked by protein score confidence interval% (Cl%), a GPS calculation based on Mascot score with the significance threshold removed to normalize results across different databases. Protein hits with protein score Cl% between 95 and 100 were accepted for identifications.
By comparing the 2D gel image of the sirrororis serum sample and that of the HCC serum sample, we found 30 down regulated protein spots and 7 up- regulated protein spots in HCC serum. The seven protein spots were analyzed as described above and the proteins were identified. Among these seven proteins upregulated in HCC serum samples, one is c-kit (Figure 1, 3h) and two are ApoE spots (Figure 2 1h and 2h). Example 2. Verification of the first candidate, c-kit
To further confirm that the above identified identified proteins (table 1 ) are up-regulated in HCC samples, we performed Western blot analysis of many more serum samples using anti-kit antibodies. The results are shown in Figure 3.
Example 3 Verification of a second candidate, phosphorylated ApoE
Since two spots are identified as ApoE, it is possible that they are ApoE isoforms or phosphorylated ApoE. To test whether they are phosphorylated ApoE, we first treated the serum samples with the phosphoprotein enrichment kit (Clontech) and performed western blot analysis using anti-ApoE antibodies to find out whether the phorphorylated form of ApoE is upregulated in HCC sera. 15 ul of human serum samples were diluted to 1 ml with buffer A provided in the kit and loaded to the phosphoryprote in affinity column. After washing the bound proteins were eluted with buffer B. Protein concentration in the eluate was determined by Bio-Rad protein assay. 0.5 ug of total protein was used for western blot analysis as shown in Figure 4.
Example 4. Fractionation of serum proteins using PF-2D LC system from Beckman Coulter.
Briefly, serum protein with six abundant proteins depleted as described was used and the buffer was exchanged to the starting buffer provided by the ProteomeLab PF 2D kit. 1 mg of the normal serum protein were resolved using the default method, and proteins were collected by interval of 0.3 pH unit from the first dimension separation using the ProteomeLab PF 2D fractionation system (Beckman Coulter). Twenty fractions were generated and each fraction was further separated by the C18 column with a linear gradient of 0%-100% ACN. The results are analyzed by proteovue program from Beckman Coulter Fractionation of serum proteins using PF-2D LC system from Beckman Coulter as shown in Figure 5.

Claims

ClaimsWhat is claimed is:
1. A method of diagnosing hepatocellular carcinoma in a subject, the method comprising: a) obtaining a serum sample from the subject; b) quantifying, in the serum sample, at least one protein that is differentially upregulated in patients diagnosed with HCC, whereby the subject is diagnosed with HCC if the quantity of at least one of the proteins is elevated relative to control levels.
2. A method of diagnosing hepatocellular carcinoma in accordance with claim 1 , wherein the at least one protein is selected from the group consisting of apolipoprotein E, Trypsin precursor, KIAA0284, Apolipoprotein A-I precursor, v-kit (c- kit homologue), c-kit and Apolipoprotein C-III precursor.
3. A method of diagnosing hepatocellular carcinoma in accordance with claim 1 , wherein the at least one protein is apolipoprotein E.
4. A method of diagnosing hepatocellular carcinoma in accordance with claim 1 , wherein the at least one protein is trypsin precursor.
5. A method of diagnosing hepatocellular carcinoma in accordance with claim 1 , wherein the at least one protein is KIAA0284.
6. A method of diagnosing hepatocellular carcinoma in accordance with claim 1 , wherein the at least one protein is Apolipoprotein A-I precursor.
7. A method of diagnosing hepatocellular carcinoma in accordance with claim 1 , wherein the at least one protein is v-kit (c-kit homologue).
8. A method of diagnosing hepatocellular carcinoma in accordance with claim 1, wherein the at least one protein is c-kit.
9. A method of diagnosing hepatocellular carcinoma in accordance with claim 1, wherein the at least one protein is Apolipoprotein C-IIl precursor.
10. The method according to Claim 1, wherein the quantifying comprises performing an assay selected from the group consisting of an aptamer-based assay, an antibody-based assay, a mass spectroscopy assay, and a high performance liquid chromatography assay.
11. The method according to claim 10, wherein the antibody-based assay is selected from the group consisting of a radioimmunoassay, an ELISA and a Western blot.
12. A method of diagnosing hepatocellular carcinoma in accordance with claim 1, further comprising removing at least one high-abundance serum protein from the serum sample prior to the quantifying.
13. A method of diagnosing hepatocellular carcinoma in accordance with claim 12, wherein the at least one high-abundance serum protein is selected from the group consisting of albumin, IgG, antitrypsin, IgA, transferrin and haptoglobin.
14. A method of diagnosing hepatocellular carcinoma in accordance with claim 12, wherein the removing at least one high-abundance serum protein comprises removing six high-abundance serum proteins.
15. A kit for diagnosing hepatocellular carcinoma, comprising: a capture antibody directed against a first epitope of an HCC serum biomarker; a solid support upon which the capture antibody is immobilized; a detection antibody directed against a second epitope of the HCC serum biomarker; and a labeled secondary antibody directed against the detection antibody.
16. The diagnostic kit of Claim 15 wherein the solid support comprises at least one plastic bead.
17. The diagnostic kit of Claim 15 wherein the solid support comprises a multiwell plate.
18. The diagnostic kit of Claim 15 wherein the hepatocellular carcinoma serum biomarker is selected from the group consisting of apolipoprotein E1 Trypsin precursor, KIAA0284, Apolipoprotein A-I precursor, v-kit (c-kit homologue), c-kit and Apolipoprotein C-III precursor.
19. The diagnostic kit of Claim 15 wherein the labeled secondary antibody comprises a label selected from the group consisting of a fluorophore, an enzyme and a radioisotope.
PCT/US2007/017690 2006-08-09 2007-08-09 Kits and methods for determining risk for primary liver cancer WO2008021163A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US82192106P 2006-08-09 2006-08-09
US60/821,921 2006-08-09

Publications (2)

Publication Number Publication Date
WO2008021163A2 true WO2008021163A2 (en) 2008-02-21
WO2008021163A3 WO2008021163A3 (en) 2009-04-16

Family

ID=39082591

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/017690 WO2008021163A2 (en) 2006-08-09 2007-08-09 Kits and methods for determining risk for primary liver cancer

Country Status (1)

Country Link
WO (1) WO2008021163A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102081100A (en) * 2010-07-20 2011-06-01 李伯安 Liver cancer multi-marker micro-array kit as well as preparation method and application thereof
CN105452865A (en) * 2013-08-06 2016-03-30 金弦起 Composition for diagnosing small hepatocellular carcinoma and hepatocellular carcinoma latent in cirrhotic liver
CN107942055A (en) * 2017-11-22 2018-04-20 南宁科城汇信息科技有限公司 The ELISA method of transcript profile and protein science in a kind of liver cancer biological process
JP2019512091A (en) * 2016-02-26 2019-05-09 ドレクセル ユニバーシティ Early detection of hepatocellular carcinoma

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030190689A1 (en) * 2002-04-05 2003-10-09 Cell Signaling Technology,Inc. Molecular profiling of disease and therapeutic response using phospho-specific antibodies
US20050112711A1 (en) * 2003-09-05 2005-05-26 Thomas Jefferson University Diagnosis and monitoring of hepatocellular carcinoma

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030190689A1 (en) * 2002-04-05 2003-10-09 Cell Signaling Technology,Inc. Molecular profiling of disease and therapeutic response using phospho-specific antibodies
US20050112711A1 (en) * 2003-09-05 2005-05-26 Thomas Jefferson University Diagnosis and monitoring of hepatocellular carcinoma

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JIANG ET AL.: 'Influence of liver cancer on lipid and lipoprotein metabolism.' LIPIDS IN HEALTH AND DISEASE vol. 5, March 2006, pages 1 - 7 *
SEKIDO ET AL.: 'Preferential expression of c-kit protooncogene transcripts in small cell lung cancer.' CANCER RESEARCH vol. 51, May 1991, pages 2416 - 2419 *
YOKOYAMA ET AL.: 'Protein level of apolipoprotein E increased in human hepatocellular carcinoma.' INTERNATIONAL JOURNAL OF ONCOLOGY vol. 28, March 2006, pages 625 - 631 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102081100A (en) * 2010-07-20 2011-06-01 李伯安 Liver cancer multi-marker micro-array kit as well as preparation method and application thereof
CN105452865A (en) * 2013-08-06 2016-03-30 金弦起 Composition for diagnosing small hepatocellular carcinoma and hepatocellular carcinoma latent in cirrhotic liver
JP2019512091A (en) * 2016-02-26 2019-05-09 ドレクセル ユニバーシティ Early detection of hepatocellular carcinoma
EP3419646A4 (en) * 2016-02-26 2019-10-23 Drexel University Early detection of hepatocellular carcinoma
JP7026390B2 (en) 2016-02-26 2022-02-28 ドレクセル ユニバーシティ Early detection of hepatocellular carcinoma
US11408888B2 (en) 2016-02-26 2022-08-09 Drexel University Early detection of hepatocellular carcinoma
CN107942055A (en) * 2017-11-22 2018-04-20 南宁科城汇信息科技有限公司 The ELISA method of transcript profile and protein science in a kind of liver cancer biological process

Also Published As

Publication number Publication date
WO2008021163A3 (en) 2009-04-16

Similar Documents

Publication Publication Date Title
JP4717810B2 (en) Biomarkers for distinguishing between type 1 diabetes and type 2 diabetes
JP6028960B2 (en) Glycan markers for liver disease condition indicators
EP1200832B1 (en) Annexins and autoantibodies used as markers for lung cancer and oesophageal cancer
Zhang et al. Serum proteomics in biomedical research: a systematic review
Tessitore et al. Serum biomarkers identification by mass spectrometry in high-mortality tumors
KR101788414B1 (en) Biomarker for diagnosis of liver cancer and use thereof
WO2012036094A1 (en) Lung cancer identification marker
WO2010141469A2 (en) Protein biomarkers and therapeutic targets for autoimmune and alloimmune diseases
Darebna et al. Changes in the expression of N-and O-glycopeptides in patients with colorectal cancer and hepatocellular carcinoma quantified by full-MS scan FT-ICR and multiple reaction monitoring
Lesur et al. Screening protein isoforms predictive for cancer using immunoaffinity capture and fast LC‐MS in PRM mode
Uto et al. Clinical proteomics for liver disease: a promising approach for discovery of novel biomarkers
Yan et al. Confounding effect of obstructive jaundice in the interpretation of proteomic plasma profiling data for pancreatic cancer
Zhang et al. Proteomic identification of down-regulation of oncoprotein DJ-1 and proteasome activator subunit 1 in hepatitis B virus-infected well-differentiated hepatocellular carcinoma
CN108351359B (en) Method for predicting risk and prognosis of hepatocellular carcinoma in patients with cirrhosis
Ma et al. A precise approach in large scale core-fucosylated glycoprotein identification with low-and high-normalized collision energy
EP2136843A1 (en) Biomarkers of prostate cancer and uses thereof
Zhang et al. Immunoaffinity LC–MS/MS for quantitative determination of a free and total protein target as a target engagement biomarker
WO2008021163A2 (en) Kits and methods for determining risk for primary liver cancer
EP3035058B1 (en) Cancer marker screening method through detection of deglycosylation of glycoprotein and hepatocellular cancer marker
CA2862834A1 (en) Srm/mrm assay for the insulin receptor protein
Liu et al. Integrative oncoproteomics strategies for anticancer drug discovery
Panis et al. How can proteomics reach cancer biomarkers?
Yin et al. Analysis of the peptides detected in atopic dermatitis and various inflammatory diseases patients-derived sera
Li et al. Plasma biomarker screening for liver fibrosis with the N-terminal isotope tagging strategy
Evans et al. Prostate cancer proteomics: the urgent need for clinically validated biomarkers

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07836647

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 07836647

Country of ref document: EP

Kind code of ref document: A2