sx001-00001-006235异常是毛fgxsx什么意思思

Means and Methods for Diagnosing Pancreatic Cancer in a Subject
United States Patent Application
The present invention relates to the field of diagnostic methods. Specifically, the present invention contemplates a method for diagnosing pancreatic cancer in a subject, a method for identifying whether a subject is in need for a therapy of pancreatic cancer or a method for determining whether a pancreatic cancer therapy is successful. The invention also relates to tools for carrying out the aforementioned methods, such as diagnostic devices.
Inventors:
Reszka, Regina (Panketal, DE)
Kamlage, Beate (Berlin, DE)
Kalthoff, Holger (Kiel, DE)
Schniewind, Bodo (Kiel, DE)
Mayerle, Julia (Greifswald, DE)
Lerch, Markus M. (Greifswald, DE)
Pilarsky, Christian (Dresden, DE)
Grützmann, Robert (Dresden, DE)
Application Number:
Publication Date:
10/30/2014
Filing Date:
11/29/2012
Export Citation:
METANOMICS HEALTH GMBH (Berlin, DE)
Primary Class:
Other Classes:
435/287.1,
International Classes:
G01N33/574
View Patent Images:
&&&&&&PDF help
Related US Applications:
August, 2009Rebek Jr. et al.December, 2003Kao et al.September, 2009Ratajczak et al.July, 2009Li et al.March, 2008Lipovsek et al.December, 2009Bagby et al.August, 2009Vermette et al.March, 2009Bergwerff et al.August, 2009Chatterjee et al.December, 2009Duer et al.October, 2008Hart et al.
Foreign References:
A method for diagnosing pancreas cancer in a subject comprising: a) determining in a sample of a subject suspected to suffer from pancreas cancer the amount of at least one biomarker from Tables 2a, 2b, 3a, 3b, 4a or 4b or at least one biomarker of Tables 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32; and b) comparing the said amount of the at least one biomarker with a reference, whereby pancreas cancer is to be diagnosed.
The method of claim 1, wherein said reference is derived from a sample of a subject or group of subjects known not to suffer from pancreatic cancer or is a calculated reference.
The method of claim 1, wherein said reference is derived from a sample of a subject or group of subjects known to suffer from pancreatic cancer.
The method of claim 1, wherein said at least one biomarker is a biomarker of category 1 or category 2.
The method of claim 4, wherein said subject exhibits pancreatitis as an underlying pancreatic disease.
The method of claim 1, wherein said reference is derived from a subject or group of subjects suffering from pancreatitis and wherein the said at least one biomarker is from table 6, 7, 8, 15, 16, 17, 27, 28 or 29.
The method of claim 1, wherein said reference is derived from a subject or group of subjects suffering from liver cirrhosis and wherein the said at least one biomarker is from table 18, 19, 20, 30, 31 or 32.
The method of claim 1, wherein said reference is derived from a subject or group of subjects suffering from pancreatitis and/or liver cirrhosis and wherein the said at least one biomarker is from table 12, 13, 14, 24, 25 or 26.
A method for identifying whether a subject is in need of a pancreas cancer therapy comprising the steps of the method of claim 1 and the further step of identifying a subject in need of a pancreas cancer therapy if said subject is to be diagnosed to suffer from pancreas cancer.
A method for determining whether a therapy against pancreatic cancer is successful in a subject comprising the steps of the method of claim 1 and the further step of determining whether a therapy is successful if no pancreatic cancer is diagnosed.
The method of claim 9, wherein said pancreas cancer therapy comprises surgery, radiotherapy or drug treatment.
The method of claim 1, wherein said sample is a plasma, blood or serum sample.
The method of claim 1, wherein said subject is a human.
The method of claim 1, wherein said pancreas cancer is pancreas adenocarcinoma.
A device for diagnosing pancreas cancer in a sample of a subject comprising: a) an analyzing unit for the said sample of the subject comprising a detector for at least one biomarker of Tables 2a, 2b, 3a, 3b, 4a or 4b or at least one biomarker of Tables 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32, said detector allowing for the determination of the amount of the said at least one bio and operatively linked thereto, b) an evaluation unit comprising a data processing unit and a data base, said data base comprising a stored reference and said data processing unit having tangibly embedded an algorithm for carrying out a comparison of the amount of the at least one biomarker determined by the analyzing unit and the stored reference and for generating an output information based on which the diagnosis can be established.
Use of at least one biomarker from Tables 2a, 2b, 3a, 3b, 4a or 4b or a detection agent therefor or at least one biomarker of Tables 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 or a detection agent therefor in a sample of a subject suspected to suffer from pancreatic cancer for diagnosing pancreatic cancer.
Description:
The present invention relates to the field of diagnostic methods. Specifically, the present invention contemplates a method for diagnosing pancreatic cancer in a subject, a method for identifying whether a subject is in need for a therapy of pancreatic cancer or a method for determining whether a pancreatic cancer therapy is successful. The invention also relates to tools for carrying out the aforementioned methods, such as diagnostic devices.Pancreatic cancer has the worst prognosis of all solid tumors with 5-year survival rates of less than 5% but an increasing incidence (Everhart 2009, Gastroenterology 136:). There is a widely acknowledged demand for the establishment of innovative tools and technologies for point-of-care utilization of specific biomarkers and novel molecular imaging tools for early diagnosis, prognostic stratification and differential diagnosis of pancreatic cancer. Advances in these areas are pivotal to improve the prognosis of this malignancy, since timely surgical resection of early stage tumors is currently the only effective means of treatment of this dismal disease.The mortality of this cancer type is the highest of any cancer type in Europe and the western world. People die soon after diagnosis due to the lack of means for early detection. Early symptoms are rare and uncharacteristic. Thus, PDACs are commonly diagnosed in an advanced stage of the disease. To date, the best imaging technologies to detect PDAC are endoscopic ultrasound (EUS), spiral computer tomography (CT), magnetic resonance cholangiopancreatography (MRCP) or endoscopic retrograde cholangiopancreatography (ERCP) (Dewitt 2006, Gastroenterol Hepatol. (4):717-25). Unfortunately, the resolution of these technologies for detecting neoplastic lesions within the pancreas is in the range of 3-10 mm. Thus, they are not able to detect pancreatic neoplasia at a curable stage. The serum concentration of conventional tumor markers such as CA19-9 is increased in a subset of pancreatic cancer patients (Fry 2008, Langenbecks Arch Surg. (393): 883-90). However, so far all available markers lack sensitivity and tumor specificity. Thus, new approaches are urgently needed to increase the diagnostic sensitivity towards the detection of very small, early stage PDAC and its precursor lesions (PanINs and IPMNs) as well as prognostic subgroups of advanced tumors.The association between chronic inflammation and the development of malignancies has been recognized for many years. For pancreatic cancer this association was only recently confirmed and a consensus conference agreed upon a new classification for pancreatic intraepithelial neoplasia as noninvasive precursor lesions (Hruban 2004, Am J Surg Path (28): 977-987). Chronic pancreatitis is defined as recurrent bouts of a sterile inflammatory disease characterized by often progressive and irreversible morphological changes, typically causing pain and permanent impairment of pancreatic function. With an incidence of 8.2, a prevalence of 27.4 per 100 000 population and a 0.04% to 5% frequency in unselected autopsy specimens chronic pancreatitis represents a frequent disorder of the gastrointestinal tract. Various etiologies are responsible for the development of chronic pancreatitis. An increased risk of patients suffering from of chronic pancreatitis to die from pancreatic cancer was shown in an international cooperative investigation conducted by AB Lowenfels and coworkers as a multicenter historical cohort study of 2015 patients with chronic pancreatitis recruited from clinical centers in 6 countries in 1993. This study found a cumulative risk of pancreatic cancer in patients with chronic pancreatitis of 1.8% after 10 years and of 4% after 20 years with a standardized incidence ratio of 14.4. For patients with a minimum of two years follow up the risk of pancreatic cancer was 16.5 fold higher than that of the general population (Lowenfels 1993, N Engl J Med (328): ). The search for an association between chronic pancreatitis and pancreatic cancer intensified when in 1996 a single point mutation in the third exon of the cationic trypsinogen gene on chromosome 7 (7q35) was found to be associated with hereditary pancreatitis and multiple kindreds were subsequently identified and reported. Only very recently the EUROPAC study group presented their work on clinical and genetic characteristics in hereditary pancreatitis. In a multilevel proportional hazard model employing data obtained from the European Registry of Hereditary Pancreatitis this group presented 112 families in 14 countries (418 affected individuals) (Howes 2004, Clinical Gastroenterology and Hepatology (2): 252-261). The cumulative risk (95% CI) of pancreatic cancer was 44.0% (8.0%-80.0%) at 70 years from symptom onset with a standardized incidence ratio of 67% (50%-82%). A previous study had also shown an estimated lifetime risk of pancreatic cancer of 40% (Lowenfels 2001, JAMA 286: 169-170, Lowenfels 1997, J Natl Cancer Inst 89: 442-44656).In pancreatic cancer imaging studies fail to detect early pancreatic malignancies in a curable stage, however in the background of chronic pancreatitis imaging studies such as EUS, CT or MRI drop sensitivity and specificity to a degree where tossing a coin is equally reliable. Thus, the detection of pancreatic malignancy in a high risk cohort would be highly desired.There are a few reports of metabolic changes in patients suffering pancreas-associated diseases. Schrader et al (Schrader 2009, Panceas 38: 416-421) suggests that patients with pancreatic cancer and chronic pancreatitis show significant changes in serum amino acid levels. It has been suggested that among the ceramides sphingomyelin on the cell surface of cancer cells takes actively part in cell signalling. Ceramides are known to induce apoptosis in cancer cells. Low levels of sphingomyelin suggest less responsiveness to gemcitabine treatment (Modrak 2009, Mol Cancer Res 7:890-896).In conclusion with a 5-year survival rate of 0.5-5%, pancreatic cancer carries the most dismal prognosis of all human tumors and represents the 4th leading cause in cancer-related deaths worldwide. It is thus a disease with a major socioeconomic impact. Accurate diagnosis and timely surgical resection of early tumors currently offer the only realistic prospect for the improvement of patient prognosis.The technical problem underlying the present invention can be seen as the provision of means and methods for complying with the aforementioned needs. The technical problem is solved by the embodiments characterized in the claims and herein below.Thus, the present invention relates to a method for diagnosing pancreas cancer in a subject comprising the steps of:
(a) determining in a sample of a subject suspected to suffer from pancreas cancer the amount of at least one biomarker from Tables 2a, 2b, 3a, 3b, 4a, or 4b; and(b) comparing the said amount of the at least one biomarker with a reference, whereby pancreas cancer is to be diagnosed.
The method as referred to in accordance with the present invention includes a method which essentially consists of the aforementioned steps or a method which includes further steps. However, it is to be understood that the method, in a preferred embodiment, is a method carried out ex vivo, i.e. not practised on the human or animal body. The method, preferably, can be assisted by automation.The term “diagnosing” as used herein refers to assessing whether a subject suffers from the pancreatic cancer, or not. As will be understood by those skilled in the art, such an assessment, although preferred to be, may usually not be correct for 100% of the investigated subjects. The term, however, requires that a statistically significant portion of subjects can be correctly assessed and, thus, diagnosed. Whether a portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p-value determination, Student's t-test, Mann-Whitney test, etc. Details are found in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. Preferred confidence intervals are at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or at least 95%. The p-values are, preferably, 0.2, 0.1, or 0.05.The term includes individual diagnosis of pancreatic cancer or its symptoms as well as continuous monitoring of a patient. Monitoring, i.e. diagnosing the presence or absence of pancreatic cancer or the symptoms accompanying it at various time points, includes monitoring of patients known to suffer from pancreatic cancer as well as monitoring of subjects known to be at risk of developing pancreatic cancer. Furthermore, monitoring can also be used to determine whether a patient is treated successfully or whether at least symptoms of pancreatic cancer can be ameliorated over time by a certain therapy.The term “pancreatic cancer” or “pancreas cancer” as used herein relates to cancer which is derived from pancreatic cells. Preferably, pancreatic cancer as used herein is pancreatic adenocarcinoma. The symptoms accompanying pancreatic cancer are well known from standard text books of medicine such as Stedmen or Pschyrembl. Moreover, the terms “cancer” and “carcinoma” may, in general, also be used herein interchangeably.The term “biomarker” as used herein refers to a molecular species which serves as an indicator for a disease or effect as referred to in this specification. Said molecular species can be a metabolite itself which is found in a sample of a subject. Moreover, the biomarker may also be a molecular species which is derived from said metabolite. In such a case, the actual metabolite will be chemically modified in the sample or during the determination process and, as a result of said modification, a chemically different molecular species, i.e. the analyte, will be the determined molecular species. It is to be understood that in such a case, the analyte represents the actual metabolite and has the same potential as an indicator for the respective medical condition.Moreover, a biomarker according to the present invention is not necessarily corresponding to one molecular species. Rather, the biomarker may comprise stereoisomers or enantiomeres of a compound. Further, a biomarker can also represent the sum of isomers of a biological class of isomeric molecules. Said isomers shall exhibit identical analytical characteristics in some cases and are, therefore, not distinguishable by various analytical methods including those applied in the accompanying Examples described below. However, the isomers will share at least identical sum formula parameters and, thus, in the case of, e.g., lipids an identical chain length and identical numbers of double bonds in the fatty acid and/or sphingo base moietiesPolar biomarkers can be, preferably, obtained by techniques referred to in this specification elsewhere and as described in Example 1, below. Lipid biomarkers can be obtained in accordance with the present invention, preferably, as described in this specification elsewhere and, in particular, either as lipid fraction by separation of a sample after protein precipitation into an aqueous polar and an organic lipid phase by, e.g., a mixture of ethanol and dichloromethane as described in Example 1, below. Those biomarkers may be marked by “lipid fraction” herein. Alternatively or in addition, complex lipids can be obtained by liquid/liquid extraction using chloroform/methanol followed by fractionation using normal phase liquid chromatography (NPLC) as described in Example 1, below. Those biomarkers may be marked or marked in addition by the specific sub-fraction, e.g., “ceramide fraction” or “CER” or “sphingomyelins” or “SM” etc. Further details are found below in the Tables.In the method according to the present invention, at least one metabolite of the biomarkers shown in Tables 2a, 2b, 3a, 3b, 4a, or 4b is to be determined. However, more preferably, a group of biomarkers will be determined in order to strengthen specificity and/or sensitivity of the assessment. Such a group, preferably, comprises at least 2, at least 3, at least 4, at least 5, at least 10 or up to all of the said biomarkers shown in the Tables 2a, 2b, 3a, 3b, 4a, or 4b.Preferably, the at least one biomarker determined in the method of the present invention is a biomarker of category 1 as shown in Table 2a or 2b. More preferably, the at least one biomarker is selected from the group consisting of the spingomyelins of the sphingomyeline fraction (SM) and the cermides of the ceramide fraction (CER) as shown in table 2b, i.e. SM_Sphingomyelin (d18:1,C19:0), SM_Sphingomyelin (d18:1,C21:0), SM_Sphingomyelin (d18:2,022:0), SM_Sphingomyelin (d17:1,024:1), SM_Sphingomyelin (d18:2,020:0), SM_Sphingomyelin (d18:2,019:0), SM_Sphingomyelin (d18:2,021:0), CER_Ceramide (d17:1,022:0), and CER_Ceramide (d18:2,022:0). Preferably, the aforementioned sphingomyeline fraction (SM) and/or ceramide fraction (CER) is obtained by a method for fractionation as set forth below or in the accompanying Examples, below.Most preferably, the aforementioned group of biomarkers is determined as the at least one biomarker in the method of the invention. More preferably, said at least one biomarker of category 1 is capable of diagnosing pancreatic carcinoma and accompanying liver cirrhosis or pancreatitis in a subject when compared to a reference derived from a healthy control subject or group of such subjects. More preferably, said diagnosis is achieved with significance levels set forth in Table 1, below. The category 1 biomarkers are capable of, preferably, diagnosing pancreatic cancer in a high risk group of subjects exhibiting the accompanying disease pancreatitis and/or liver cirrhosis.Preferably, the at least one biomarker determined in the method of the present invention is a biomarker of category 2 as shown in Table 3a or 3b. More preferably, the at least one biomarker is selected from the group consisting of: 1-Hydroxy-2-amino-(cis,trans)-3,5-octadecadiene (from sphingolipids), Sphingomyelin (d18:2,018:0), Glycerol phosphate, lipid fraction, Arachidonic acid (C20:cis[5,8,11,14]4), Phosphate, lipid fraction, myo-Inositol-2-phosphate, lipid fraction (myo-Inositolphospholipids), Cholesta-2,4,6-triene, erythro-C16-Sphingosine, SM_Sphingomyelin (d18:2,022:1), SM_Sphingomyelin (d18:2,020:1), SM_Sphingomyelin (d18:2,024:1), SM_Sphingomyelin (d18:1,020:0), SM_Sphingomyelin (d18:1,019:0), SM_Sphingomyelin (d18:2,023:1), CER_Ceramide (d18:2,018:0), SM_Sphingomyelin (d18:2,018:1), SM_Sphingomyelin (d18:1,022:0), SM_Sphingomyelin (d18:2,016:0), SM_Sphingomyelin (d18:1,021:0), CER_Ceramide (d18:2,020:0), CER_Ceramide (d18:1,022:0), CER_Ceramide (d16:1,018:0), SM_Sphingomyelin (d16:1,022:1), SM_Sphingomyelin (d16:1,020:0), SM_Sphingomyelin (d16:1,018:1), CER_Ceramide (d16:1,020:0), SM_Sphingomyelin (d17:1,024:1), CER_Ceramide (d18:2,024:1), CER_Ceramide (d17:1,024:1), Sphingosine-1-phosphate (d18:1), SM_Sphingomyelin (d16:1,024:1) and CER_Ceramide (d16:1,024:1).Most preferably, the aforementioned group of biomarkers is determined as the at least one biomarker in the method of the invention. More preferably, said at least one biomarker of category 2 is capable of diagnosing pancreatic carcinoma and accompanying pancreatitis in a subject when compared to a reference derived from a healthy control subject or group of such subjects. More preferably, said diagnosis is achieved with significance levels set forth in Table 1, below. The category 2 biomarkers are capable of, preferably, diagnosing pancreatic cancer in a risk group of subjects exhibiting the accompanying disease pancreatitis.Preferably, the at least one biomarker determined in the method of the present invention is a biomarker as shown in Table 4a or 4b. More preferably, the at least one biomarker is selected from the group consisting of: Sphingomyelin (d18:2,C18:0), 0-Acetylcarnitine, 1-Hydroxy-2-amino-(cis,trans)-3,5-octadecadiene (from sphingolipids), erythro-Sphingosine, Behenic acid (C22:0), Eicosanoic acid (C20:0), 3-O-Methylsphingosine (d18:1), 5-O-Methylsphingosine (d18:1), 3-Hydroxyisobutyrate, threo-Sphingosine, Nervonic acid (C24:cis[15]1), 7-Methylguanosine, erythro-C16-Sphingosine, Tetradecanoylcarnitine, Sarcosine, Phytosphingosine (t18:0), total, Sphingomyelin (d18:1,C23:0), 5-O-Methylsphingosine (d16:1), Sphingomyelin (d18:2,C16:0), Tricosanoic acid (C23:0), Hexadecanoylcarnitine, Sphingomyelin (d18:1,C24:0), 3-Hydroxybutyrate, Octadecanoylcarnitine, SM_Sphingomyelin (d18:1,C19:0), SM_Sphingomyelin (d18:2,C18:1), SM_Sphingomyelin (d18:2,C22:1), SM_Sphingomyelin (d18:2,020:1), SM_Sphingomyelin (d18:2,020:0), SM_Sphingomyelin (d18:2,019:0), SM_Sphingomyelin (d18:2,C18:0), CER_Ceramide (d18:2,020:0), SM_Sphingomyelin (d18:1,C20:0), CER_Ceramide (d18:1,C20:0), SM_Sphingomyelin (d18:2,C22:0), SM_Sphingomyelin (d17:1,C18:0), CER_Ceramide (d18:2,018:0), SM_Sphingomyelin (d17:1,C20:0), CER_Ceramide (d18:1,C18:0), SM_Sphingomyelin (d18:2,021:0), SM_Sphingomyelin (d18:2,C24:2), SM_Sphingomyelin (d18:2,C23:1), SM_Sphingomyelin (d18:1,C21:0), CER_Ceramide (d18:1,C21:0), CER_Ceramide (d18:2,C22:0), SM_Sphingomyelin (d18:2,C24:1), CER_Ceramide (d16:1,C18:0), SM_Sphingomyelin (d18:1,C22:0), CER_Ceramide (d17:1,C22:0), CER_Ceramide (d18:1,C22:0), SM_Sphingomyelin (d17:1,C22:0), CER_Ceramide (d17:1,C16:0), SM_Sphingomyelin (d17:1,C24:1), CER_Ceramide (d16:1,C20:0), CER_Ceramide (d17:1,C24:1), SM_Sphingomyelin (d16:1,C20:0), CER_Ceramide (d18:1,C22:1), CER_Ceramide (d18:2,016:0), SM_Sphingomyelin (d16:1,C18:1), SM_Sphingomyelin (d18:2,016:0), CER_Ceramide (d18:2,C24:1), CER_Ceramide (d18:1,C23:1), CER_Ceramide (d18:1,C24:2), SM_Sphingomyelin (d18:2,C24:0), SM_Sphingomyelin (d17:1,C16:0), SM_Sphingomyelin (d16:1,C18:0), SM_Sphingomyelin (d16:1,C22:1), CER_Ceramide (d18:1,C24:1), SM_Sphingomyelin (d18:2,C23:0), CER_Ceramide (d18:2,024:0), CE_Cholesterylester C20:4, CER_Ceramide (d18:1,C16:0), CER_Ceramide (d16:1,C22:0), Sphingosine-1-phosphate (d17:1), CER_Ceramide (d18:2,023:0), SM_Sphingomyelin (d18:2,C14:0), SM_Sphingomyelin (d17:1,C24:0), CER_Ceramide (d18:1,C14:0), SM_Sphingomyelin (d17:1,C23:0), CER_Ceramide (d16:1,C16:0), CER_Ceramide (d18:1,C24:0), SM_Sphingomyelin (d16:1,C22:0), CER_Ceramide (d16:1,C24:1), CER_Ceramide (d17:1,C24:0), SM_Sphingomyelin (d18:1,C16:0), SM_Sphingomyelin (d18:1,C23:0), SM_Sphingomyelin (d16:1,C21:0), SM_Sphingomyelin (d18:1,C24:0), Sphingadienine-1-phosphate (d18:2), CER_Ceramide (d18:1,C23:0), SM_Sphingomyelin (d16:1,C24:1), SM_Sphingomyelin (d18:1,C14:0), and CER_Ceramide (d17:1,C23:0).Most preferably, the aforementioned group of biomarkers is determined as the at least one biomarker in the method of the invention. More preferably, said aforementioned at least one biomarker is capable of diagnosing pancreatic carcinoma in a subject when compared to a reference derived from a group of subjects comprising healthy subjects and/or subjects suffering from pancreatitis and/or subjects suffering from liver cirrhosis (eg non-cancer group). More preferably, said diagnosis is achieved with analysis approaches set forth in Table 1, below. Preferably, the aforementioned group of biomarkers comprises at least two ceramides and at least one sphingomyeline, 3 ceramides and at least 2 sphingomyelines, 4 ceramides and at least 3 sphingomyelines, 5 ceramides and at least 4 sphingomyelines, or 6 ceramides and at least 5 sphingomyelines,A metabolite as used herein refers to at least one molecule of a specific metabolite up to a plurality of molecules of the said specific metabolite. It is to be understood further that a group of metabolites means a plurality of chemically different molecules wherein for each metabolite at least one molecule up to a plurality of molecules may be present. A metabolite in accordance with the present invention encompasses all classes of organic or inorganic chemical compounds including those being comprised by biological material such as organisms. Preferably, the metabolite in accordance with the present invention is a small molecule compound. More preferably, in case a plurality of metabolites is envisaged, said plurality of metabolites representing a metabolome, i.e. the collection of metabolites being comprised by an organism, an organ, a tissue, a body fluid or a cell at a specific time and under specific conditions.In addition to the specific biomarkers recited in the specification, other biomarkers may be, preferably, determined as well in the methods of the present invention. Such biomarkers may include peptide or polypeptide biomarkers or glycosides such as the CA19.9 antigen.The term “sample” as used herein refers to samples from body fluids, preferably, blood, plasma, serum, saliva or urine, or samples derived, e.g., by biopsy, from cells, tissues or organs, in particular from the heart. More preferably, the sample is a blood, plasma or serum sample, most preferably, a plasma sample. Biological samples can be derived from a subject as specified elsewhere herein. Techniques for obtaining the aforementioned different types of biological samples are well known in the art. For example, blood samples may be obtained by blood taking while tissue or organ samples are to be obtained, e.g., by biopsy.The aforementioned samples are, preferably, pre-treated before they are used for the method of the present invention. As described in more detail below, said pre-treatment may include treatments required to release or separate the compounds or to remove excessive material or waste. Suitable techniques comprise centrifugation, extraction, fractioning, ultrafiltration, protein precipitation followed by filtration and purification and/or enrichment of compounds. Moreover, other pre-treatments are carried out in order to provide the compounds in a form or concentration suitable for compound analysis. For example, if gas-chromatography coupled mass spectrometry is used in the method of the present invention, it will be required to derivatize the compounds prior to the said gas chromatography. Suitable and necessary pre-treatments depend on the means used for carrying out the method of the invention and are well known to the person skilled in the art. Pre-treated samples as described before are also comprised by the term “sample” as used in accordance with the present invention.The term “subject” as used herein relates to animals and, preferably, to mammals. More preferably, the subject is a primate and, most preferably, a human. The subject, preferably, is suspected to suffer from pancreatic cancer, i.e. it may already show some or all of the symptoms associated with the disease. Moreover, the subject may also preferably suffer from or shall be suspected to suffer from pancreatitis and/or liver cirrhosis in addition. Preferably, the subject, however, is besides the aforementioned diseases and disorders apparently healthy. The said subject, preferably, is at increased risk of developing pancreatic cancer (Brand R E et al, Gut. 0-9). More preferably, such a subject being at increased risk has one or more relatives suffering from pancreatic cancer, has a defined genetic predisposition for developing pancreatic cancer, including but not exclusive to Peutz-Jeghers Syndrome, has one or more relatives suffering from pancreatitis, and/or has a defined genetic predisposition for developing pancreatitis.The term “determining the amount” as used herein refers to determining at least one characteristic feature of a biomarker to be determined by the method of the present invention in the sample. Characteristic features in accordance with the present invention are features which characterize the physical and/or chemical properties including biochemical properties of a biomarker. Such properties include, e.g., molecular weight, viscosity, density, electrical charge, spin, optical activity, colour, fluorescence, chemoluminescence, elementary composition, chemical structure, capability to react with other compounds, capability to elicit a response in a biological read out system (e.g., induction of a reporter gene) and the like. Values for said properties may serve as characteristic features and can be determined by techniques well known in the art. Moreover, the characteristic feature may be any feature which is derived from the values of the physical and/or chemical properties of a biomarker by standard operations, e.g., mathematical calculations such as multiplication, division or logarithmic calculus. Most preferably, the at least one characteristic feature allows the determination and/or chemical identification of the said at least one biomarker and its amount. Accordingly, the characteristic value, preferably, also comprises information relating to the abundance of the biomarker from which the characteristic value is derived. For example, a characteristic value of a biomarker may be a peak in a mass spectrum. Such a peak contains characteristic information of the biomarker, i.e. the m/z information, as well as an intensity value being related to the abundance of the said biomarker (i.e. its amount) in the sample.As discussed before, each biomarker comprised by a sample may be, preferably, determined in accordance with the present invention quantitatively or semi-quantitatively. For quantitative determination, either the absolute or precise amount of the biomarker will be determined or the relative amount of the biomarker will be determined based on the value determined for the characteristic feature(s) referred to herein above. The relative amount may be determined in a case were the precise amount of a biomarker can or shall not be determined. In said case, it can be determined whether the amount in which the biomarker is present is enlarged or diminished with respect to a second sample comprising said biomarker in a second amount. In a preferred embodiment said second sample comprising said biomarker shall be a calculated reference as specified elsewhere herein. Quantitatively analysing a biomarker, thus, also includes what is sometimes referred to as semi-quantitative analysis of a biomarker.Moreover, determining as used in the method of the present invention, preferably, includes using a compound separation step prior to the analysis step referred to before. Preferably, said compound separation step yields a time resolved separation of the metabolites comprised by the sample. Suitable techniques for separation to be used preferably in accordance with the present invention, therefore, include all chromatographic separation techniques such as liquid chromatography (LC), high performance liquid chromatography (HPLC), gas chromatography (GC), thin layer chromatography, size exclusion or affinity chromatography. These techniques are well known in the art and can be applied by the person skilled in the art without further ado. Most preferably, LC and/or GC are chromatographic techniques to be envisaged by the method of the present invention. Suitable devices for such determination of biomarkers are well known in the art. Preferably, mass spectrometry is used in particular gas chromatography mass spectrometry (GC-MS), liquid chromatography mass spectrometry (LC-MS), direct infusion mass spectrometry or Fourier transform ion-cyclotrone-resonance mass spectrometry (FT-ICR-MS), capillary electrophoresis mass spectrometry (CE-MS), high-performance liquid chromatography coupled mass spectrometry (HPLC-MS), quadrupole mass spectrometry, any sequentially coupled mass spectrometry, such as MS-MS or MS-MS-MS, inductively coupled plasma mass spectrometry (ICP-MS), pyrolysis mass spectrometry (Py-MS), ion mobility mass spectrometry or time of flight mass spectrometry (TOF). Most preferably, LC-MS and/or GC-MS are used as described in detail below. Said techniques are disclosed in, e.g., Nissen 1995, Journal of Chromatography A, 703: 37-57, U.S. Pat. No. 4,540,884 or U.S. Pat. No. 5,397,894, the disclosure content of which is hereby incorporated by reference. As an alternative or in addition to mass spectrometry techniques, the following techniques may be used for compound determination: nuclear magnetic resonance (NMR), magnetic resonance imaging (MRI), Fourier transform infrared analysis (FTIR), ultraviolet (UV) spectroscopy, refraction index (RI), fluorescent detection, radiochemical detection, electrochemical detection, light scattering (LS), dispersive Raman spectroscopy or flame ionisation detection (FID). These techniques are well known to the person skilled in the art and can be applied without further ado. The method of the present invention shall be, preferably, assisted by automation. For example, sample processing or pre-treatment can be automated by robotics. Data processing and comparison is, preferably, assisted by suitable computer programs and databases. Automation as described herein before allows using the method of the present invention in high-throughput approaches.Moreover, the at least one biomarker can also be determined by a specific chemical or biological assay. Said assay shall comprise means which allow to specifically detect the at least one biomarker in the sample. Preferably, said means are capable of specifically recognizing the chemical structure of the biomarker or are capable of specifically identifying the biomarker based on its capability to react with other compounds or its capability to elicit a response in a biological read out system (e.g., induction of a reporter gene). Means which are capable of specifically recognizing the chemical structure of a biomarker are, preferably, antibodies or other proteins which specifically interact with chemical structures, such as receptors or enzymes. Specific antibodies, for instance, may be obtained using the biomarker as antigen by methods well known in the art. Antibodies as referred to herein include both polyclonal and monoclonal antibodies, as well as fragments thereof, such as Fv, Fab and F(ab)2 fragments that are capable of binding the antigen or hapten. The present invention also includes humanized hybrid antibodies wherein amino acid sequences of a non-human donor antibody exhibiting a desired antigen-specificity are combined with sequences of a human acceptor antibody. Moreover, encompassed are single chain antibodies. The donor sequences will usually include at least the antigen-binding amino acid residues of the donor but may comprise other structurally and/or functionally relevant amino acid residues of the donor antibody as well. Such hybrids can be prepared by several methods well known in the art. Suitable proteins which are capable of specifically recognizing the biomarker are, preferably, enzymes which are involved in the metabolic conversion of the said biomarker. Said enzymes may either use the biomarker as a substrate or may convert a substrate into the biomarker. Moreover, said antibodies may be used as a basis to generate oligopeptides which specifically recognize the biomarker. These oligopeptides shall, for example, comprise the enzyme's binding domains or pockets for the said biomarker. Suitable antibody and/or enzyme based assays may be RIA (radioimmunoassay), ELISA (enzyme-linked immunosorbent assay), sandwich enzyme immune tests, electrochemiluminescence sandwich immunoassays (ECLIA), dissociation-enhanced lanthanide fluoro immuno assay (DELFIA) or solid phase immune tests. Moreover, the biomarker may also be determined based on its capability to react with other compounds, i.e. by a specific chemical reaction. Further, the biomarker may be determined in a sample due to its capability to elicit a response in a biological read out system. The biological response shall be detected as read out indicating the presence and/or the amount of the biomarker comprised by the sample. The biological response may be, e.g., the induction of gene expression or a phenotypic response of a cell or an organism. In a preferred embodiment the determination of the least one biomarker is a quantitative process, e.g., allowing also the determination of the amount of the at least one biomarker in the sampleAs described above, said determining of the at least one biomarker can, preferably, comprise mass spectrometry (MS). Mass spectrometry as used herein encompasses all techniques which allow for the determination of the molecular weight (i.e. the mass) or a mass variable corresponding to a compound, i.e. a biomarker, to be determined in accordance with the present invention. Preferably, mass spectrometry as used herein relates to GC-MS, LC-MS, direct infusion mass spectrometry, FT-ICR-MS, CE-MS, HPLC-MS, quadrupole mass spectrometry, any sequentially coupled mass spectrometry such as MS-MS or MS-MS-MS, ICP-MS, Py-MS, TOF or any combined approaches using the aforementioned techniques. How to apply these techniques is well known to the person skilled in the art. Moreover, suitable devices are commercially available. More preferably, mass spectrometry as used herein relates to LC-MS and/or GC-MS, i.e. to mass spectrometry being operatively linked to a prior chromatographic separation step. More preferably, mass spectrometry as used herein encompasses quadrupole MS. Most preferably, said quadrupole MS is carried out as follows: a) selection of a mass/charge quotient (m/z) of an ion created by ionisation in a first analytical quadrupole of the mass spectrometer, b) fragmentation of the ion selected in step a) by applying an acceleration voltage in an additional subsequent quadrupole which is filled with a collision gas and acts as a collision chamber, c) selection of a mass/charge quotient of an ion created by the fragmentation process in step b) in an additional subsequent quadrupole, whereby steps a) to c) of the method are carried out at least once and analysis of the mass/charge quotient of all the ions present in the mixture of substances as a result of the ionisation process, whereby the quadrupole is filled with collision gas but no acceleration voltage is applied during the analysis. Details on said most preferred mass spectrometry to be used in accordance with the present invention can be found in WO 03/073464.More preferably, said mass spectrometry is liquid chromatography (LC) MS and/or gas chromatography (GC) MS. Liquid chromatography as used herein refers to all techniques which allow for separation of compounds (i.e. metabolites) in liquid or supercritical phase. Liquid chromatography is characterized in that compounds in a mobile phase are passed through the stationary phase. When compounds pass through the stationary phase at different rates they become separated in time since each individual compound has its specific retention time (i.e. the time which is required by the compound to pass through the system). Liquid chromatography as used herein also includes HPLC. Devices for liquid chromatography are commercially available, e.g. from Agilent Technologies, USA. Gas chromatography as applied in accordance with the present invention, in principle, operates comparable to liquid chromatography. However, rather than having the compounds (i.e. metabolites) in a liquid mobile phase which is passed through the stationary phase, the compounds will be present in a gaseous volume. The compounds pass the column which may contain solid support materials as stationary phase or the walls of which may serve as or are coated with the stationary phase. Again, each compound has a specific time which is required for passing through the column. Moreover, in the case of gas chromatography it is preferably envisaged that the compounds are derivatised prior to gas chromatography. Suitable techniques for derivatisation are well known in the art. Preferably, derivatisation in accordance with the present invention relates to methoxymation and trimethylsilylation of, preferably, polar compounds and transmethylation, methoxymation and trimethylsilylation of, preferably, non-polar (i.e. lipophilic) compounds.The term “reference” refers to values of characteristic features of each of the biomarker which can be correlated to a medical condition, i.e. the presence or absence of the disease, diseases status or an effect referred to herein. Preferably, a reference is a threshold value (e.g., an amount or ratio of amounts) for a biomarker whereby values found in a sample to be investigated which are higher than or essentially identical to the threshold are indicative for the presence of a medical condition while those being lower are indicative for the absence of the medical condition. It will be understood that also preferably, a reference may be a threshold value for a biomarker whereby values found in a sample to be investigated which are lower or identical than the threshold are indicative for the presence of a medical condition while those being higher are indicative for the absence of the medical condition.In accordance with the aforementioned method of the present invention, a reference is, preferably, a reference obtained from a sample from a subject or group of subjects known to suffer from pancreatic cancer. In such a case, a value for the at least one biomarker found in the test sample being essentially identical is indicative for the presence of the disease.Moreover, the reference, also preferably, could be from a subject or group of subjects known not to suffer from pancreatic cancer, preferably, an apparently healthy subject. In such a case, a value for the at least one biomarker found in the test sample being altered with respect to the reference is indicative for the presence of the disease. The same applies mutatis mutandis for a calculated reference being, most preferably, the average or median for the relative value or the value for a degree of change of the at least one biomarker in a population of individuals (comprising the subject to be investigated). The relative values or degrees of changes of the at least one biomarker of said individuals of the population can be determined as specified elsewhere herein. How to calculate a suitable reference value, preferably, the average or median, is well known in the art. The population of subjects referred to before shall comprise a plurality of subjects, preferably, at least 5, 10, 50, 100, 1,000 or 10,000 subjects. It is to be understood that the subject to be diagnosed by the method of the present invention and the subjects of the said plurality of subjects are of the same species.The value for the at least one biomarker of the test sample and the reference values are essentially identical, if the values for the characteristic features and, in the case of quantitative determination, the intensity values are essentially identical. Essentially identical means that the difference between two values is, preferably, not significant and shall be characterized in that the values for the intensity are within at least the interval between 1st and 99th percentile, 5th and 95th percentile, 10th and 90th percentile, 20th and 80th percentile, 30th and 70th percentile, 40th and 60th percentile of the reference value, preferably, the 50th, 60th, 70th, 80th, 90th or 95th percentile of the reference value. Statistical test for determining whether two amounts are essentially identical are well known in the art and are also described elsewhere herein.An observed difference for two values, on the other hand, shall be statistically significant. A difference in the relative or absolute value is, preferably, significant outside of the interval between 45th and 55th percentile, 40th and 60th percentile, 30th and 70th percentile, 20th and 80th percentile, 10th and 90th percentile, 5th and 95th percentile, 1st and 99th percentile of the reference value. Preferred relative changes of the medians or degrees of changes are described in the accompanying Tables as well as in the Examples. In the Tables below, a preferred relative change for the biomarkers is indicated as “up” for an increase and “down” for a decrease in column “direction of change”. Values for preferred degrees of changes are indicated in the column “estimated fold change”. The preferred references for the aforementioned relative changes or degrees of changes are indicated in the Tables below as well. It will be understood that these changes are, preferably, observed in comparison to the references indicated in the respective Tables, below. Preferably, the reference, i.e. values for at least one characteristic feature of the at least one biomarker or ratios thereof, will be stored in a suitable data storage medium such as a database and are, thus, also available for future assessments.The term “comparing” refers to determining whether the determined value of a biomarker is essentially identical to a reference or differs there from. Preferably, a value for a biomarker is deemed to differ from a reference if the observed difference is statistically significant which can be determined by statistical techniques referred to elsewhere in this description. If the difference is not statistically significant, the biomarker value and the reference are essentially identical. Based on the comparison referred to above, a subject can be assessed to suffer from the disease, or not.For the specific biomarkers referred to in this specification, preferred values for the changes in the relative amounts or ratios (i.e. the changes expressed as the ratios of the medians) are found in the Tables, below. Based on the ratios of the metabolites found in a subject suffering from pancreatic cancer and an apparently healthy control (blood donors) or a non-cancer group (blood donors, chronic pancreatitis and liver cirrhosis), respectively, and the calculated t-values as shown in Tables 2a, 2b, 3a, 3b, 4a, or 4b below, it can be derived whether an increase or a decrease of a given biomarker of Tables 2a, 2b, 3a, 3b, 4a, or 4b is indicative for the presence of pancreatic cancer. Negative t-values for a biomarker indicate that a decrease is indicative while positive t-values indicate that an increase of the biomarker is indicative for pancreatic cancer. It will be understood that the reference in said cases is derived from a subject or group of subjects known not to suffer from pancreatic cancer or is a calculated reference as defined elsewhere herein.The comparison is, preferably, assisted by automation. For example, a suitable computer program comprising algorithms for the comparison of two different data sets (e.g., data sets comprising the values of the characteristic feature(s)) may be used. Such computer programs and algorithms are well known in the art. Notwithstanding the above, a comparison can also be carried out manually.Advantageously, it has been found in the study underlying the present invention that the amounts of the specific biomarkers referred to above are indicators for pancreatic cancer. Accordingly, the at least one biomarker as specified above in a sample can, in principle, be used for assessing whether a subject suffers from pancreatic cancer, or not. This is particularly helpful for an efficient diagnosis of the disease as well as for improving of the pre-clinical and clinical management of pancreatic cancer as well as an efficient monitoring of patients. Moreover, the findings underlying the present invention will also facilitate the development of efficient drug-based therapies or other interventions against pancreatic cancer as set forth in detail below. The definitions and explanations of the terms made above apply mutatis mutandis for the following embodiments of the present invention except specified otherwise herein below.The present invention further contemplates a method for diagnosing pancreas cancer in a subject comprising the steps of:
(a) determining in a sample of a subject suspected to suffer from pancreas cancer the amount of at least one ceramide or the amoun and(b) comparing the said amount of the at least one biomarker with a reference, whereby pancreas cancer is to be diagnosed.
Preferably, said at least one ceramide is not ceramide (d18:1, C24:1).The sample in the aforementioned method has been, preferably, pre-treated by lipid fractionation.Lipid fractionation as used in this context refers to a process as, preferably, described in the accompanying Examples below. In particular, lipid fractionation can be achieved by extracting the total lipids from plasma by liquid/liquid extraction using chloroform/methanol. The lipid extracts obtained thereby are subsequently fractionated by normal phase liquid chromatography (NPLC) into eleven different lipid groups according to Christie (Journal of Lipid Research (26), 2). The fractions were analyzed by LC-MS/MS using electrospray ionization (ESI) and atmospheric pressure chemical ionization (APCI) with detection of specific multiple reaction monitoring (MRM) transitions for cholesterol esters (CE), free sterols (FS), sphingoymelins (SM), and ceramides (CER) respectively. Sphingosines and sphingosine-1-phosphates (SP) were analyzed by LC-MS/MS using electrospray ionization (ESI) with detection of specific multiple reaction monitoring (MRM) transitions as described by Schmidt H et. al., Prostaglandins & other Lipid Mediators 81(2006), 162-170. The fractions are further analyzed by GC-MS after derivatization with TMSH (Trimethyl sulfonium hydroxide), yielding the fatty acid methyl esters (FAME) corresponding to the acyl moieties of the class-separated lipids. The concentrations of FAME from C14 to C24 are determined in each fraction.For the aforementioned method of the invention, either the amount of at least one ceramide is used as a biomarker or the total ceramide amount. Preferably, the amount of at least one sphinogemyelin or the amount of total sphingomyelins is determined.The present invention further contemplates a method for diagnosing pancreas cancer in a subject comprising the steps of:
(a) determining in a sample of a subject suspected to suffer from pancreas cancer the amount of at least one biomarker of Tables 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32; and(b) comparing the said amount of the at least one biomarker with a reference, whereby pancreas cancer is to be diagnosed.
Preferably, the said sample applied in the aforementioned method is a plasma sample and the at least one metabolite is from table 6, 7 or 8. Also preferably, the said sample in the aforementioned method is a serum sample and the at least one metabolite is from table 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20. Moreover, preferably, the sample is a serum or plasma sample and the at least one metabolite is from table 21, 22, 23, 24, 25 or 26, 27, 28, 29, 30, 31 or 32.Preferably, the reference is derived from a subject or a group of subjects which are apparently healthy, e.g. blood donors. More preferably, the at least one biomarker in such a case is from table 9, 10, 11, 21, 22 or 23. Most preferably, an increase in the at least one biomarker is indicative for pancreatic cancer, if the said at least one biomarker is from table 10 or 22, whereas a decrease of the at least one biomarker is indicative for pancreatic cancer, if the said biomarker is from table 11 or 23.Preferably, the reference is derived from a subject or a group of subjects which exhibit symptoms of pancreatic cancer but which do not suffer from pancreatic cancer. Such critical control subjects, preferably, suffer from pancreatitis and/or liver cirrhosis. More preferably, the at least one biomarker in such a case is from table 12, 13, 14, 24, 25 or 26. Most preferably, an increase in the at least one biomarker is indicative for pancreatic cancer, if the said at least one biomarker is from table 13 or 25, whereas a decrease of the at least one biomarker is indicative for pancreatic cancer, if the said biomarker is from table 14 or 26. Accordingly, when determining at least one biomarker of tables 12, 13, 14, 24, 25 or 26, the method of the invention can be applied for differentiating in a subject between pancreatic cancer and a disease of the critical controls, i.e. pancreatitis and/or liver cirrhosis.Preferably, the reference is derived from a subject or a group of subjects which suffer from pancreatitis. More preferably, the at least one biomarker in such a case is from table 6, 7, 8, 15, 16, 17, 27, 28 or 29. Most preferably, an increase in the at least one biomarker is indicative for pancreatic cancer, if the said at least one biomarker is from table 7, 16 or 28, whereas a decrease of the at least one biomarker is indicative for pancreatic cancer, if the said biomarker is from table 8, 17 or 29. Accordingly, when determining at least one biomarker of tables 6, 7, 8, 15, 16, 17, 27, 28 or 29, the method of the invention can be applied for differentiating in a subject between pancreatic cancer and pancreatitis.Preferably, the reference is derived from a subject or a group of subjects which suffer from liver cirrhosis. More preferably, the at least one biomarker in such a case is from table 18, 19, 20, 30, 31 or 32. Most preferably, an increase in the at least one biomarker is indicative for pancreatic cancer, if the said at least one biomarker is from table 19 or 31, whereas a decrease of the at least one biomarker is indicative for pancreatic cancer, if the said biomarker is from table 20 or 32. Accordingly, when determining at least one biomarker of tables 18, 19, 20, 30, 31 or 32, the method of the invention can be applied for differentiating in a subject between pancreatic cancer and liver cirrhosis.The present invention also relates to a method for health care management comprising the steps of the methods of the present invention and the further step of recommending and/or applying a health care measure to the subject if pancreatic cancer is diagnosed.Health care management as used herein refers to governing the application of suitable health care measures to a subject according to its individual needs. Health care management is decisive for estimating the need for hospitalization and/or ambulant patient management.The term “health care measure” as used herein, accordingly, refers to any measure which is applied to a subject within the context of managing its disease. Health care measures, preferably, encompass recommendations, and the application thereof, on monitoring measures and/or monitoring frequencies. Moreover, health care measures include recommendations, and the application thereof, on hospitalization (i.e. the admission to a hospital) or ambulant care or support. Health care measures also include life style recommendations aiming to improve the disease situation and the application thereof as well as therapeutic measures such as the recommendation and/or the application of therapies referred to elsewhere herein in detail.The present invention also relates to a method for identifying whether a subject is in need for a therapy of pancreatic cancer or a change of therapy comprising the steps of the methods of the present invention and the further step of identifying a subject in need if pancreatic cancer is diagnosed.The phrase “in need for a therapy of pancreatic cancer” as used herein means that the disease in the subject is in a status where therapeutic intervention is necessary or beneficial in order to ameliorate or treat pancreatic cancer or the symptoms associated therewith. Accordingly, the findings of the studies underlying the present invention do not only allow diagnosing pancreatic cancer in a subject but also allow for identifying subjects which should be treated by a pancreatic cancer therapy or whose pancreatic cancer therapy needs adjustment. Once the subject has been identified, the method may further include a step of making recommendations for a therapy of pancreatic cancer.A therapy of pancreatic cancer as used in accordance with the present invention, preferably, comprises surgery, radiotherapy or drug treatment. Preferred surgery-based therapies include resection of the pancreas or parts thereof such as pancreaticoduodenectomy, tail pancreatectomy. total or partial pancreatoctomy, palliative bridging procedures. Drug-based therapies, preferably, include the administration of one or more drugs with antitumour properties including but not exclusive to platinum derivatives, fluoropyrimidines, pyrimidine analogues, Gemcitabine, antimetabolites, alkylating agents, anthracyclines, plant alkaloids, topoisomerase inhibitors, targeted antibodies and tryosine kinase inhibitors.The present invention further relates to a method for determining whether a therapy against pancreatic cancer is successful in a subject comprising the steps of the methods of the present invention and the further step of determining whether a therapy is successful if no pancreatic cancer is diagnosed.It is to be understood that a pancreatic cancer therapy will be successful if pancreatic cancer or at least some symptoms thereof are treated or ameliorated compared to an untreated subject. Moreover, a therapy is also successful as meant herein if the disease progression can be prevented or at least slowed down compared to an untreated subject.The present invention also relates to a device or system for diagnosing pancreas cancer in a sample of a subject comprising: (a) an analyzing unit for the said sample of the subject comprising a detector for at least one biomarker of Tables 2a, 2b, 3a, 3b, 4a, or 4b or at least one biomarker of Tables 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 said detector allowing for the determination of the amount of the said at least one bio and operatively linked thereto,(b) an evaluation unit comprising a data processing unit and a data base, said data base comprising a stored reference, preferably a reference as specified above in connection with the method of the invention and, more preferably, a reference derived from a subject or group of subjects known to suffer from pancreas cancer, and said data processing unit having tangibly embedded an algorithm for carrying out a comparison, preferably as specified above in connection with the method of the invention, of the amount of the at least one biomarker determined by the analyzing unit and the stored reference and for generating an output information based on which the diagnosis can be established. A device as used herein shall comprise at least the aforementioned units. The units of the device are operatively linked to each other. How to link the means in an operating manner will depend on the type of units included into the device. For example, where the detector allows for automatic qualitative or quantitative determination of the biomarker, the data obtained by said automatically operating analyzing unit can be processed by, e.g., a computer program in order to facilitate the assessment in the evaluation unit. Preferably, the units are comprised by a single device in such a case. Said device may accordingly include an analyzing unit for the biomarker and a computer or data processing device as evaluation unit for processing the resulting data for the assessment and for stabling the output information. Preferred devices are those which can be applied without the particular knowledge of a specialized clinician, e.g., electronic devices which merely require loading with a sample. The output information of the device, preferably, is a numerical value which allows drawing conclusions on the presence or absence of pancreatic cancer and, thus, is an aid for diagnosis. More preferably, the output information is a preliminary diagnosis or an aid for diagnosis based on the aforementioned numerical value, i.e. a classifier which indicates whether the subject suffers from pancreatic cancer or not. Such a preliminary diagnosis may need the evaluation of further information which can be provided in the device of the invention by including an expert knowledge database system.A preferred reference to be used as a stored reference in accordance with the device of the present invention is an amount for the at least one biomarker to be analyzed or values derived therefrom which are derived from a subject or group of subjects known to suffer from pancreatic cancer. In such a case, the algorithm tangibly embedded, preferably, compares the determined amount for the at least one biomarker with the reference wherein an identical or essentially identical amount or value shall be indicative for the presence of pancreatic cancer in the subject.Alternatively, another preferred reference to be used as a stored reference in accordance with the device of the present invention is an amount for the at least one biomarker to be analyzed or values derived therefrom which are derived from a subject or group of subjects known not to suffer from pancreatic cancer. In such a case, the algorithm tangibly embedded, preferably, compares the determined amount for the at least one biomarker with the reference wherein an amount or value which differs from the reference shall be indicative for the presence of pancreatic cancer in the subject. Preferred differences are those indicated as relative changes or degrees of changes for the individual biomarkers in the Tables below.The units of the device, also preferably, can be implemented into a system comprising several devices which are operatively linked to each other. Depending on the units to be used for the system of the present invention, said means may be functionally linked by connecting each mean with the other by means which allow data transport in between said means, e.g., glass fiber cables, and other cables for high throughput data transport. Nevertheless, wireless data transfer between the means is also envisaged by the present invention, e.g., via LAN (Wireless LAN, W-LAN). A preferred system comprises means for determining biomarkers. Means for determining biomarkers as used herein encompass means for separating biomarkers, such as chromatographic devices, and means for metabolite determination, such as mass spectrometry devices. Suitable devices have been described in detail above. Preferred means for compound separation to be used in the system of the present invention include chromatographic devices, more preferably devices for liquid chromatography, HPLC, and/or gas chromatography. Preferred devices for compound determination comprise mass spectrometry devices, more preferably, GC-MS, LC-MS, direct infusion mass spectrometry, FT-ICR-MS, CE-MS, HPLC-MS, quadrupole mass spectrometry, sequentially coupled mass spectrometry (including MS-MS or MS-MS-MS), ICP-MS, Py-MS or TOF. The separation and determination means are, preferably, coupled to each other. Most preferably, LC-MS and/or GC-MS are used in the system of the present invention as described in detail elsewhere in the specification. Further comprised shall be means for comparing and/or analyzing the results obtained from the means for determination of biomarkers. The means for comparing and/or analyzing the results may comprise at least one databases and an implemented computer program for comparison of the results. Preferred embodiments of the aforementioned systems and devices are also described in detail below.Furthermore, the present invention relates to a data collection comprising characteristic values of at least one biomarker being indicative for a medical condition or effect as set forth above (i.e. diagnosing pancreatic cancer in a subject, identifying whether a subject is in need for a therapy of pancreatic cancer or determining whether a pancreatic cancer therapy is successful).The term “data collection” refers to a collection of data which may be physically and/or logically grouped together. Accordingly, the data collection may be implemented in a single data storage medium or in physically separated data storage media being operatively linked to each other. Preferably, the data collection is implemented by means of a database. Thus, a database as used herein comprises the data collection on a suitable}

我要回帖

更多关于 fgxsx什么意思 的文章

更多推荐

版权声明:文章内容来源于网络,版权归原作者所有,如有侵权请点击这里与我们联系,我们将及时删除。

点击添加站长微信