WO2010079253A2 - Bio-markers for diagnosing fibrosis - Google Patents

Bio-markers for diagnosing fibrosis Download PDF

Info

Publication number
WO2010079253A2
WO2010079253A2 PCT/ES2010/070007 ES2010070007W WO2010079253A2 WO 2010079253 A2 WO2010079253 A2 WO 2010079253A2 ES 2010070007 W ES2010070007 W ES 2010070007W WO 2010079253 A2 WO2010079253 A2 WO 2010079253A2
Authority
WO
WIPO (PCT)
Prior art keywords
fibrosis
slurpl
hamp
gsn
fragment
Prior art date
Application number
PCT/ES2010/070007
Other languages
Spanish (es)
French (fr)
Other versions
WO2010079253A3 (en
Inventor
Fernando José CORRALES IZQUIERDO
Joaquín FERNÁNDEZ IRIGOYEN
Leticia Odriozola Moncayola
Jesús María PRIETO VALTUEÑA
Enrique SANTAMARÍA MARTÍNEZ
Original Assignee
Proyecto De Biomedicina Cima, S.L.
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 Proyecto De Biomedicina Cima, S.L. filed Critical Proyecto De Biomedicina Cima, S.L.
Publication of WO2010079253A2 publication Critical patent/WO2010079253A2/en
Publication of WO2010079253A3 publication Critical patent/WO2010079253A3/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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/08Hepato-biliairy disorders other than hepatitis
    • G01N2800/085Liver diseases, e.g. portal hypertension, fibrosis, cirrhosis, bilirubin

Definitions

  • the invention relates to the field of diagnosis and, more particularly, to the diagnosis of diseases associated with an increase in deposition of connective tissue in an organ or tissue, preferably in the liver.
  • the diagnosis is carried out using biomarkers whose level of expression correlates with the appearance of f ⁇ brotic diseases.
  • Hepatic fibrosis is a central feature of most chronic liver lesions due to metabolic, genetic, viral, and cholestatic diseases.
  • Chronic inflammatory diseases of the liver cause destruction of the parenchyma of the liver and its exchange for scar tissue (fibrosis).
  • Fibrosis is characterized by excess extracellular matrix deposition (ECM) that involves the molecular and histological reorganization of various types of collagen, proteoglycans, structural glycoproteins and hyaluronic acid (hyaluronan).
  • ECM extracellular matrix deposition
  • Fibrosis is a brand of liver cirrhosis, which is associated with impaired liver function and significant morbidity and mortality.
  • liver fibrosis has been made in the past and is currently practiced mostly through the aggressive puncture biopsy procedure and consecutive histological evaluation based on several numerical scoring systems that produce graduation of necroinflammatory activity and staging (extension) of fibrosis.
  • this "gold standard” has many disadvantages in addition to aggressiveness such as sampling error, irreproducible quality of the sample depending on the length and size of the tissue sample and a histological evaluation strictly dependent on the pathologist's experience.
  • liver fibrosis uses a panel of 5 markers: ⁇ 2-macroglobulin, haptoglobulin, apolipoprotein Al, ⁇ -glutamyl transpeptidase, and bilirubin (Imbert-Bismut et al. Lancet 2001; 357: 1069- 1075 and WO0216949).
  • this test eliminates the need for biopsy in only 26% of patients and does not accurately predict the presence or absence of fibrosis.
  • WO0373822 provides a method of diagnosing the presence or severity of liver fibrosis in an individual based on the presence or level of ⁇ -MG, HA and TIMP-1.
  • WO2005116901 discloses several methods to diagnose the presence and / or severity of a liver pathology based on different combinations of markers such as ⁇ -2 macroglobulin, hyaluronic acid, apoliprotein Al, N-terminal propeptide of type III collagen, ⁇ -glutamyltranspeptidase, bilirubin , ⁇ -globulins, platelets, prothrombin time, aspartate amino transferase, alanine aminotransferase, urea, sodium, glycemia, triglycerides, albumin, alkaline phosphatases, YKL-40 (human cartilage glycoprotein 39), Tissue matrix metalloproteinase inhibitor 1 (TIMP-I), matrix 2 metalloproteinase (MMP-2) and ferritin.
  • markers such as ⁇ -2 macroglobulin, hyaluronic acid, apoliprotein Al, N-terminal propeptide of type III collagen, ⁇ -glutamyltranspeptida
  • EPl 811041 discloses methods for the diagnosis of fibrosis and / or liver cirrhosis based on the use of markers identified in a rat model for liver fibrosis (rats treated with dimethylnitrosamine) using mRNA expression profiles in micromatrices and quantitative protein profiles of samples of liver and serum.
  • WO2007003670 discloses methods for the diagnosis of fibrotic alterations by using any of four detectable markers in urine: uromodulin, MAC2BP, AGP 1 and cathepsin A.
  • WO2008031051 discloses a method for the detection and diagnosis of liver fibrosis using a panel of human serum biomarker proteins. These methods have also been disclosed in Gangadharan et al. [Clinical Chemistry 2007; 53: 1792-1799].
  • liver fibrosis Another non-aggressive alternative approach for the evaluation of liver fibrosis is transient elastography with Fibroscan® (Echosens, Paris, France) that operates by measuring liver stiffness (Sandrin L et al. Ultrasound Med. Biol. 2003; 29: 1705- 1711). A mechanical pulse is generated on the surface of the skin, which spreads through the liver. The speed of the wave is measured by ultrasound. Speed correlates directly with the stiffness of the liver, which in turn reflects the degree of fibrosis - the stiffer the liver is, the greater the degree of fibrosis.
  • liver fibrosis Although there is a significant need for non-aggressive systemic biomarkers, organ-specific and disease for liver fibrosis (and fibrosis of other organs as well), there is currently no individual parameter or combination of them available, which satisfies all the diagnostic criteria required for extended, cost effective, and reliable use. Individual measurements of biochemical markers in serum, plasma or even urine are currently not valid enough to replace liver biopsy.
  • the invention relates to the use of at least one biomarker for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis in a subject where said biomarker is selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
  • the invention in a second aspect, relates to a method for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis that comprises comparing the expression of one or more biomarkers in a sample of a subject with a predetermined standard for each of said one or more biomarkers; wherein said one or more biomarkers are selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2; and wherein a significant difference in the expression of said one or more biomarkers in said sample compared to the predetermined standard of each of said one or more biomarkers is indicative of onset, phase, evolution of fibrosis and / or disease associated with fibrosis or of the efficacy of a treatment for fibrosis and / or disease associated with fibrosis.
  • the invention relates to a kit comprising reagents for detecting at least two or more biomarkers selected from the group of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
  • Control sample GeI Stains marked with circles were collected and identified by LC-MS / MS.
  • the spot in the white circle was identified as gelsolin, and the spot in the black circle was identified as apolipoprotein D.
  • FIG. 1 Decreased levels of ApoD and gelsolin proteins identified in cirrhotic urine samples were confirmed by immunoblot.
  • A The Urine proteins were separated on an SDS-acrylamide gel and the proteins were transferred on a nitrocellulose membrane. ApoD and gelsolin levels were detected using antibodies specific for these proteins.
  • B The intensity of the bands was quantified and represented. The average value of protein levels showed a two-fold decrease in cirrhotic samples for both proteins.
  • FIG. 3 The intensity of hepcidin-25 (peak of 2793 Da), hepcidin-20 (peak of 2193) and SLURP-I (peak of 8855 Da) for cirrhotic, control and low fibrosis samples are represented.
  • the levels of this protein are significantly decreased in cirrhotic patients, on the other hand the low fibrosis group shows an intermediate expression level.
  • Figure 4 Decreased levels of the SLURPl protein in urine samples of cirrhotic patients with respect to the controls detected by Western Blot.
  • A The control urine (C1-C8) and eight urine from cirrhotic patients (P1-P8) were subjected to SDS-acrylamide gel electrophoresis and the proteins were transferred on a nitrocellulose membrane. SLURPl levels were detected using antibodies specific for these proteins.
  • B Graph of the optical density corresponding to the SLURP-I bands obtained in the control urine and in cirrhotic patients.
  • FIG. 5 The intensities of Osteopontin (7655 Da peak) and its phosphorylated form (7735 Da peak) for cirrhotic, control and low fibrosis samples are depicted. The levels of this protein are significantly decreased in cirrhotic patients, on the other hand the low fibrosis group shows an intermediate expression level.
  • the authors of the present invention have identified protein biomarkers in human urine samples whose expression correlates with the presence of cirrhosis and liver fibrosis.
  • the authors have identified gelsolin, apoliprotein D, hepcidin (25 and 20), SLURP-I, Osteopontin (SPPl, OPN or OSTP), Beta 1 Defensin
  • the invention relates to the use of a biomarker for the detection, diagnosis and evaluation of fibrosis and / or of a disease involving fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or of a disease associated with fibrosis in a subject
  • said biomarker is selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
  • biomarker or, alternatively, “molecular marker”, as used herein, is used to refer to a molecule or the expression product of a gene or fragments and variants thereof that show substantial changes in a given disease. and that can be used both for the detection and for the diagnosis of a disease by detecting the appearance of said changes in the biomarker and / or to follow the efficacy of a treatment for that disease by detecting changes in the biomarker as opposed to those that occur in the disease or clinical situation.
  • changes in the biomarker are changes in expression levels.
  • expression refers to a process by which a polypeptide is produced from DNA. This process involves the transcription of the gene to a messenger RNA (mRNA), and the translation of this mRNA into the polypeptide.
  • mRNA messenger RNA
  • changes in the levels of expression refers to any change in the production of mRNA, polypeptide or both that produces altered relative levels of mRNA, protein or both in a sample with respect to other molecules in the same sample.
  • the expression levels of a biomarker can be determined by determining the levels of mRNA in a sample or by determining the levels of the corresponding polypeptide
  • the polypeptide biomarkers can be variants resulting from post-translational modifications, including fragments thereof.
  • diagnosis refers to the assessment of the probability according to which a subject suffers from a disease as well as the assessment of its onset, developmental state , evolution, or its regression, and / or the prognosis of the course of the disease in the future.
  • assessment although preferred, may not be correct for 100% of the subjects to be diagnosed.
  • the term requires that a statistically significant part of the subjects can be identified as having the disease or having a predisposition to it.
  • a part is statistically significant, it can be determined by the person skilled in the art using several well-known statistical evaluation tools, for example, determination of confidence intervals, determination of p-values, 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%, at least 95%.
  • P values are preferably 0.2, 0.1, 0.05.
  • the expression "follow the effectiveness of a treatment” refers to the assessment of the results of a therapy in a patient suffering from fibrosis or a disease associated with fibrosis.
  • Suitable therapies for the treatment of fibrosis include treatment with PCP inhibitors, IFN- ⁇ or imatinib mesylate (Gleevec).
  • the usefulness of the biomarkers of the invention to track the efficacy of a treatment can also be applied to methods for selecting and screening drugs with potential activity.
  • This process comprises a) administering to the subject (preferably an animal) the drug to be studied; b) at different points of the study (before, during and / or after administration) take biological samples of the animal and determine the marker levels according to the present invention; and c) compare the determinations made in the samples obtained in the different phases of the treatment and compare them to control animals, for example untreated.
  • SLURP-I refers to the gene known as "containing the secreted domain LY6 / PLAUR 1", also identified by the accession number HGNC ID. 18746 (HUGO Gene Nomenclature Committee, Human Genome Organization), which is located in chromosomal region 8q24.3.
  • the protein encoded by SLURP-I is identified in the Entrez database by GeneID access number: 57152 (updated July 27, 2008) and is also known as "ARS component B” (ARS, ArsB), " urinary antineoplastic protein "(ANUP), MDM,” similar to secreted lymphocyte antigen 6 (LY6LS).
  • the expression product of the SLURP1 gene comprises SEQ ID NO: 1, which is a 103 amino acid polypeptide identified with the UniProt ID accession number P55000 (last modified July 22, 2008. Version 75) (SEQ ID NO: 1) as well as orthologs, isoforms and fragments of said polypeptide.
  • the biomarker is a fragment of said polypeptide comprising amino acids 22-103 (SEQ ID NO: 2), corresponding to the secreted mature polypeptide.
  • HAMP refers to the "hepcidin antimicrobial peptide” gene, identified by the HGNC ID accession number. 15598, and which is located in chromosomal region 19ql3.1.
  • the "hepcidin antimicrobial peptide” refers to the "hepcidin antimicrobial peptide” gene, identified by the HGNC ID accession number. 15598, and which is located in chromosomal region 19ql3.1.
  • HAMP liver-expressed antimicrobial peptide
  • HEPC hepcidin
  • HFE2B hepcidin
  • LEAPl hepcidin
  • PLTR circulating liver tumor regressor
  • the expression product of the HAMP gene comprises any of the sequences SEQ ID NO: 3, which is the 84-amino acid preproprotein identified by the UniProt ID accession number. P81172 (last modified July 22, 2008.
  • HAMP expression product is Hepc25, Hepc20 or both.
  • GSN refers to the gene "gelsolin (amyloidosis, of the Finnish type), identified by accession number HGNC ID. 4620, and which is located in the chromosomal region 9q33.
  • the expression product of the gene is also identified in the Entrez database as GeneID: 2934 (updated September 28, 2008) or DKFZp313L0718.
  • the protein is known as gelsolin, and alternatively as "actin depolymerizing factor” (ADF), brevina or AGEL
  • ADF actin depolymerizing factor
  • brevina a transcriptional variant for GSN that encode different isoforms
  • the GSN expression product comprises any of the sequences SEQ ID NO: 6, which corresponds to the secreted precursor polypeptide identified by UniProt ID. P06396 (last modified October 14, 2008. Version 114) as well as orthologs, isoforms and fragments of said polypeptide.
  • APOD refers to the "apoliprotein D” gene, also identified by the HGNC accession number: 612, which is located in the chromosomal region 3q26.2-qter. APOD is also identified by "Entrez GeneID: 347" (updated October 12, 2008).
  • the APOD expression product comprises any of the sequences SEQ ID NO: 7, a precursor of the polypeptide identified as UniProt ID. P05090 (last modified July 22, 2008. Version 107) as well as orthologs, isoforms and fragments of said polypeptide.
  • SPPl refers to the gene "Osteopontin” or "phosphoprotein secreted type 1"; OPN, and known alternatively as BNSP; BSPI; ETA-I;
  • HGNC 11255 which is located in the 4q21-q25 chromosomal region.
  • SPPl too is identified by "Entrez Gene ID: 6696" (updated on December 4, 2009).
  • the SPP1 gene expression product comprises SEQ ID NO: 8, which is a 314 amino acid polypeptide identified with the UniProt accession number P 10451 (last modified November 24, 2009. Version 120); as well as orthologs, iso forms and fragments of said polypeptide, including the SPP1 isoforms identified as SEQ ID NO: 8, 9, 10 and 11.
  • DEFBl refers to the gene "Beta 1 defensin or Defensin beta 1" and alternatively known as BDl; HBDl; DEFB-I; DEFB101; MGC51822, corresponding to the polypeptide identified by HGNC accession number: 2766, which is located in chromosomal region 8p23.2-p23.1.
  • DEFBl is also identified by "Entrez GeneID: 1672" (updated on December 4, 2009).
  • the DEFBl expression product comprises the sequences SEQ ID NO: 9, identified as UniProt P60022.1 (last modified November 3, 2009. Version 72) as well as orthologs, isoforms and fragments of said polypeptide .
  • MASP2 refers to the "Manina-binding lectin serine peptidase 2" and "alternatively, sMAP; MAP 19; MASP-2, also identified by HGNC accession number: 6902, which is located in the chromosomal region Ip36.3-p36.2. MASP2 is also identified by "Entrez GeneID: 10747” (updated on December 4, 2009).
  • the SPP1 expression product comprises the sequence SEQ ID NO: 10, identified as UniProt O00187.3 (last modified November 24, 2009. Version 118); as well as orthologs, isoforms and fragments of said polypeptide, including the isoforms identified in SEQ ID NO: 13 and 14 of the present invention.
  • fibrosis refers to an excess of extracellular matrix deposition (ECM) that involves molecular and histological reorganization of various types of collagen, proteoglycans, structural glycoproteins and hyaluronic acid.
  • ECM extracellular matrix deposition
  • fibrosis is a mark of many liver diseases, Other organs may also suffer from excess deposition of connective tissue.
  • the fibrosis is hepatic fibrosis.
  • disease associated with fibrosis refers to any disease where one or more organs suffer from excessive deposition of fibrous connective tissue.
  • diseases include cystic fibrosis of the pancreas and lungs, injection fibrosis, which can occur as a complication of intramuscular injections, especially in children, endomyocardiac fibrosis, idiopathic pulmonary fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, progressive massive fibrosis, a complication of miner's pneumoconiosis, systemic nephrogenic fibrosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, tuberculosis (TB) can cause fibrosis of the lungs, sickle cell anemia can lead to increase and finally spleen fibrosis and rheumatoid arthritis and cirrhosis , which can cause liver fibrosis.
  • the invention also relates to a method for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis comprising comparing the expression of one or more biomarkers in a sample of a subject with a predetermined standard for each of said one or more biomarkers; wherein said one or more biomarkers are selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2; and wherein a significant difference in the expression of said one or more biomarkers in said sample compared to the predetermined standard of each of said one or more biomarkers is indicative of onset, phase, evolution of fibrosis and / or disease associated with fibrosis or of the efficacy of a treatment for fibrosis and / or disease associated with fibrosis.
  • the method of the invention allows the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis as well as monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis.
  • the method of the invention comprises determining the expression of one or more biomarkers selected from the group of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl, MASP2 in a biological sample of an individual.
  • sample means any biological tissue or fluid taken from a subject.
  • biological liquid for example, blood, serum or, more preferably urine.
  • biomarker of the invention can be evaluated by any of a wide variety of well known methods to detect the expression of a transcribed molecule or its corresponding protein. If the biomarkers are nucleic acid molecules, expression of the marker polynucleotide can be detected using nucleic acid hybridization methods, nucleic acid reverse transcription methods, nucleic acid amplification methods and the like.
  • expression of a marker gene is evaluated by preparing mRNA / cDNA (i.e., a transcribed polynucleotide) of cells in a sample of a patient, and by hybridizing the mRNA / cDNA with a reference polynucleotide that It is complementary to a polynucleotide comprising the marker gene, and fragments thereof.
  • the cDNA can, optionally, be amplified using any of several polymerase chain reaction methods before hybridization with the reference polynucleotide although it is preferred that it is not amplified.
  • the biomarkers are polypeptides, in which case detection can be carried out using immunological methods for the detection of secreted proteins, protein purification methods, protein function or activity assays.
  • the expression of a marker protein is evaluated using an antibody (eg, a radiolabeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), a antibody derivative (for example, an antibody conjugated to a substrate or to the protein or ligand of a protein-ligand pair (e.g., biotin-streptavidin), or an antibody fragment (e.g., a single chain antibody, the hypervariable domain of an isolated antibody, etc.) that specifically binds with a protein corresponding to the marker gene, such as the protein encoded by the open reading frame corresponding to the marker gene or such a protein that has undergone all or part of its modification normal post-translational
  • ELISA adsorption enzyme immunoassay
  • an antibody for example anti-enzyme
  • a solid phase ie, to a microtiter plate
  • antigen for example, enzyme
  • the solid phase is then washed to remove unbound antigen.
  • a labeled antibody for example, enzyme bound
  • enzymes that can bind to the antibody are alkaline phosphatase, horseradish peroxidase, luciferase, urease, and B-galactosidase.
  • the enzyme-bound antibody reacts with a substrate to generate a colored reaction product that can be measured.
  • the antibody is incubated with a sample containing antigen (ie, enzyme).
  • a sample containing antigen ie, enzyme
  • the antigen-antibody mixture is then contacted with a solid phase (for example, a microtiter plate) that is coated with antigen (i.e., enzyme).
  • antigen i.e., enzyme
  • the more antigen present in the sample the less free antibody will be available to bind to the solid phase. It is added after a labeled secondary antibody (eg, enzyme bound) to the solid phase to determine the amount of primary antibody bound to the solid phase.
  • a labeled secondary antibody eg, enzyme bound
  • a section of tissue is tested for specific proteins by exposing the tissue to antibodies that are specific to the protein being tested.
  • the antibodies are then visualized by any of a number of methods to determine the presence and amount of the protein present. Examples of the methods used to visualize antibodies are, for example, by means of enzymes bound to the antibodies (for example, luciferase, alkaline phosphatase, horseradish peroxidase, or beta-galactosidase), or chemical methods (for example, DAB chromogen /substratum).
  • the sample is then analyzed microscopically, most preferably by optical microscopy of a sample stained with a dye that is detected in the visible spectrum, using any of a variety of such methods and staining reagents known to the person skilled in the art.
  • radioimmunoassays can be used.
  • a radioimmunoassay is a technique to detect and measure the concentration of an antigen using a labeled form (for example, radioactively or fluorescently labeled) of the antigen.
  • radioactive labels for antigens include 3 H, 14 C, and 125 I.
  • the concentration of antigen enzyme in a biological sample is measured by causing the antigen in the biological sample to compete with the labeled antigen (eg radioactively) for binding. to an antibody to the antigen.
  • the labeled antigen is present in a concentration sufficient to saturate the antibody binding sites.
  • the concentration of the antigen in the sample the lower the concentration of the labeled antigen that will bind to the antibody.
  • the antigen-antibody complex must be separated from the free antigen.
  • One method of separating the antigen-antibody complex from the free antigen is to precipitate the antigen-antibody complex with anti-isotype antiserum.
  • Another method of separating the antigen-antibody complex from the free antigen is to precipitate the antigen-antibody complex with S. aureus annihilated with formalin.
  • Yet another method to separate the Antigen-antibody complex of the free antigen is to perform a "solid phase radioimmunoassay" where the antibody is bound (for example, covalently) to Sepharose beads, polystyrene wells, polyvinylchloride wells, or microtiter wells.
  • concentration of labeled antigen bound to the antibody can be determined.
  • An “immunoradiometric assay” is an immunoassay in which the antibody reagent is radioactively labeled.
  • An IRMA requires the production of a multivalent antigen conjugate, by techniques such as conjugation to a protein, for example, rabbit serum albumin (RSA).
  • the multivalent antigenic conjugate must have at least 2 antigen residues per molecule and the antigen residues must be separated at a sufficient distance to allow the binding of at least two antibodies to the antigen.
  • the multivalent antigen conjugate may be attached to a solid surface such as a plastic sphere.
  • sample antigen and antibody for the antigen that is radioactively labeled are added to a test tube containing the sphere coated with the multivalent antigen conjugate.
  • the antigen in the sample competes with the multivalent antigen conjugate for the binding sites of the antibody to the antigen.
  • unbound reagents are removed by washing and the amount of radioactivity in the solid phase is determined. The amount of radioactive antibody bound is inversely proportional to the concentration of antigen in the sample.
  • biomarker's protein levels in a biological sample can be performed according to the preferences of the practitioner, and based on the present disclosure and the type of biological sample (i.e., plasma, urine, sample of tissue etc.).
  • One such technique is immunoblotting (Towbin et al., Proc. Nat. Acad. Sci. 76: 4350 (1979)), where a sample treated in a suitable manner is run on an SDS-PAGE gel before being transferred to a solid support, such as a nitrocellulose filter.
  • Detectable labeled anti-enzyme antibodies can then be used to assess enzyme levels, where the intensity of The detectable brand signal corresponds to the amount of enzyme present.
  • the levels can be quantified, for example by densitometry.
  • the levels of biomarkers as disclosed herein, and / or their polypeptides can be detected in a tissue sample by mass spectrometry such as MALDI / TOF (flight time), SELDI / TOF, liquid chromatography.
  • mass spectrometry such as MALDI / TOF (flight time), SELDI / TOF, liquid chromatography.
  • LC-MS mass spectrometry
  • GC-MS gas chromatography-mass spectrometry
  • HPLC-MS high performance liquid chromatography-mass spectrometry
  • capillary electrophoresis-mass spectrometry for example MS / MS, MS / MS / MS, ESI-MS / MS, etc.
  • tandem mass spectrometry for example MS / MS, MS / MS / MS, ESI-MS / MS, etc.
  • Mass spectrometry methods are well known in the art and have been used to quantify and / or identify biomolecules, such as proteins (see, for example, Li et al. (2000) Tibtech 18: 151-160; Rowley et al . (2000) Methods 20: 383-397; and Kuster and Mann (1998) Curr. Opin. Structural Biol. 8: 393-400).
  • mass spectrometry techniques have been developed that allow at least partial de novo sequencing of isolated proteins. Chait et al., Science 262: 89-92 (1993); Keough et al, Proc. Nati Acad. Sci. USA. 96: 7131-6 (1999); reviewed in Bergman, EXS 88: 133-44 (2000).
  • a gas phase ion spectrometer is used.
  • laser desorption / ionization mass spectrometry is used to analyze the sample.
  • Modern laser desorption / ionization (“LDI-MS”) mass spectrometry can be carried out in two main variations: matrix-assisted laser desorption / ionization mass spectrometry (“MALDI”) and surface-enhanced laser desorption / ionization (“SELDI").
  • MALDI matrix-assisted laser desorption / ionization mass spectrometry
  • SELDI surface-enhanced laser desorption / ionization
  • the surface of the substrate is modified so that it is an active participant in the desorption process.
  • the surface is derived with an adsorbent and / or capture reagents that selectively bind to the protein of interest.
  • the surface is derived with molecules that absorb energy that are not desorbed when they are reached by the laser.
  • the surface is derived with molecules that bind to the protein of interest and that contain a photolytic bond that breaks after laser application.
  • the bypass agent is generally located at a specific location on the surface of the substrate where the sample is applied. See, for example, US Pat. No. 5719060 and WO 98/59361.
  • the two methods can be combined by, for example, the use of a SELDI affinity surface to capture an analyte and adding a matrix containing liquid to the captured analyte to provide the energy absorbing material.
  • biomarker levels will typically depend on the detection of signal strength. This, in fact, may reflect the amount and character of a polypeptide bound to the substrate. For example, in certain embodiments, the strength of the peak value signal of the spectra of a first sample and a second sample (for example, visually, by computer analysis, etc.) can be compared, to determine the quantities relative of particular biomolecules. Software programs such as the Biomarker Wizard program (Ciphergen Biosystems, Inc., Fremont, Calif.) Can be used as an aid in the analysis of mass spectra. Mass spectrometers and their techniques are well known to those skilled in the art. Alternatively, the levels of a polypeptide biomarker can be determine obtaining a spectrometry spectrum of mass by laser desorption / ionization increased by surface flight time (SELDI-TOF MS) of the sample.
  • SELDI-TOF MS surface flight time
  • a decrease in expression with the reference value is considered where the difference between the test sample and the reference sample of at least 0.9 times, 0.75 times, 0.2 times, 0.1 times, 0 , 05 times, 0.025 times, 0.02 times, 0.01 times, 0.005 times or even less.
  • the reference value is the level of expression of the biomarker in a reference sample that is obtained by combining equal amounts of samples from a population of subjects.
  • typical reference samples will be obtained from subjects that are clinically well documented and free of the disease.
  • normal (reference) concentrations of the biomarker can be determined, for example by providing the average concentration over the reference population.
  • considerations are taken into account. Among such considerations are the type of sample involved (eg tissue or CSF), age, weight, sex, general physical condition of the patient and the like. For example, equal amounts of a group of at least 2, at least 10, at least 100 to preferably more than 1000 subjects are taken as reference group, preferably classified according to the above considerations, for example of various age categories.
  • the reference value used to determine whether a therapy is effective is usually the level of expression of the biomarker or biomarkers under consideration with a patient sample before the start of therapy.
  • the variation with respect to the reference value is an increase in the expression above of the reference value.
  • the level of expression is considered to be increased where the difference between the test sample and the reference sample is at least 1.1 times,
  • the method of the invention can be carried out by determining the level of a variable number of biomarkers.
  • the level (s) of a biomarker, two or more biomarkers, three or more biomarkers or four or more biomarkers as defined in the present invention may allow greater sensitivity and specificity in the diagnosis of fibrosis and / or a disease associated with fibrosis or may allow better differentiation of fibrosis from other diseases that may have similar or overlapping biomarkers.
  • the present invention comprises the simultaneous determination of the following groups of biomarkers:
  • SLURPl and MASP2 SLURPl and HAMP fragment, SLURPl and Hepc25 fragment, SLURPl and Hepc20 fragment, SLURPl and GSN fragment, SLURPl and APOD fragment, SLURP 1 and SPP 1 fragment, SLURPl and DEFBl fragment, SLURPl and MASP2 fragment; HAMP and Hepc25,
  • Hepc20 and GSN Hepc20yAPOD,
  • SLURPl 5 GSN and MASP2 SLURPl 5 HAMP and Hepc25 fragment; SLURP 1, HAMP and Hepc20 fragment; SLURPl 5 HAMP and GSN fragment; SLURPl 5 HAMP and APOD fragment; SLURPl 5 HAMP and SPPl fragment; SLURPl 5 HAMP and DEFBl fragment; SLURP 1, HAMP and MASP2 fragment; SLURPl 5 Hepc25 and Hepc20 fragment; SLURPl 5 Hepc25 and GSN fragment; SLURPl, Hepc25 and APOD fragment; SLURPl fragment, Hepc25 and SPPl SLURPl fragment, Hepc25 and DEFBl SLURPl fragment, Hepc25 and MASP2 SLURP fragment 1, Hepc20 and GSN; SLURPl, Hepc20 and APOD fragment; SLURPl fragment, Hepc20 and SPPl SLURPl fragment, Hepc25 and DEFBl SLURPl fragment, Hepc25
  • HAMP Hepc25 and Hepc20
  • HAMP Hepc25 and GSN
  • HAMP Hepc25 and MASP2
  • HAMP Hepc20 and GSN
  • HAMP HAMP, Hepc20 and MASP2; HAMP, GSN and APOD;
  • Hepc25, GSN and DEFBl Hepc25, GSN and MASP2;
  • SLURPl SLURPl fragment, Hepc20 and GSN
  • SLURP 1 SLURP 1 fragment, Hepc20 and APOD
  • SLURPl, HAMP, GSN and MASP2 SLURP1, HAMP, Hepc25 and Hepc20 fragment; SLURPl, HAMP, Hepc25 and GSN fragment; SLURPl, HAMP, Hepc25 and APOD fragment; SLURP 1, HAMP, Hepc25 and SPP 1 fragment; SLURPl, HAMP, Hepc25 and DEFBl fragment; SLURPl, HAMP, Hepc25 and MASP2 fragment; SLURPl, HAMP, Hepc20 and GSN fragment; SLURPl, HAMP, Hepc20 and APOD fragment; SLURP 1, HAMP, Hepc20 and SPP 1 fragment; SLURPl, HAMP, Hepc20 and DEFBl fragment; SLURPl, HAMP, Hepc20 and MASP2 fragment; SLURPl, HAMP, GSN and APOD fragment; SLURPl, HAMP, GSN and APOD fragment; SLURPl, HAMP, GSN and APOD
  • HAMP Hepc25, Hepc20 and GSN
  • HAMP Hepc25, Hepc20 and APOD
  • HAMP Hepc25, Hepc20 and SPPl
  • HAMP Hepc25, Hepc20 and DEFBl
  • HAMP Hepc25, Hepc20 and MASP2;
  • HAMP Hepc25, Hepc20, GSN and SPPl
  • HAMP Hepc25, Hepc20, GSN and DEFBl
  • HAMP Hepc25, Hepc20, GSN and MASP2;
  • HAMP HAMP, Hepc20, GSN and APOD
  • SLURPl SLURPl, HAMP, Hepc25, Hepc20 and SPPl fragment
  • SLURP fragment SLURP 1
  • HAMP Hepc25, Hepc20 and DEFB 1
  • SLURPl SLURPl fragment, Hepc25, Hepc20, GSN and MASP2; SLURP 1, SLURP 1 fragment, HAMP, Hepc20, GSN and APOD;
  • the methods of the invention can be combined with other methods known in the art for the detection of fibrosis or a disease associated with fibrosis and for the determination of the efficacy of a therapy. for fibrosis or a disease associated with fibrosis.
  • the biomarkers of the invention can be measured simultaneously with the series of 5 biomarkers defined in WO0216949 ( ⁇ 2-macroglobulin, haptoglobulin, apolipoprotein Al, ⁇ -glutamyl transpeptidase, and bilirubin), with the series of three biomarkers described in WO0373822 ( ⁇ -MG, HA and TIMP-I), with the different series of biomarkers described in WO2005116901 (comprising ⁇ -2 macroglobulin, hyaluronic acid, apolipoprotein Al, N-terminal propeptide of type III collagen, ⁇ -glutamyltranspeptidase, bilirubin, ⁇ -globulins, platelets, prothrombin time, aspartate amino transferase, alanine aminotransferase, urea, sodium, glycemia, triglycerides, albumin, alkaline phosphatases, YKL-40 (human carti
  • the method of the invention can be used together with other methods to detect fibrosis not based on the determination of expression levels of one or more biomarkers, such as liver biopsy and elastography as described in Sandrin L et al. (Ultrasound Med. Biol. 2003; 29: 1705-1711).
  • kits comprising reagents for detecting at least two or more biomarkers selected from the group of SLURPl, HAMP, GSN ,, APOD, SPPl, DEFBl, MASP2.
  • biomarkers are polypeptides and thus, the reagents that form the kit can be any compound capable of binding with high affinity to the above polypeptide biomarkers.
  • the kit reagents can be, without limitation, any type of antibody or immunoglobulin molecule or fragment thereof such as polyclonal, monoclonal, human or humanized or recombinant antibodies as well as single chain antibodies, for example scFv constructs, or synthetic antibodies as such and that they can belong to any of the following classes of immunoglobulins: IgG, IgM, IgE, IgA, and where where applicable, a subclass of any of the aforementioned classes, for example subclasses of the IgG class such as IgGl, IgG2, IgG2a, IgG2b, IgG3 or IgGM.
  • any type of antibody or immunoglobulin molecule or fragment thereof such as polyclonal, monoclonal, human or humanized or recombinant antibodies as well as single chain antibodies, for example scFv constructs, or synthetic antibodies as such and that they can belong to any of the following classes of immunoglobulins: IgG, IgM,
  • the kit may also comprise antibody fragments such as Fv, Fab or F (ab ') 2 fragments or single chain fragments such as scFv. Double chain fragments such as Fv, Fab or F (ab ') 2 are preferred.
  • Fab and F (ab ') 2 fragments have no Fc fragment contained in intact antibodies. As a beneficial consequence, such fragments are transported faster in the circulatory system, and show less non-specific binding to tissue compared to complete antibody species.
  • Such fragments can be produced from intact antibodies by proteolytic digestion using proteases such as papain (for the production of Fab fragments) or pepsin (for the production of F (ab ') 2 fragments), or chemical oxidation.
  • the antibodies can be bound to a solid support forming antibody matrices or protein chips.
  • Protein matrices are solid phase ligand binding assay systems that use immobilized proteins on surfaces that include glass, membranes, microtiter wells, mass spectrometer plates, and balls or other particles.
  • the matrices are highly parallel (multiplexed) and with miniaturized frequencies (microarrays, protein chips). Its advantages include that they are fast and automatable, capable of great sensitivity, economical in reagents, and give plenty of data for a single experiment. Bioinformatic support is important; Data management demands sophisticated software and data comparison analysis. However, the software adapts to that used for DNA matrices, such as much of the hardware and detection systems.
  • the capture matrix in which ligand binding reagents are used, which are normally antibodies but which can also alternatively be protein skeletons, peptides or nucleic acid aptamers, to detect target molecules in mixtures such as plasma or tissue extracts.
  • ligand binding reagents which are normally antibodies but which can also alternatively be protein skeletons, peptides or nucleic acid aptamers, to detect target molecules in mixtures such as plasma or tissue extracts.
  • capture matrices are used to perform multiple immunoassays in parallel, testing both for several analytes in individual sera for example and testing many serum samples simultaneously.
  • proteomics the Capture matrices are used to quantify and compare protein levels in different samples in health and disease, that is, protein expression profile.
  • Different proteins are used than those that bind specific ligands in the matrix format for in vitro functional screening such as protein-protein, protein-DNA, protein-drug, receptor-ligand, enzyme-substrate, etc.
  • the capture reagents themselves are selected and screened against many proteins, optionally in multiplex matrix format against many target proteins.
  • protein sources include cell-based expression systems for recombinant proteins, purification of natural sources, in vitro production by cell-free translation systems, and synthetic methods for peptides. Many of these methods are automated for high performance production.
  • capture matrices and protein function analysis it is important that the proteins are correctly folded and functional; This is not always the case, for example, where recombinant bacteria proteins are extracted under denaturing conditions.
  • denatured protein matrices are useful in the screening of antibodies for cross-reactivity, identification of autoantibodies and selection of ligand-binding proteins.
  • Protein matrices have been designed as a miniaturization of familiar immunoassay methods such as ELISA and drop transfer, often using fluorescent readers, and facilitated by robotics and high performance detection systems to allow multiple tests to be carried out in parallel.
  • Physical supports include glass slides, silicone, microwells, nitrocellulose or PVDF membranes, and magnetic microbeads and others. While the most familiar format is protein droplets that leave on planar surfaces, alternative architectures include microfluidic-based CD centrifugation devices (Gyros, Monmouth Junction, NJ) and specialized chip designs, such as built-in microchannels on a plate (for example, The Living Chip TM, Biotrove, Woburn, MA) and tiny 3D posts on a silicone surface (Zyomyx, Hayward CA).
  • Suspended particles are also used as a base for matrices, provided they are coded for identification;
  • the systems include color codes for microballs (Luminex, Austin, TX; Bio-Rad Laboratories), semiconductor nanocrystals (e.g., QDOTS TM, Quantum Dot, Hayward, CA), ball barcode (ULTRAPLEX TM balls, SmartBead Technologies Ltd, Babraham, Cambridge, RU) and multimetallic microbeads (for example, NANOBARCODES TM particles, Nanoplex Technologies, Mountain View, CA).
  • the balls are optionally assembled in planar matrices on semiconductor chips (LEAPS TM technology, BioArray Solutions, Warren, NJ).
  • Protein immobilization involves both the coupling reagent and the nature of the surface to which it is attached.
  • a good protein matrix support surface is chemically stable before and after coupling procedures, allows good spot morphology, shows minimal non-specific binding, does not contribute to the background in detection systems, and is compatible with different detection systems .
  • the immobilization method used is reproducible, applicable to proteins of different properties (size, hydrophilic, hydrophobic), manageable at high performance and automation, and compatible with the retention of fully functional protein activity.
  • the orientation of the surface bound protein is recognized as an important factor when presenting it to the ligand or substrate in an active state; for capture matrices the most effective binding results are obtained with oriented capture reagents, which generally require site specific protein marking.
  • Both covalent and non-covalent methods of protein immobilization are used and have several pros and cons. Passive adsorption to surfaces is methodologically simple, but allows little quantitative or orientation control. It may or may not alter the functional properties of the protein, and the reproducibility and efficacy are variable. Covalent coupling methods provide stable binding, apply to a range of proteins and have good reproducibility. However, the orientation is variable. In addition, chemical shunt can alter protein function and requires a stable interactive surface. Biological capture methods that use a tag on the protein provide stable binding and bind to the protein specifically and in reproducible orientation, but the biological reagents must first be properly immobilized, and the matrix can require special handling and have variable stability.
  • Substrates for covalent bonding include glass slides coated with silane reagents containing amino or aldehyde.
  • silane reagents containing amino or aldehyde In the VERSALINX TM system (Prolinx, Bothell, WA), reversible covalent coupling is achieved by interaction between the protein derived with phenyldiboronic acid and salicylic hydroxamic acid immobilized on the support surface. This also has low background binding and low intrinsic fluorescence and allows immobilized proteins to retain function.
  • Non-covalent binding of unmodified proteins occurs with porous structures such as HYDROGEL TM (PerkinElmer, Wellesley, MA), based on a three-dimensional polyacrylamide gel; It has been described that this substrate gives a particularly low background in glass microarrays, and high capacity and retention of protein function.
  • the widely used biological coupling methods are through biotin / streptavidin or hexahistidine / Ni interactions, having modified the protein appropriately.
  • Biotin can be conjugated to a polylysine skeleton immobilized on a surface such as titanium dioxide (Zyomyx, Inc., Hayward, CA) or tantalum pentoxide (Zeptosens, Witterswil, Switzerland).
  • Matrix manufacturing methods include robotic contact printing, inkjet, piezoelectric tapping and photolithography.
  • a number of commercial matrix manufacturers are available [for example, Packard Biosciences, Affymetrix Inc. and Genetix] as well as manual equipment [for example, V and P Scientific].
  • Bacteria colonies are optionally placed in robotic grids on PVDF membranes for induction of protein expression in situ.
  • spot size and density are the nanomatrices, with spots on the nanometer space scale, which allow thousands of reactions to be carried out on a single chip of less than 1 mm square.
  • BioForce Nanosciences Inc. and Nanolink Inc. for example, have developed commercially available nanomatrices.
  • the methods of marking and fluorescence detection are widely used.
  • the same instruments that are used for reading DNA microarrays are applicable for protein matrices.
  • capture matrices for example, antibodies
  • fluorescently labeled proteins from two different cell states, in which the used cell phones are they conjugate directly with different fluorophores (for example, Cy-3, Cy-5) and mix, so that the color acts as a reader for changes in the abundance of the target.
  • the fluorescent reading sensitivity is amplified 10-100 times by tyramide signal amplification (TSA) (PerkinElmer Lifesciences).
  • TSA tyramide signal amplification
  • Planar guided wave technology Zeptosens
  • High sensitivity is achieved with the suspension of balls and particles, using phycoerythrin as a brand (Luminex) or the properties of semiconductor nanocrystals (Quantum Dot).
  • Phycoerythrin as a brand
  • Quantum Dot the properties of semiconductor nanocrystals
  • a number of alternative readers have been developed, especially in the area of commercial biotechnology. These include nuclear plasmon resonance adaptations (HTS Biosystems, Intrinsic Bioprobes, Tempe, AZ), rolling circle DNA amplification (Molecular Staging, New Haven, CT), mass spectrometry (Intrinsic Bioprobes; Ciphergen, Fremont, CA), resonance light scattering (Genicon Sciences, San Diego, CA) and atomic force microscopy [BioForce Laboratories].
  • HTS Biosystems Intrinsic Bioprobes, Tempe, AZ
  • rolling circle DNA amplification Molecular Staging, New Haven, CT
  • mass spectrometry Intrinsic Bioprobes; Ciphergen
  • Capture matrices form the basis of diagnostic chips for the expression profile. They employ high affinity capture reagents, such as conventional antibodies, individual domains, constructed skeletons, peptides or nucleic acid aptamers, to bind and detect specific target ligands in a high yield manner.
  • Antibody matrices have the required specificity and acceptable background properties, and some are commercially available (BD Biosciences, San Jose, CA; Clontech, Mountain View, CA; BioRad; Sigma, St. Louis, MO). Antibodies to capture matrices are produced either by conventional immunization (polyclonal sera and hybridomas), or as recombinant fragments, normally expressed in E. coli, after selection of phage or ribosome display libraries (Cambridge Antibody Technology, Cambridge , RU; Biolnvent, Lund, Sweden; Affitech, Walnut Creek, CA; Biosite, San Diego, CA).
  • skeleton refers to ligand-binding protein domains, which are constructed in multiple variants capable of binding to various target molecules with specificity and affinity properties similar to antibody. Variants are produced in a genetic library format and selected against individual targets by phage, bacteria or ribosome presentation. Such skeletons or ligand-binding frameworks include Affybodies based on S.
  • aureus protein A (Affibody, Bromma, Sweden), Trronectins based on fibronectins (Phylos, Lexington, MA) and Anticalins based on lipocalin structure (Pieris Proteolab , Freising- Weihenstephan, Germany). These are used in capture matrices in a manner similar to antibodies and have the advantages of consistency and ease of production.
  • Non-protein capture molecules notably single chain nucleic acid aptamers that bind protein ligands with high specificity and affinity
  • matrices SomaLogic, Boulder, CO
  • the aptamers are selected from oligonucleotide libraries by the Selex TM method (SomaLogic, Boulder, CO) and their interaction with proteins is increased by covalent binding, by the incorporation of brominated deoxyuridine and UV-activated cross-linking (photoaptamers). Photo-cross-linking to ligands reduces cross-reactivity of aptamers due to specific steric requirements.
  • Aptamers have the advantages of ease of production by automated oligonucleotide synthesis and DNA stability and consistency; in the photoaptamer matrices, universal fluorescent protein dyes are used to detect binding.
  • Protein analytes that bind to antibody matrices are detected directly or indirectly, for example, through a secondary antibody.
  • Direct marking is used to compare different samples with different colors.
  • sandwich immunoassays provide high specificity and sensitivity and are therefore the method of choice for low abundance proteins such as cytokines; They also give the possibility of detection of protein modifications.
  • Unmarked detection including mass spectrometry, surface plasmon resonance and atomic force microscopy, prevents ligand alteration. What is required of any method is optimal sensitivity and specificity, with a low background to give a signal at high noise. Since analyte concentrations cover a wide range, sensitivity has to be adapted appropriately.
  • the proteins of interest are often those at low concentrations in body fluids and extracts, which require detection in the range of pg or less, such as cytokines or low-expression products in cells.
  • An alternative to a matrix of capture molecules is one made by means of molecular fingerprint technology, in which peptides (for example, from C-terminal regions of proteins) are used as templates to generate structurally complementary, specific cavities sequence in a polymerizable matrix; the cavities can then specifically capture (denatured) proteins that have the appropriate primary amino acid sequence (ProteinPrint TM, Aspira Biosystems, Burlingame, CA).
  • ProteinChip® matrix (Ciphergen, Fremont, CA), in which solid-phase chromatographic surfaces bind to proteins with similar loading or hydrophobicity characteristics of mixtures such as plasma or tumor extracts, and SELDI-TOF mass spectrometry is used for the detection of retained proteins.
  • Large-scale functional chips have been constructed by immobilizing large numbers of purified proteins and are used to test a wide range of biological functions, such as protein interactions with other proteins, drug-target, enzyme-substrate interactions, etc. They generally require an expression library, cloned in E. coli, yeast or the like from which the expressed proteins are purified, for example, through a His tag and immobilized. Transcription / translation of cell-free proteins is a viable alternative for the synthesis of proteins that do not express well in bacterial systems or other systems in vivo.
  • protein matrices are alternatives in vitro to the system of the two cell-based yeast hybrids and are useful where the latter is deficient, such as interactions involving secreted proteins or proteins with disulfuric bonds.
  • the high-performance analysis of biochemical activities in matrices for yeast protein kinases and for various functions (protein-protein and protein-lipid interactions) of yeast proteome has been described, where a large proportion of all open reading frames was expressed of yeast and immobilized in a microarray. Large-scale proteome chips are also useful in identifying functional interactions, drug screening, etc. (Proteometrix, Branford, CT).
  • a protein matrix is used to screen phage display libraries or ribosomes, to select binding partners, including antibodies, synthetic skeletons, peptides and aptamers.
  • binding partners including antibodies, synthetic skeletons, peptides and aptamers.
  • a library vs. library screening is carried out.
  • the screening of candidate drugs in combinatorial chemical libraries against a matrix of target proteins identified from genomic projects is another application of this approach.
  • Multiplexed ball assays use a series of spectrally discrete particles that are used to capture and quantify soluble analytes. The analyte is then measured by detection of a fluorescence based emission and flow cytometric analysis. Multiplexed ball assays generate data that is comparable to ELISA based assays, but in a multiplexed or simultaneous manner. The concentration of strangers for the cytometric ball matrix is calculated as with any sandwich test, that is, through the use of known standards and representing the unknown ones against the standard curve. In addition, multiplexed ball assays allow quantification of soluble analytes in samples never before considered due to sample volume limitations.
  • the kit of the invention may additionally contain instructions for use in determining the protein levels of the biomarkers present in the sample. These instructions can be found in the form of printed material or in the form of electronic support that can store instructions so that they can be read by a subject, such as electronic storage media (optical discs, tapes and the like), optical media (CD- ROM, DVD) and the like.
  • the media may additionally or alternatively contain Internet pages that provide such instructions.
  • Urine samples were from 35 patients with F4 cirrhosis (Metavir index), 10 patients with Fl fibrosis (Metavir index), and 35 healthy controls.
  • the criteria used to classify liver damage was liver biopsy for patients with cirrhosis and fibrosis, and standard blood tests for control individuals. 81.5% of the patients were men and 18.5% women and all were between 38 and 75 years old.
  • Urine samples were centrifuged at 4000xg for 5 minutes at 4 0 C, and 30 ml supernatants were concentrated using centrifugal filter devices
  • Triplicate analyzes were performed alternating fluorescent tide of study samples.
  • the first dimension was performed on an Ettan IPGphor (GE Healthcare) using Immobiline DryStrips pH3-l l NL, 24 cm (GE Healthcare).
  • the samples were loaded at the acid end of the strip with a cup loading device.
  • the isoelectric focusing conditions were those recommended by the manufacturer for the type of strip used.
  • the SDS-PAGE (12.5%) was developed in an Ettan DALTsix (GE Healthcare) at 25 0 C and lW / gel for 12 hours.
  • the images of the gels were captured with a Typhoon Trio (GE Healthcare) at a resolution of 100 ⁇ m with ⁇ ex / ⁇ em of 488/520, 532/580, and 633/670 nm for Cy2, Cy3, and Cy5 respectively.
  • Image analysis and statistics were performed with DeCyder Differential version 6.5 software (GE Healthcare) using the Biological Variation Analysis (BVA) module. Only differences with p ⁇ 0.05 (t test) were accepted.
  • the preparative gels were run with 400 ⁇ g of protein following the same procedure indicated above.
  • the proteins were visualized by staining with SYPRO Ruby protein gel staining (Bio-Rad) and the images were acquired with a Typhoon Trio using ⁇ ex / ⁇ em 532/560 nm. Differentially represented spots were cut manually and gel samples were processed with a MassPrep (Waters) station. Triptych digestion in the gel was performed with 12.5 ng / ⁇ l trypsin in 50 mM ammonium bicarbonate for 12 hours at 37 ° C. Microcapillary reverse phase CL was performed with a CapLC TM (Waters) capillary system.
  • Reverse phase separation of the tryptic digested was performed with a fused silica capillary column Atlantis, C 18, 3 ⁇ m, 75 ⁇ m x 10 cm Nano Ease TM (Waters) balanced in 5% acetonitrile, 0.2% formic acid .
  • the column was washed for 5 minutes with the same buffer and the peptides were eluted using a linear gradient of 5-50% acetonitrile in 45 minutes at a constant flow rate of 0.2 ⁇ l / min.
  • the column was coupled in line to a Q-TOF Micro (Waters) using a PicoTip nanospray ionization source (Waters).
  • the temperature of the heated capillary was 8O 0 C and the potential difference of the spray was 1.8-2.2 kV.
  • MS / MS data was collected in an automated mode dependent on the data. The three most intense ions in each study were sequentially fractionated by collision-induced dissociation (CID) using an isolation width of 2.0 and a relative collision energy of 35%. Data processing was performed with MassLynx 4.0. The database search was done with Phenyx 2.2 (GeneBio, Geneva, Switzerland) against the Uniprot Relay 12.3 knowledge base consisting of UniprotKB / Swiss-Prot Relay 54.3 and UniprotKB / TrEMBL Relay 37.3 with entries 285.335 and 4.932. 421, respectively.
  • search was enzymatically restricted for trypsin and a failed cut site was allowed. Additional search parameters were as follows: no restriction on molecular weight and isoelectric point; fixed modification, cistern carbamidomethylation; variable modification, methionine oxidation.
  • protein solutions are applied to ProteinChip matrix stains, which have been derived with planar chromatographic chemicals.
  • the proteins actively interact with the surface of the chromatographic matrix, and are sequestered according to their potential for surface interaction as well as separated from salts and other contaminants in the sample by subsequent washing in the stain with the appropriate buffer solutions.
  • the proteins to be analyzed are co-crystallized with UV absorbing components and volatilized by a pulsed UV laser beam. The ionized proteins are then accelerated in an electric field, and the mass to charge ratios of the different ionic species of the proteins can be deduced from their velocity.
  • binding buffers were used for each specific type of chip: shallow CMlO buffer (0.1 M sodium acetate, pH 4.0); H50 buffer (the surface was initially loaded with 50% ACN and binding was performed in 10% ACN, 0.1% TFA); QlO buffer (100 mM Tris HCl pH 9); IMAC30 buffer (the surface was initially loaded with 0.1 M cupric sulfate, neutralized with 0.1 M sodium acetate pH 4.0, and the binding was performed in 0.1 M sodium phosphate, 0 sodium chloride , 5 M pH 7.0).
  • the SELDI-TOF profile was performed as follows: the chips were equilibrated twice with 150 ⁇ l of the appropriate binding buffer for 5 minutes and loaded twice (30 minutes each load) with 25 ⁇ l of sample mixed with 175 ⁇ l of buffer of Union. Unbound proteins were removed by washing the chips three times with 150 ⁇ l of binding buffer and three times with 200 ⁇ l of deionized water. After drying the charged surface, 1 ⁇ l of synapine acid (Bio-Rad, Hercules, CA) dissolved in 50% ACN and 0.5% TFA was applied twice and allowed to dry. The analysis of bound proteins / peptides was performed using ProteinChip System, Series 4000 (Bio-Rad, Hercules, CA).
  • the spectra were collected in positive ion mode between the mass range of 1-100 kDa with a focus mass of 48 kDa and 8 kDa.
  • the obtained spectra were processed, normalized and compared with the Ciphergen Express, a data group analysis program, using the default parameters.
  • the statistical analysis chosen was the univariate non-parametric U-Mann-Whitney test.
  • linear models were used using the LIMMA Bioconductor package that allowed the selection of peptides using a correction of the p-value based on FDR or the B statistic (logarithm of the quotient probability that a peptide is differentially expressed / probability of not being differential).
  • the discriminant capacity of the selected peptides was evaluated with the ROC curve (sensitivity versus specificity) and particularly with the AUC value (area under the curve). Once the most reliable individual markers were established, a multivariate analysis was performed to evaluate the combination that provides the most effective classification of the samples.
  • Urine samples were mixed with 5x loading buffer (250 mM Tris-HCl pH 6.8, 10% SDS, 50% glycerol, 5% mercaptoethanol, 62.5 mM EDTA, 0 bromine phenol blue, 1%) and 1.2 ⁇ g of protein were loaded and run on a 12.5% sodium glycine-dodecyl sulfate-polyacrylamide gel. After electrophoresis, the proteins were transferred to a nitrocellulose membrane by wet transfer (100 V at 4 0 C for 1 hour).
  • PBST phosphate buffered saline plus Tween
  • the membranes were incubated with a polyclonal rabbit anti-gelsolin antibody (Santa Cruz Biotechnology, Santa Cruz, California), (diluted 1: 1000) and rabbit polyclonal antibody apolipoprotein D (Santa Cruz Biotechnology, Santa Cruz, California) in PBST containing 1% milk, 4 0 C overnight.
  • PBST phosphate buffered saline plus Tween
  • the immunoreactive protein bands were visualized by incubating the membranes with anti-rabbit antibody conjugated to peroxidase (diluted 1: 5000 in 5% milk in PBST) for 1 hour at room temperature.
  • anti-rabbit antibody conjugated to peroxidase diluted 1: 5000 in 5% milk in PBST
  • immunoreactive bands were represented on a chemiluminescent film by normal film development.
  • Retained chromatography To test the proteins of interest during purification, the fractions profile was made in ProteinChip CMlO and / or QlO matrices. The matrices were first equilibrated in binding buffer (50 mM Tris, pH 9.0 for QlO matrices and 100 mM Na acetate, pH 4.0 for CMlO matrices). Aliquots of each sample were diluted 10 times in binding buffer and allowed to bind to the surface of the matrix for 30 minutes. The matrix was washed three times with 150 ⁇ l of binding buffer, rinsed with deionized water, and allowed to dry before adding matrix.
  • binding buffer 50 mM Tris, pH 9.0 for QlO matrices and 100 mM Na acetate, pH 4.0 for CMlO matrices.
  • NP20 matrices 1-2 ⁇ L aliquots were also bound to NP20 matrices and allowed to dry before adding the matrix.
  • SPA or CHCA were used as matrices.
  • Q HyperD® F resin (PaIl Corporation) was equilibrated with Ul buffer (1 M urea, 0.2% CHAPS, 50 mM Tris-HCl, pH 9) and incubated with each urine sample. Denatured urine samples were incubated in batches for 1 hour at room temperature with 800 ⁇ L of Q HyperD® F resin.
  • the unbound fraction was collected and the resin was washed with 1 mL of 50 mM Tris, pH 9 in 0.1 OGP %.
  • the bound proteins were eluted successively with 1 mL of buffers of pH 7, 6, 5, 4, and 3 in 0.1% OGP and finally with a solution of 33% isopropanol / 17% ACN / 0% TFA ,one%.
  • the profile of an aliquot of each fraction was made on ProteinChip CMlO and / or QlO matrices to be tested for the proteins of interest.
  • the profile of a 2 ⁇ L aliquot of each fraction was made in a ProteinChip NP20 matrix to determine the elution pattern of the markers of interest.
  • the aliquot profile of the selected fractions was also made in a ProteinChip CMlO matrix.
  • Fractionation using a proteominer system To enrich the 4.6 kDa peak before purification, fractionation was used as a preliminary step using the proteominer system: 1 ml of Proteominer resin in PBS was equilibrated, 40 ml of urine was added and incubated at 4 or C overnight in rotation. The unbound fraction was collected and the resin was transferred to a column for elution.
  • the bound fraction was eluted successively in 1 ml of 1) 2.2 M thiourea, 7.7 M urea, 4.4% CHAPS, 2) 9 M urea, 25 mM citric acid, pH 3.8 and 3) 33.3% isopropyl alcohol, 16.7% acetonitrile, 0.5% trifluoroacetic acid.
  • the column was incubated 10 minutes at room temperature in rotation before collecting each elution to ensure maximum efficiency.
  • IMAC-Cu fractionation IMAC HyperCel resin was loaded with 100 mM copper sulfate and equilibrated in PBS, 0.5 M NaCl. The samples were diluted with PBS / 0.5 M NaCl, and incubated with the resin at 4 ° C overnight in rotation. For the purification of the 4.6 kDa peak, the unbound fraction of the proteominer was diluted 1: 3 in PBS / 0.5 M NaCl and incubated with 1.5 ml of copper-charged resin. For the purification of peaks 5.8 and 7.6 / 7.7 kDa, the urine was diluted 1: 2 in PBS / 0.5 M NaCl.
  • Resin purification 18 Purification of the 5.8 kDa peptide includes a purification step by hydrophobic exchange in resin 18. This resin was equilibrated in binding buffer ((150 mM NaCl in 100 mM Sodium acetate pH 5) and incubated throughout the overnight at 4 o C. Patient urine and control urine was incubated with 100 ul and 200 ul of resin respectively 18.
  • the unbound fraction was collected and the resin bound proteins were eluted successively with 1.5 volumes of the following buffers : El) 4.5 M Urea, E2) 9 M urea, E3) 9 M urea, 0.6% ammonium hydroxide, E4) 9 M urea, 1.2% ammonium hydroxide, and E5) 9 M urea, 2.4% ammonium hydroxide. Fractions from elution with ammonium hydroxide were neutralized with 5M acetic acid. An aliquot of each fraction was diluted 1: 10 and its peptide profiles were analyzed on CMlO chips.
  • Analytical extraction The selected protein bands were cut from the polyacrylamide gel with a pasteur pipette and extracted to confirm the m / z of the protein and prepare the sample for digestion.
  • the pieces of gels were washed twice with 200 ⁇ L of 50% methanol / 10% acetic acid for 20 minutes, dehydrated with 100 ⁇ L of ACN for 15 minutes, and extracted with 70 ⁇ L of 50% formic acid / 25% ACN / 15% IPA incubating 2 hours at room temperature with vigorous stirring.
  • the profile of a 1-2 ⁇ L aliquot of the extract was made on a ProteinChip NP20 matrix.
  • the 8.85 kDa marker was reduced and rented after gel extraction.
  • the analytical extract was dried in a CentriVap centrifuge concentrator (Labconco), rinsed twice with ammonium hydroxide, and dried.
  • the precipitate was then resuspended in 10 .mu.l of 2.5 mM DTT, 50 mM ammonium bicarbonate pH 8 and incubated at 60 0 C for 15 minutes. An excess of iodoacetamide was then added to the reaction. and incubated for 15 minutes in the dark. The alkylation reaction was quenched with an excess of DTT for 15 minutes in the dark. Before digestion with trypsin, this sample was dried again in the CentriVap.
  • the samples were reduced and rented before purification.
  • DTT was added to the samples at a concentration of 5 mM and incubated for 1 hour at 22 ° C. An excess of iodoacetamide was then added to the reaction and incubated for 15 minutes in the dark. The alkylation reaction was quenched with an excess of DTT for 15 minutes in the dark. Digestion in solution for trypsin digested, alkylated dried gels extracts were rehydrated with 20 ⁇ L of 50 mM ammonium bicarbonate (pH 8) containing 5 ng / ⁇ L of modified trypsin (Roche Applied Science) and incubated for 3 hours at 37 0 C.
  • the cut bands were treated to remove Coomassie stain and SDS, incubating successively with 400 ⁇ l of 50% methanol / 10% acetic acid (one hour for the first incubation and 30 minutes for the second) and with 400 ⁇ l of acetonitrile 50% / 100 mM pH8 ammonium bicarbonate for 30 minutes, and with 100 ⁇ l of acetronitrile for 10 minutes.
  • the gel bands were dried on a CentriVap evaporator. For digestion, the dried bands were rehydrated in 20 ⁇ l of 50 mM pH8 ammonium bicarbonate with, 5 ng / ⁇ l trypsin or 10 ng / ⁇ l AspN.
  • DIGE gels were obtained from five healthy controls and five cirrhotic patients (Fig. 1). The spots were detected and compared using the DeCyder software with the independent T-test statistical analysis. In this analysis, eighteen differential spots were found with a P value of less than 0.05 and an average ratio greater than 1.5. Seventeen of these proteins decreased in cirrhotic patients and only one of them increased in patients. To identify the differential proteins, a preparative gel was made by combining several control samples, and the proteins were stained with the Sypro Ruby stain
  • the plasma depolymerizing gelsolin precursor f is a plasma secreted actin modulating protein that binds actin and f ybronectin. It can promote the assembly of monomers in filaments as well as separate filaments already formed.
  • SELDI TOF Two studies were performed using the SELDI-TOF technique. The first was carried out in the Spanish National Biotechnology Center (CNB), and the second in the Biomarker Research Center in the Bio-Rad laboratories.
  • the purpose of this study was to obtain additional information, increasing both the number of samples and the types of chips.
  • the number of samples was increased to 35 patients with cirrhosis and 35 healthy controls.
  • a group of 10 patients with low fibrosis (Metavir Fl index) was added.
  • the number of chips used also increased to four: CMlO (cation exchange), IMAC30 (metal affinity), QlO (anion exchange) and H5 (hydrophobic exchange).
  • the data analysis produced 61 peaks that showed statistically significant differences (P ⁇ 0.01) between cirrhotic patients and healthy controls.
  • the fibrosis group was too small for a consistent statistical analysis, but provided some information on the specificity of the markers, showing an intermediate behavior between the cirrhotic samples and controls (Figs. 3, 5 and 6).
  • This second study provided confirmation of four markers detected in the CNB analysis.
  • the peptide analysis performed in the Bio-Rad laboratories provides, first, confirmation of one of the peptides identified in the previous study, and second, new candidate markers identified by means of the QlO chip.
  • Table 1 lists those peptides identified in the two univariate statistical studies that showed significantly different levels of expression between controls and cirrhotic patients. The tabal indicates the protein that has been assigned to each peptide and the ROC p values obtained when discriminating between control and cirrhotic patients. Table 1
  • ROC curve A useful tool to evaluate the diagnostic capacity of a quantitative test is the so-called ROC curve. It will also help us compare different tests.
  • sensitivity proportion of true positives
  • specificity proportion of true negatives
  • Multivariate statistical analysis of the combination of markers 4623/7655/7735/8853/2193/2793/5804 produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.91.
  • Multivariate statistical analysis of the combination of markers 4623/7655 / / 8853/2193/2793/5804 produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.87.
  • Multivariate statistical analysis of the combination of markers 4623/7655/8853/2793 produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.87.
  • Hepcidin is translated as an 84 amino acid peptide and processed to obtain hepcidin-25 (C-terminal residues 60-84 of the peptide) or hepcidin-20 (C-terminal residues 65-84). Hepcidin has been shown to be involved in the maintenance of iron homeostasis. Regulates both intestinal iron absorption and iron storage in macrophages and hepatocytes. Hepcidin increases in response to increased iron levels and inflammation, and decreases in response to hypoxia, anemia and oxidative stress. In addition, hepcidin also has a strong antimicrobial activity against several microorganisms.
  • SLURP-I has been found to be a marker of late skin differentiation and has been shown to have antitumor activity. It is involved in the maintenance of the physiological and structural integrity of the keratinocyte layers of the skin, defects in SLURP-I are a cause of Meleda Mal.
  • the MASP2 protein is a serine protease that binds to the malignant binding lectin (MBL), causing activation of the complement pathway. MBL is able to bind with repeated structures of sugars present in a wide variety of bacteria and other microorganisms promoting their elimination. Stengaard et al. (New Eng. J. Med. 349: 554-560, 2003) have described that congenital deficiency in the gene encoding MASP2 results in a greater susceptibility to infections and immune diseases.
  • Beta-Defensins function as natural antibiotics that are found on the surface of the skin. They are active against bacteria, fungi and enveloped viruses. Beta-Defensins have 36 to 42 amino acids. Human beta-Defensin 1 (hBD-1) or DEFBl is constitutively expressed at the level of the urinary and respiratory genital tract. It is an antimicrobial peptide involved in the resistance of epithelial surfaces to microbial colonization. These peptides interact with several CDi and lymphocyte receptors, thereby activating adaptive immunity mechanisms.
  • Osteopontin is a multifunctional glycoprotein with a stimulating effect on fibroblasts and extracellular matrix synthesis. Osteopontin is found in various tissues and has various functions such as the biomineralization of bone tissue and tissue repair, fibrosis and dystrophic calcifications after immunological lesions, it also participates in tumor growth, in the development of cancer and metastasis. It also participates in the initiation of the immune response in the early activation of T lymphocytes and macrophages.

Abstract

The invention relates to methods for detecting and diagnosing fibrosis and diseases associated with fibrosis, and to methods for determining the effectiveness of a therapy against fibrosis by determining the expression levels of bio-markers, where the one or more bio-markers are selected from the group formed by SLURP1, HAMP, GSN and APOD. The invention also relates to kits comprising reagents suitable for carrying out the methods according to the invention.

Description

BIOMARCADORES PARA EL DIAGNOSTICO DE FIBROSIS BIOMARKERS FOR THE FIBROSIS DIAGNOSIS
CAMPO DE LA INVENCIÓNFIELD OF THE INVENTION
La invención se refiere al campo del diagnóstico y, más en particular, al diagnóstico de enfermedades asociadas con un aumento en la deposición de tejido conjuntivo en un órgano o tejido, preferiblemente en el hígado. El diagnóstico se lleva a cabo usando biomarcadores cuyo nivel de expresión se correlaciona con la aparición de enfermedades fϊbróticas.The invention relates to the field of diagnosis and, more particularly, to the diagnosis of diseases associated with an increase in deposition of connective tissue in an organ or tissue, preferably in the liver. The diagnosis is carried out using biomarkers whose level of expression correlates with the appearance of fϊbrotic diseases.
ANTECEDENTES TÉCNICOSTECHNICAL BACKGROUND
La fibrosis hepática es una característica central de la mayoría de las lesiones crónicas del hígado debidas a enfermedades metabólicas, genéticas, víricas, y colestásicas. Las enfermedades inflamatorias crónicas del hígado producen la destrucción del parénquima del hígado y su cambio por tejido cicatricial (fibrosis). La fibrosis se caracteriza por el exceso de deposición de matriz extracelular (MEC) que implica la reorganización molecular e histológica de varios tipos de colágenos, proteoglicanos, glicoproteínas estructurales y ácido hialurónico (hialuronano). La fibrosis es una marca de cirrosis hepática, que se asocia con perturbación de la función del hígado y morbilidad y mortalidad significativas.Hepatic fibrosis is a central feature of most chronic liver lesions due to metabolic, genetic, viral, and cholestatic diseases. Chronic inflammatory diseases of the liver cause destruction of the parenchyma of the liver and its exchange for scar tissue (fibrosis). Fibrosis is characterized by excess extracellular matrix deposition (ECM) that involves the molecular and histological reorganization of various types of collagen, proteoglycans, structural glycoproteins and hyaluronic acid (hyaluronan). Fibrosis is a brand of liver cirrhosis, which is associated with impaired liver function and significant morbidity and mortality.
La deposición de MEC en el espacio de Disse (fibrosis perisinusoidal), la generación de membranas básales subendoteliales (incompletas), y la estrangulación de hepatocitos por la matriz adyacente deterioran no sólo el flujo de sangre a través del órgano, sino también la función biosintética de los hepatocitos y la capacidad de depuración de estas y otras células, por ejemplo de factores de coagulación y proteínas de transporte en el plasma, hormonas, y amoniaco. De esta manera, el diagnóstico, seguimiento y evaluación terapéutica de la fϊbrosis y fϊbrogénesis, es decir, el proceso activo de la generación de tejido conjuntivo nuevo en el hígado enfermo, es de gran importancia clínica.The deposition of ECM in the space of Disse (perisinusoidal fibrosis), the generation of subendothelial (incomplete) basal membranes, and the strangulation of hepatocytes by the adjacent matrix impair not only blood flow through the organ, but also biosynthetic function of hepatocytes and the purification capacity of these and other cells, for example of coagulation factors and transport proteins in plasma, hormones, and ammonia. In this way, the diagnosis, follow-up and therapeutic evaluation of phobrosis and phogenesis, that is, the active process of generating new connective tissue in the diseased liver, is of great clinical importance.
El diagnóstico de la fϊbrosis hepática se ha hecho en el pasado y se practica actualmente mayoritariamente mediante el procedimiento agresivo de biopsia por punción y evaluación histológica consecutiva basada en varios sistemas de puntuación numérica que producen graduación de actividad necroinflamatoria y estadificación (extensión) de fϊbrosis. Sin embargo, este "estándar de oro" tiene muchos inconvenientes además de la agresividad tal como error de muestreo, calidad irreproducible de la muestra dependiendo de la longitud y tamaño de la muestra de tejido y una evaluación histológica estrictamente dependiente de la experiencia del patólogo.The diagnosis of liver fibrosis has been made in the past and is currently practiced mostly through the aggressive puncture biopsy procedure and consecutive histological evaluation based on several numerical scoring systems that produce graduation of necroinflammatory activity and staging (extension) of fibrosis. However, this "gold standard" has many disadvantages in addition to aggressiveness such as sampling error, irreproducible quality of the sample depending on the length and size of the tissue sample and a histological evaluation strictly dependent on the pathologist's experience.
Actualmente, el método serológico más fiable para la evaluación de fibrosis hepática utiliza un panel de 5 marcadores: α2-macroglobulina, haptoglobulina, apolipoproteína Al, γ-glutamil transpeptidasa, y bilirrubina (Imbert-Bismut et al. Lancet 2001; 357:1069-1075 y WO0216949). Sin embargo, esta prueba elimina la necesidad de biopsia en sólo el 26% de los pacientes y no predice de forma exacta la presencia o ausencia de fibrosis.Currently, the most reliable serological method for the evaluation of liver fibrosis uses a panel of 5 markers: α2-macroglobulin, haptoglobulin, apolipoprotein Al, γ-glutamyl transpeptidase, and bilirubin (Imbert-Bismut et al. Lancet 2001; 357: 1069- 1075 and WO0216949). However, this test eliminates the need for biopsy in only 26% of patients and does not accurately predict the presence or absence of fibrosis.
WO0373822 proporciona un método de diagnóstico de la presencia o gravedad de fibrosis hepática en un individuo basado en la presencia o nivel de α-MG, HA y TIMP- 1.WO0373822 provides a method of diagnosing the presence or severity of liver fibrosis in an individual based on the presence or level of α-MG, HA and TIMP-1.
WO2005116901 divulga varios métodos para diagnosticar la presencia y/o gravedad de una patología hepática basados en diferentes combinaciones de marcadores tal como α-2 macroglobulina, ácido hialurónico, apoliproteína Al, propéptido N-terminal de colágeno de tipo III, γ-glutamiltranspeptidasa, bilirrubina, γ-globulinas, plaquetas, tiempo de protrombina, aspartato amino-transferasa, alanina aminotransferasa, urea, sodio, glucemia, triglicéridos, albúmina, fosfatasas alcalinas, YKL-40 (glicoproteína de cartílago humana 39), Inhibidor tisular de metaloproteinasa de matriz 1 (TIMP-I), metaloproteinasa de matriz 2 (MMP-2) y ferritina. EPl 811041 divulga métodos para el diagnóstico de fibrosis y/o cirrosis hepática basados en el uso de marcadores identificados en un modelo de rata para fibrosis hepática (ratas tratadas con dimetilnitrosamina) mediante perfiles de expresión de ARNm en micromatrices y perfiles cuantitativos de proteínas de muestras de hígado y suero.WO2005116901 discloses several methods to diagnose the presence and / or severity of a liver pathology based on different combinations of markers such as α-2 macroglobulin, hyaluronic acid, apoliprotein Al, N-terminal propeptide of type III collagen, γ-glutamyltranspeptidase, bilirubin , γ-globulins, platelets, prothrombin time, aspartate amino transferase, alanine aminotransferase, urea, sodium, glycemia, triglycerides, albumin, alkaline phosphatases, YKL-40 (human cartilage glycoprotein 39), Tissue matrix metalloproteinase inhibitor 1 (TIMP-I), matrix 2 metalloproteinase (MMP-2) and ferritin. EPl 811041 discloses methods for the diagnosis of fibrosis and / or liver cirrhosis based on the use of markers identified in a rat model for liver fibrosis (rats treated with dimethylnitrosamine) using mRNA expression profiles in micromatrices and quantitative protein profiles of samples of liver and serum.
WO2007003670 divulga métodos para el diagnóstico de alteraciones fibróticas mediante el uso de cualquiera de cuatro marcadores detectables en orina: uromodulina, MAC2BP, AGP 1 y catepsina A.WO2007003670 discloses methods for the diagnosis of fibrotic alterations by using any of four detectable markers in urine: uromodulin, MAC2BP, AGP 1 and cathepsin A.
WO2008031051 divulga un método para la detección y diagnóstico de fibrosis hepática usando un panel de proteínas biomarcadoras de suero humano. Estos métodos también se han divulgado en Gangadharan et al. [Clinical Chemistry 2007; 53:1792-1799].WO2008031051 discloses a method for the detection and diagnosis of liver fibrosis using a panel of human serum biomarker proteins. These methods have also been disclosed in Gangadharan et al. [Clinical Chemistry 2007; 53: 1792-1799].
Otra aproximación alternativa no agresiva para la evaluación de la fibrosis hepática es la elastografϊa transitoria con Fibroscan® (Echosens, Paris, Francia) que opera midiendo la rigidez del hígado (Sandrin L et al. Ultrasound Med. Biol. 2003; 29:1705-1711). Se genera un pulso mecánico en la superficie de la piel, que se propaga a través del hígado. Se mide la velocidad de la onda por ultrasonido. La velocidad se correlaciona directamente con la rigidez del hígado, que a su vez refleja el grado de fibrosis - cuanto más rígido está el hígado mayor es el grado de fibrosis.Another non-aggressive alternative approach for the evaluation of liver fibrosis is transient elastography with Fibroscan® (Echosens, Paris, France) that operates by measuring liver stiffness (Sandrin L et al. Ultrasound Med. Biol. 2003; 29: 1705- 1711). A mechanical pulse is generated on the surface of the skin, which spreads through the liver. The speed of the wave is measured by ultrasound. Speed correlates directly with the stiffness of the liver, which in turn reflects the degree of fibrosis - the stiffer the liver is, the greater the degree of fibrosis.
Aunque hay una necesidad importante de biomarcadores sistémicos no agresivos, específicos de órgano y enfermedad para la fibrosis hepática (y fibrosis de otros órganos también), actualmente no hay un parámetro individual o combinación de ellos disponible, que satisfaga todos los criterios de diagnóstico requeridos para un uso extendido, rentable, y fiable. Las medidas individuales de marcadores bioquímicos en suero, plasma o incluso orina actualmente no son lo suficientemente válidas para remplazar a la biopsia hepática.Although there is a significant need for non-aggressive systemic biomarkers, organ-specific and disease for liver fibrosis (and fibrosis of other organs as well), there is currently no individual parameter or combination of them available, which satisfies all the diagnostic criteria required for extended, cost effective, and reliable use. Individual measurements of biochemical markers in serum, plasma or even urine are currently not valid enough to replace liver biopsy.
COMPENDIO DE LA INVENCIÓN En un primer aspecto, la invención se refiere al uso de la menos un biomarcador para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis o para seguir la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis en un sujeto en donde dicho biomarcador se selecciona del grupo que consiste en SLURPl, HAMP, GSN, APOD, SPPl, DEFBl y MASP2.SUMMARY OF THE INVENTION In a first aspect, the invention relates to the use of at least one biomarker for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis in a subject where said biomarker is selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
En un segundo aspecto, la invención se refiere a un método para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis o para seguir la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis que comprende comparar la expresión de uno o más biomarcadores en una muestra de un sujeto con un estándar predeterminado para cada uno de dichos uno o más biomarcadores; en donde dicho uno o más biomarcadores se seleccionan del grupo que consiste en SLURPl, HAMP, GSN, APOD, SPPl, DEFBl y MASP2; y en donde una diferencia significativa en la expresión de dicho uno o más biomarcadores en dicha muestra comparado con el estándar predeterminado de cada uno de dicho uno o más biomarcadores es indicativo de inicio, fase, evolución de fibrosis y/o enfermedad asociada con fibrosis o de la eficacia de un tratamiento para la fibrosis y/o enfermedad asociada con fibrosis.In a second aspect, the invention relates to a method for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis that comprises comparing the expression of one or more biomarkers in a sample of a subject with a predetermined standard for each of said one or more biomarkers; wherein said one or more biomarkers are selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2; and wherein a significant difference in the expression of said one or more biomarkers in said sample compared to the predetermined standard of each of said one or more biomarkers is indicative of onset, phase, evolution of fibrosis and / or disease associated with fibrosis or of the efficacy of a treatment for fibrosis and / or disease associated with fibrosis.
En aún otro aspecto, la invención se refiere a un kit que comprende reactivos para detectar al menos dos o más biomarcadores seleccionados del grupo de SLURPl, HAMP, GSN , APOD, SPPl, DEFBl y MASP2.In yet another aspect, the invention relates to a kit comprising reagents for detecting at least two or more biomarkers selected from the group of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
BREVE DESCRIPCIÓN DE LAS FIGURASBRIEF DESCRIPTION OF THE FIGURES
Figura 1. GeI de muestra control. Se recogieron las manchas marcadas con círculos y se identificaron mediante LC-MS/MS. La mancha en el círculo blanco se identificó como gelsolina, y la mancha en el círculo negro se identificó como apolipoproteína D.Figure 1. Control sample GeI. Stains marked with circles were collected and identified by LC-MS / MS. The spot in the white circle was identified as gelsolin, and the spot in the black circle was identified as apolipoprotein D.
Figura 2. Los niveles disminuidos de las proteínas identificadas ApoD y gelsolina en muestras de orina cirrótica se confirmaron mediante inmunotransferencia. (A) Las proteínas de la orina se separaron en un gel de SDS-acrilamida y las proteínas se transfirieron en una membrana de nitrocelulosa. Los niveles de ApoD y gelsolina se detectaron usando anticuerpos específicos para estas proteínas. (B) Se cuantificó y representó la intensidad de las bandas. El valor medio de los niveles de proteína mostró un descenso de dos veces en las muestras cirróticas para ambas proteínas.Figure 2. Decreased levels of ApoD and gelsolin proteins identified in cirrhotic urine samples were confirmed by immunoblot. (A) The Urine proteins were separated on an SDS-acrylamide gel and the proteins were transferred on a nitrocellulose membrane. ApoD and gelsolin levels were detected using antibodies specific for these proteins. (B) The intensity of the bands was quantified and represented. The average value of protein levels showed a two-fold decrease in cirrhotic samples for both proteins.
Figura 3. Se representan las intensidad de hepcidina-25 (pico de 2793 Da), hepcidina- 20 (pico de 2193) y SLURP-I (pico de 8855 Da) para las muestras cirróticas, control y de baja fibrosis. Los niveles de esta proteína están significativamente disminuidos en los pacientes cirróticos, por otra parte el grupo de baja fibrosis muestra un nivel de expresión intermedio.Figure 3. The intensity of hepcidin-25 (peak of 2793 Da), hepcidin-20 (peak of 2193) and SLURP-I (peak of 8855 Da) for cirrhotic, control and low fibrosis samples are represented. The levels of this protein are significantly decreased in cirrhotic patients, on the other hand the low fibrosis group shows an intermediate expression level.
Figura 4. Disminución de los niveles de la proteína SLURPl en muestras de orina de pacientes cirróticos con respecto a los controles detectada mediante Western Blot. (A) Las orinas controles (C1-C8) y ocho orinas de pacientes cirróticos (P1-P8) se sometieron a electroforesis en gel de SDS-acrilamida y las proteínas se transfirieron en una membrana de nitrocelulosa. Los niveles de SLURPl se detectaron usando anticuerpos específicos para estas proteínas. (B) Gráfico de la densidad óptica correspondiente a las bandas de SLURP-I obtenidas en las orinas controles y de pacientes cirróticos.Figure 4. Decreased levels of the SLURPl protein in urine samples of cirrhotic patients with respect to the controls detected by Western Blot. (A) The control urine (C1-C8) and eight urine from cirrhotic patients (P1-P8) were subjected to SDS-acrylamide gel electrophoresis and the proteins were transferred on a nitrocellulose membrane. SLURPl levels were detected using antibodies specific for these proteins. (B) Graph of the optical density corresponding to the SLURP-I bands obtained in the control urine and in cirrhotic patients.
Figura 5. Se representan las intensidades de Osteopontina (pico de 7655 Da) y su forma fosforilada (pico de 7735 Da) para las muestras cirróticas, control y de baja fibrosis. Los niveles de esta proteína están significativamente disminuidos en los pacientes cirróticos, por otra parte el grupo de baja fibrosis muestra un nivel de expresión intermedio.Figure 5. The intensities of Osteopontin (7655 Da peak) and its phosphorylated form (7735 Da peak) for cirrhotic, control and low fibrosis samples are depicted. The levels of this protein are significantly decreased in cirrhotic patients, on the other hand the low fibrosis group shows an intermediate expression level.
Figura 6. Se representan las intensidades de Defensina Beta 1 (pico de 4623 Da) y MASP2 (pico de 5804 Da) para las muestras cirróticas, control y de baja fibrosis. Los niveles de esta proteína están significativamente disminuidos en los pacientes cirróticos, por otra parte el grupo de baja fibrosis muestra un nivel de expresión intermedio. DESCRIPCIÓN DETALLADA DE LA INVENCIÓNFigure 6. The intensities of Defensin Beta 1 (peak of 4623 Da) and MASP2 (peak of 5804 Da) for cirrhotic, control and low fibrosis samples are depicted. The levels of this protein are significantly decreased in cirrhotic patients, on the other hand the low fibrosis group shows an intermediate expression level. DETAILED DESCRIPTION OF THE INVENTION
Los autores de la presente invención han identificado biomarcadores proteicos en muestras de orina humana cuya expresión se correlaciona con la presencia de cirrosis y fϊbrosis hepática. En particular, los autores han identificado gelsolina, apoliproteína D, hepcidina (25 y 20), SLURP-I, Osteopontina (SPPl, OPN ó OSTP), Beta 1 DefensinaThe authors of the present invention have identified protein biomarkers in human urine samples whose expression correlates with the presence of cirrhosis and liver fibrosis. In particular, the authors have identified gelsolin, apoliprotein D, hepcidin (25 and 20), SLURP-I, Osteopontin (SPPl, OPN or OSTP), Beta 1 Defensin
(DEFBl) y Serina proteasa asociada a lectina unida a mañosa (MASP2) como proteínas cuyos niveles disminuyen significativamente en las muestras de orina de pacientes cirróticos cuando se comparan con muestras control. Estas proteínas son marcadores potenciales de estados avanzados de fibrosis hepática.(DEFBl) and Serine-linked lectin-linked serine protease (MASP2) as proteins whose levels decrease significantly in the urine samples of cirrhotic patients when compared to control samples. These proteins are potential markers of advanced states of liver fibrosis.
De esta manera, en un primer aspecto, la invención se refiere al uso de un biomarcador para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad que implica fibrosis o para seguir la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis en un sujeto en donde dicho biomarcador se selecciona del grupo que consiste en SLURPl, HAMP, GSN, APOD, SPPl, DEFBl y MASP2.Thus, in a first aspect, the invention relates to the use of a biomarker for the detection, diagnosis and evaluation of fibrosis and / or of a disease involving fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or of a disease associated with fibrosis in a subject where said biomarker is selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
El término "biomarcador" o, de forma alternativa, "marcador molecular", como se usa aquí, se usa para referirse a una molécula o al producto de expresión de un gen o fragmentos y variantes del mismo que muestran cambios sustanciales en una enfermedad determinada y que se pueden usar tanto para la detección como para el diagnóstico de una enfermedad detectando la aparición de dichos cambios en el biomarcador y/o para seguir la eficacia de un tratamiento para esa enfermedad detectando cambios en el biomarcador en oposición a aquellos que se dan en la enfermedad o situación clínica.The term "biomarker" or, alternatively, "molecular marker", as used herein, is used to refer to a molecule or the expression product of a gene or fragments and variants thereof that show substantial changes in a given disease. and that can be used both for the detection and for the diagnosis of a disease by detecting the appearance of said changes in the biomarker and / or to follow the efficacy of a treatment for that disease by detecting changes in the biomarker as opposed to those that occur in the disease or clinical situation.
En una forma de realización preferida, los cambios en el biomarcador son cambios en los niveles de expresión. El término "expresión", como se usa aquí, se refiere a un proceso mediante el cual se produce un polipéptido a partir del ADN. Este proceso implica la transcripción del gen a un ARN mensajero (ARNm), y la traducción de este ARNm al polipéptido. En el contexto de la invención "cambios en los niveles de expresión" se refiere a cualquier cambio en la producción del ARNm, del polipéptido o de ambos que produce niveles relativos alterados del ARNm, proteína o ambos en una muestra con respecto a otras moléculas en la misma muestra. Se apreciará que los niveles de expresión de un biomarcador se pueden determinar mediante la determinación de los niveles de ARNm en una muestra o mediante la determinación de los niveles del polipéptido correspondiente. De forma alternativa, los biomarcadores polipeptídicos pueden ser variantes resultantes de modificaciones postraduccionales, incluyendo fragmentos de los mismos.In a preferred embodiment, changes in the biomarker are changes in expression levels. The term "expression", as used herein, refers to a process by which a polypeptide is produced from DNA. This process involves the transcription of the gene to a messenger RNA (mRNA), and the translation of this mRNA into the polypeptide. In the context of the invention "changes in the levels of expression "refers to any change in the production of mRNA, polypeptide or both that produces altered relative levels of mRNA, protein or both in a sample with respect to other molecules in the same sample. It will be appreciated that the expression levels of a biomarker can be determined by determining the levels of mRNA in a sample or by determining the levels of the corresponding polypeptide Alternatively, the polypeptide biomarkers can be variants resulting from post-translational modifications, including fragments thereof.
Los términos "diagnóstico", "detección" y "evaluación", se usan aquí de forma intercambiable, y se refiere a la valoración de la probabilidad según la cual un sujeto padece una enfermedad así como a la valoración de su inicio, estado de desarrollo, evolución, o de su regresión, y/o el pronóstico del curso de la enfermedad en el futuro. Como entenderán los expertos en la materia, tal valoración, aunque se prefiere que sea, normalmente puede no ser correcta para el 100% de los sujetos que se va a diagnosticar. El término, sin embargo, requiere que una parte estadísticamente significativa de los sujetos se pueda identificar como que padecen la enfermedad o que tienen predisposición a la misma. Si una parte es estadísticamente significativa se puede determinar sin más por el experto en la materia usando varias herramientas de evaluación estadística bien conocidas, por ejemplo, determinación de intervalos de confianza, determinación de valores p, prueba t de Student, prueba de Mann-Whitney, etc. Los detalles se encuentran en Dowdy y Wearden, Statistics for Research, John Wiley & Sons, Nueva York 1983. Los intervalos de confianza preferidos son al menos el 50%, al menos el 60%, al menos el 70%, al menos el 80%, al menos el 90%, al menos el 95%. Los valores de p son, preferiblemente, 0,2, 0,1, 0,05.The terms "diagnosis", "detection" and "evaluation" are used interchangeably here, and refers to the assessment of the probability according to which a subject suffers from a disease as well as the assessment of its onset, developmental state , evolution, or its regression, and / or the prognosis of the course of the disease in the future. As those skilled in the art will understand, such assessment, although preferred, may not be correct for 100% of the subjects to be diagnosed. The term, however, requires that a statistically significant part of the subjects can be identified as having the disease or having a predisposition to it. If a part is statistically significant, it can be determined by the person skilled in the art using several well-known statistical evaluation tools, for example, determination of confidence intervals, determination of p-values, 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%, at least 95%. P values are preferably 0.2, 0.1, 0.05.
La expresión "seguir la eficacia de un tratamiento" se refiere a la valoración de los resultados de una terapia en un paciente que padece fibrosis o una enfermedad asociada con fibrosis. Las terapias adecuadas para el tratamiento de fibrosis incluyen el tratamiento con inhibidores de PCP, IFN-γ o mesilato de imatinib (Gleevec). La utilidad de los biomarcadores de la invención para seguir la eficacia de un tratamiento también se puede aplicar a métodos para seleccionar y cribar fármacos con potencial actividad antifibrosante. Este proceso comprende a) administrar al sujeto (preferiblemente un animal) el fármaco a estudiar; b) a diferentes puntos del estudio (antes, durante y/o después de la administración) coger muestras biológicas del animal y determinar los niveles de marcador según la presente invención; y c) comparar las determinaciones realizadas en las muestras obtenidas en las diferentes fases del tratamiento y compararlas a animales control, por ejemplo sin tratar.The expression "follow the effectiveness of a treatment" refers to the assessment of the results of a therapy in a patient suffering from fibrosis or a disease associated with fibrosis. Suitable therapies for the treatment of fibrosis include treatment with PCP inhibitors, IFN-γ or imatinib mesylate (Gleevec). The usefulness of the biomarkers of the invention to track the efficacy of a treatment can also be applied to methods for selecting and screening drugs with potential activity. antifibrosant This process comprises a) administering to the subject (preferably an animal) the drug to be studied; b) at different points of the study (before, during and / or after administration) take biological samples of the animal and determine the marker levels according to the present invention; and c) compare the determinations made in the samples obtained in the different phases of the treatment and compare them to control animals, for example untreated.
El término "SLURP-I", como se usa aquí, se refiere al gen conocido como "que contiene el dominio LY6/PLAUR secretado 1", también identificado mediante el número de acceso HGNC ID. 18746 (Comité de Nomenclatura de Genes de HUGO, Organización del Genoma Humano), que está localizado en la región cromosómica 8q24.3. La proteína codificada por SLURP-I se identifica en la base de datos Entrez mediante el número de acceso GeneID:57152 (actualizado el 27 de julio de 2008) y también se conoce como "componente B de ARS" (ARS, ArsB), "proteína urinaria antineoplásica" (ANUP), MDM, "similar al antígeno 6 de linfocitos secretado" (LY6LS). En una forma de realización particular el producto de expresión del gen SLURPl comprende SEQ ID NO: 1, que es un polipéptido de 103 aminoácidos identificado con el número de acceso UniProt ID P55000 (última modificación 22 de julio, 2008. Versión 75) (SEQ ID NO: 1) así como ortólogos, isoformas y fragmentos de dicho polipéptido. En otra forma de realización preferida, el biomarcador es un fragmento de dicho polipéptido que comprende los aminoácidos 22-103 (SEQ ID NO:2), correspondiente al polipéptido maduro secretado.The term "SLURP-I", as used herein, refers to the gene known as "containing the secreted domain LY6 / PLAUR 1", also identified by the accession number HGNC ID. 18746 (HUGO Gene Nomenclature Committee, Human Genome Organization), which is located in chromosomal region 8q24.3. The protein encoded by SLURP-I is identified in the Entrez database by GeneID access number: 57152 (updated July 27, 2008) and is also known as "ARS component B" (ARS, ArsB), " urinary antineoplastic protein "(ANUP), MDM," similar to secreted lymphocyte antigen 6 (LY6LS). In a particular embodiment, the expression product of the SLURP1 gene comprises SEQ ID NO: 1, which is a 103 amino acid polypeptide identified with the UniProt ID accession number P55000 (last modified July 22, 2008. Version 75) (SEQ ID NO: 1) as well as orthologs, isoforms and fragments of said polypeptide. In another preferred embodiment, the biomarker is a fragment of said polypeptide comprising amino acids 22-103 (SEQ ID NO: 2), corresponding to the secreted mature polypeptide.
El término "HAMP", como se usa aquí, se refiere al gen "péptido antimicrobiano hepcidina", identificado mediante el número de acceso HGNC ID. 15598, y que está localizado en la región cromosómica 19ql3.1. El "péptido antimicrobiano hepcidina"The term "HAMP", as used herein, refers to the "hepcidin antimicrobial peptide" gene, identified by the HGNC ID accession number. 15598, and which is located in chromosomal region 19ql3.1. The "hepcidin antimicrobial peptide"
(HAMP) se identifica en la base de datos Entrez con GeneID:57817 (actualizado el 12 de octubre de 2008) y también se conoce como "péptido antimicrobiano expresado en hígado" (LEAP-I), hepcidina, HEPC, HFE2B, LEAPl, y "regresor tumoral putativo de hígado" (PLTR). En una forma de realización particular, el producto de expresión del gen HAMP comprende cualquiera de las secuencias SEQ ID NO :3, que es la pre-pro- proteína de 84 aminoácidos identificada por el número de acceso de UniProt ID. P81172 (última modificación 22 de julio, 2008. Versión 82); SEQ ID NO:4 (aminoácidos 60-84, conocido como hepcidina 25 o Hepc25); SEQ ID NO:5 (aminoácidos 65-84, conocido como hepcidina 20 o Hepc20); así como ortólogos, isoformas y fragmentos de dicho polipéptido. En una forma de realización preferida el producto de expresión de HAMP es Hepc25, Hepc20 o ambos.(HAMP) is identified in the Entrez database with GeneID: 57817 (updated October 12, 2008) and is also known as "liver-expressed antimicrobial peptide" (LEAP-I), hepcidin, HEPC, HFE2B, LEAPl, and "putative liver tumor regressor" (PLTR). In a particular embodiment, the expression product of the HAMP gene comprises any of the sequences SEQ ID NO: 3, which is the 84-amino acid preproprotein identified by the UniProt ID accession number. P81172 (last modified July 22, 2008. Version 82); SEQ ID NO: 4 (amino acids 60-84, known as hepcidin 25 or Hepc25); SEQ ID NO: 5 (amino acids 65-84, known as hepcidin 20 or Hepc20); as well as orthologs, isoforms and fragments of said polypeptide. In a preferred embodiment the HAMP expression product is Hepc25, Hepc20 or both.
El término "GSN", como se usa aquí, se refiere al gen "gelsolina (amiloidosis, de tipo finlandés), identificado mediante el número de acceso HGNC ID. 4620, y que está localizado en la región cromosómica 9q33. El producto de expresión del gen también se identifica en la base de datos Entrez como GeneID: 2934 (actualizado el 28 de septiembre de 2008) o DKFZp313L0718. La proteína se conoce como gelsolina, y de forma alternativa como "factor despolimerizador de actina" (ADF), brevina o AGEL. Se han descrito diversas variantes transcripcionales para GSN que codifican diferentes isoformas. En una forma de realización particular el producto de expresión de GSN comprende cualquiera de las secuencias SEQ ID NO:6, que corresponde al polipéptido precursor secretado identificado mediante UniProt ID. P06396 (última modificación 14 de octubre, 2008. Versión 114) así como ortólogos, isoformas y fragmentos de dicho polipéptido.The term "GSN", as used herein, refers to the gene "gelsolin (amyloidosis, of the Finnish type), identified by accession number HGNC ID. 4620, and which is located in the chromosomal region 9q33. The expression product of the gene is also identified in the Entrez database as GeneID: 2934 (updated September 28, 2008) or DKFZp313L0718. The protein is known as gelsolin, and alternatively as "actin depolymerizing factor" (ADF), brevina or AGEL Various transcriptional variants for GSN that encode different isoforms have been described In a particular embodiment the GSN expression product comprises any of the sequences SEQ ID NO: 6, which corresponds to the secreted precursor polypeptide identified by UniProt ID. P06396 (last modified October 14, 2008. Version 114) as well as orthologs, isoforms and fragments of said polypeptide.
El término "APOD", como se usa aquí, se refiere al gen "apoliproteína D", también identificado mediante el número de acceso HGNC: 612, que está localizado en la región cromosómica 3q26.2-qter. APOD también se identifica mediante "Entrez GeneID: 347" (actualizado el 12 de octubre de 2008), En una forma de realización particular el producto de expresión de APOD comprende cualquiera de las secuencias SEQ ID NO:7, un precursor del polipéptido identificado como UniProt ID. P05090 (última modificación 22 de julio, 2008. Versión 107) así como ortólogos, isoformas y fragmentos de dicho polipéptido.The term "APOD", as used herein, refers to the "apoliprotein D" gene, also identified by the HGNC accession number: 612, which is located in the chromosomal region 3q26.2-qter. APOD is also identified by "Entrez GeneID: 347" (updated October 12, 2008). In a particular embodiment, the APOD expression product comprises any of the sequences SEQ ID NO: 7, a precursor of the polypeptide identified as UniProt ID. P05090 (last modified July 22, 2008. Version 107) as well as orthologs, isoforms and fragments of said polypeptide.
El término "SPPl", como se usa aquí, se refiere al gen "Osteopontina" ó "fosfoproteína secretada tipo 1"; OPN, y conocido de forma alternativa como BNSP; BSPI; ETA-I;The term "SPPl", as used herein, refers to the gene "Osteopontin" or "phosphoprotein secreted type 1"; OPN, and known alternatively as BNSP; BSPI; ETA-I;
MGCl 10940, correspondiente al polipéptido identificado mediante el número de accesoMGCl 10940, corresponding to the polypeptide identified by the accession number
HGNC: 11255 que está localizado en la región cromosómica 4q21-q25. SPPl también se identifica mediante "Entrez Gene ID: 6696" (actualizado el 4 de diciembre de 2009). En una forma de realización particular el producto de expresión del gen SPPl comprende la SEQ ID NO: 8, que es un polipéptido de 314 aminoácidos identificado con el número de acceso UniProt P 10451 (última modificación 24 de noviembre, 2009. Versión 120); así como ortólogos, iso formas y fragmentos de dicho polipéptido, incluyendo las isoformas de SPPl identificadas como SEQ ID NO:8, 9, 10 y 11.HGNC: 11255 which is located in the 4q21-q25 chromosomal region. SPPl too is identified by "Entrez Gene ID: 6696" (updated on December 4, 2009). In a particular embodiment, the SPP1 gene expression product comprises SEQ ID NO: 8, which is a 314 amino acid polypeptide identified with the UniProt accession number P 10451 (last modified November 24, 2009. Version 120); as well as orthologs, iso forms and fragments of said polypeptide, including the SPP1 isoforms identified as SEQ ID NO: 8, 9, 10 and 11.
El término "DEFBl", como se usa aquí, se refiere al gen "Beta 1 defensina o Defensina beta 1" y conocido de forma alternativa como BDl; HBDl; DEFB-I; DEFB101; MGC51822, correspondiente al polipéptido identificado mediante el número de acceso HGNC:2766, que está localizado en la región cromosómica 8p23.2-p23.1. DEFBl también se identifica mediante "Entrez GeneID: 1672" (actualizado el 4 de diciembre de 2009). En una forma de realización particular, el producto de expresión de DEFBl comprende la secuencias SEQ ID NO:9, identificada como UniProt P60022.1 (última modificación 3 de noviembre, 2009. Versión 72) así como ortólogos, isoformas y fragmentos de dicho polipéptido.The term "DEFBl", as used herein, refers to the gene "Beta 1 defensin or Defensin beta 1" and alternatively known as BDl; HBDl; DEFB-I; DEFB101; MGC51822, corresponding to the polypeptide identified by HGNC accession number: 2766, which is located in chromosomal region 8p23.2-p23.1. DEFBl is also identified by "Entrez GeneID: 1672" (updated on December 4, 2009). In a particular embodiment, the DEFBl expression product comprises the sequences SEQ ID NO: 9, identified as UniProt P60022.1 (last modified November 3, 2009. Version 72) as well as orthologs, isoforms and fragments of said polypeptide .
El término "MASP2", como se usa aquí, se refiere al gen "Serina proteasa asociada a lectina unida a mañosa" del inglés "Mannan-binding lectin serine peptidase 2" y de forma alternativa, sMAP; MAP 19; MASP-2, también identificado mediante el número de acceso HGNC:6902, que está localizado en la región cromosómica Ip36.3-p36.2. MASP2 también se identifica mediante "Entrez GeneID: 10747" (actualizado el 4 de diciembre de 2009). En una forma de realización particular el producto de expresión de SPPl comprende la secuencia SEQ ID NO:10, identificada como UniProt O00187.3 (última modificación 24 de noviembre, 2009. Versión 118); así como ortólogos, isoformas y fragmentos de dicho polipéptido, incluyendo las isoformas identificadas en SEQ ID NO: 13 y 14 de la presente invención.The term "MASP2", as used herein, refers to the "Manina-binding lectin serine peptidase 2" and "alternatively, sMAP; MAP 19; MASP-2, also identified by HGNC accession number: 6902, which is located in the chromosomal region Ip36.3-p36.2. MASP2 is also identified by "Entrez GeneID: 10747" (updated on December 4, 2009). In a particular embodiment, the SPP1 expression product comprises the sequence SEQ ID NO: 10, identified as UniProt O00187.3 (last modified November 24, 2009. Version 118); as well as orthologs, isoforms and fragments of said polypeptide, including the isoforms identified in SEQ ID NO: 13 and 14 of the present invention.
El término "fibrosis", como se usa aquí, se refiere un exceso de deposición de matriz extracelular (MEC) que implica reorganización molecular e histológica de varios tipos de colágenos, proteoglicanos, glicoproteínas estructurales y ácido hialurónicoThe term "fibrosis," as used herein, refers to an excess of extracellular matrix deposition (ECM) that involves molecular and histological reorganization of various types of collagen, proteoglycans, structural glycoproteins and hyaluronic acid.
(hialuronano). Aunque la fibrosis es una marca de muchas enfermedades del hígado, otros órganos también pueden sufrir un exceso de deposición de tejido conjuntivo. En una forma de realización preferida, la fϊbrosis es fϊbrosis hepática.(hyaluronan). Although fibrosis is a mark of many liver diseases, Other organs may also suffer from excess deposition of connective tissue. In a preferred embodiment, the fibrosis is hepatic fibrosis.
El término "enfermedad asociada con fϊbrosis", se refiere a cualquier enfermedad en donde uno o más órganos padecen una deposición excesiva de tejido conjuntivo fibroso. Estas enfermedades incluyen fibrosis quística del páncreas y pulmones, fibrosis por inyección, que puede ocurrir como complicación de inyecciones intramusculares, especialmente en niños, fibrosis endomiocardiaca, fibrosis pulmonar idiopática, fibrosis mediastinal, mielofibrosis, fibrosis retroperitoneal, fibrosis masiva progresiva, una complicación de la pneumoconiosis de los mineros, fibrosis nefrogénica sistémica, enfermedad pulmonar parenquimatosa difusa, síndrome de dolor post-vasectomía, la tuberculosis (TB) puede producir fibrosis de los pulmones, la anemia falciforme puede producir aumento y por último fibrosis del bazo y artritis reumatoide y cirrosis, que pueden causar fibrosis hepática. En una forma de realización preferida, la enfermedad asociada con la fibrosis es cirrosis.The term "disease associated with fibrosis" refers to any disease where one or more organs suffer from excessive deposition of fibrous connective tissue. These diseases include cystic fibrosis of the pancreas and lungs, injection fibrosis, which can occur as a complication of intramuscular injections, especially in children, endomyocardiac fibrosis, idiopathic pulmonary fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, progressive massive fibrosis, a complication of miner's pneumoconiosis, systemic nephrogenic fibrosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, tuberculosis (TB) can cause fibrosis of the lungs, sickle cell anemia can lead to increase and finally spleen fibrosis and rheumatoid arthritis and cirrhosis , which can cause liver fibrosis. In a preferred embodiment, the disease associated with fibrosis is cirrhosis.
La invención también se refiere a un método para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis o para seguir la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis que comprende comparar la expresión de uno o más biomarcadores en una muestra de un sujeto con un estándar predeterminado para cada uno de dichos uno o más biomarcadores; en donde dicho uno o más biomarcadores se seleccionan del grupo que consiste en SLURPl, HAMP, GSN, APOD, SPPl, DEFBl y MASP2; y en donde una diferencia significativa en la expresión de dicho uno o más biomarcadores en dicha muestra comparado con el estándar predeterminado de cada uno de dicho uno o más biomarcadores es indicativo de inicio, fase, evolución de fibrosis y/o enfermedad asociada con fibrosis o de la eficacia de un tratamiento para la fibrosis y/o enfermedad asociada con fibrosis.The invention also relates to a method for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis comprising comparing the expression of one or more biomarkers in a sample of a subject with a predetermined standard for each of said one or more biomarkers; wherein said one or more biomarkers are selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2; and wherein a significant difference in the expression of said one or more biomarkers in said sample compared to the predetermined standard of each of said one or more biomarkers is indicative of onset, phase, evolution of fibrosis and / or disease associated with fibrosis or of the efficacy of a treatment for fibrosis and / or disease associated with fibrosis.
Los términos y expresiones "diagnóstico", "seguir la eficacia de un tratamiento", "fibrosis", "enfermedad asociada con fibrosis", "biomarcador", "SLURPl", "HAMP", "GSN", "APOD", "SPPl", "DEFBl" y "MASP2"se han descrito previamente en relación con el uso de la invención. El método de la invención permite la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis así como el seguimiento de la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis. El método de la invención comprende la determinación de la expresión de uno o más biomarcadores seleccionados del grupo de SLURPl, HAMP, GSN, APOD, SPPl, DEFBl, MASP2 en una muestra biológica de un individuo.The terms and expressions "diagnosis", "follow the efficacy of a treatment", "fibrosis", "disease associated with fibrosis", "biomarker", "SLURPl", "HAMP", "GSN", "APOD", "SPPl "," DEFBl "and" MASP2 "have been previously described in relation to the use of the invention. The method of the invention allows the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis as well as monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis. The method of the invention comprises determining the expression of one or more biomarkers selected from the group of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl, MASP2 in a biological sample of an individual.
El término "muestra", como se usa aquí, significa cualquier tejido o líquido biológico tomado de un sujeto. En una forma de realización preferida es líquido biológico, por ejemplo, sangre, suero o, más preferiblemente orina.The term "sample", as used herein, means any biological tissue or fluid taken from a subject. In a preferred embodiment it is biological liquid, for example, blood, serum or, more preferably urine.
La expresión de un biomarcador de la invención se puede evaluar mediante cualquiera de una amplia variedad de métodos bien conocidos para detectar la expresión de una molécula transcrita o su proteína correspondiente. Si los biomarcadores son moléculas de ácido nucleico, la expresión del polinucleótido marcador se puede detectar usando métodos de hibridación de ácidos nucleicos, métodos de transcripción inversa de ácidos nucleicos, métodos de amplificación de ácidos nucleicos y similares. En una forma de realización preferida, la expresión de un gen marcador se evalúa preparando ARNm/ ADNc (es decir, un polinucleótido transcrito) de células en una muestra de un paciente, y mediante la hibridación del ARNm/ADNc con un polinucleótido de referencia que es complementario a un polinucleótido que comprende el gen marcador, y fragmentos del mismo. El ADNc se puede, opcionalmente, amplificar usando cualquiera de varios métodos de la reacción en cadena de la polimerasa antes de la hibridación con el polinucleótido de referencia aunque se prefiere que no se amplifique.The expression of a biomarker of the invention can be evaluated by any of a wide variety of well known methods to detect the expression of a transcribed molecule or its corresponding protein. If the biomarkers are nucleic acid molecules, expression of the marker polynucleotide can be detected using nucleic acid hybridization methods, nucleic acid reverse transcription methods, nucleic acid amplification methods and the like. In a preferred embodiment, expression of a marker gene is evaluated by preparing mRNA / cDNA (i.e., a transcribed polynucleotide) of cells in a sample of a patient, and by hybridizing the mRNA / cDNA with a reference polynucleotide that It is complementary to a polynucleotide comprising the marker gene, and fragments thereof. The cDNA can, optionally, be amplified using any of several polymerase chain reaction methods before hybridization with the reference polynucleotide although it is preferred that it is not amplified.
En una forma de realización preferida, los biomarcadores son polipéptidos, en cuyo caso la detección se puede llevar a cabo usando métodos inmunológicos para la detección de proteínas secretadas, métodos de purificación de proteínas, ensayos de función o actividad de proteína. En una forma de realización preferida, la expresión de una proteína marcadora se evalúa usando un anticuerpo (por ejemplo, un anticuerpo radio- marcado, marcado con cromóforo, marcado con fluoróforo, o marcado con enzima), un derivado de anticuerpo (por ejemplo, un anticuerpo conjugado con un sustrato o con la proteína o ligando de un par proteína-ligando (por ejemplo, biotina-estreptavidina), o un fragmento de anticuerpo (por ejemplo, un anticuerpo de cadena sencilla, el dominio hipervariable de un anticuerpo aislado, etc.) que se une específicamente con una proteína correspondiente al gen marcador, tal como la proteína codificada por el marco abierto de lectura correspondiente al gen marcador o tal proteína que ha sufrido toda o una parte de su modificación postraduccional normal.In a preferred embodiment, the biomarkers are polypeptides, in which case detection can be carried out using immunological methods for the detection of secreted proteins, protein purification methods, protein function or activity assays. In a preferred embodiment, the expression of a marker protein is evaluated using an antibody (eg, a radiolabeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), a antibody derivative (for example, an antibody conjugated to a substrate or to the protein or ligand of a protein-ligand pair (e.g., biotin-streptavidin), or an antibody fragment (e.g., a single chain antibody, the hypervariable domain of an isolated antibody, etc.) that specifically binds with a protein corresponding to the marker gene, such as the protein encoded by the open reading frame corresponding to the marker gene or such a protein that has undergone all or part of its modification normal post-translational
El inmunoensayo más normal es el "enzimoinmunoanálisis de adsorción (ELISA)". El ELISA es una técnica para detectar y medir la concentración de un antígeno usando una forma marcada (por ejemplo unida a enzima) del anticuerpo. Hay diferentes formas deThe most normal immunoassay is the "adsorption enzyme immunoassay (ELISA)". ELISA is a technique to detect and measure the concentration of an antigen using a labeled (for example enzyme-bound) form of the antibody. There are different ways of
ELISA, que son bien conocidas para los expertos en la materia. Las técnicas estándar conocidas en la técnica para ELISA se describen en "Methods in Immunodiagnosis", 2a ELISA, which are well known to those skilled in the art. Standard techniques known in the art for ELISA are described in "Methods in Immunodiagnosis", 2 a
Edición, Rose y Bigazzi, eds. John Wiley AND Sons, 1980; Campbell et al, "Methods and Immunology", W. A. Benjamín, Inc., 1964; y Oellerich, M. 1984, J. Clin. Chem.Edition, Rose and Bigazzi, eds. John Wiley AND Sons, 1980; Campbell et al, "Methods and Immunology", W. A. Benjamín, Inc., 1964; and Oellerich, M. 1984, J. Clin. Chem
Clin. Biochem., 22:895-904.Clin. Biochem., 22: 895-904.
En un "ELISA sandwich", un anticuerpo (por ejemplo anti-enzima) está unido a una fase sólida (es decir, a una placa de microtitulación) y se expone a una muestra biológica que contiene antígeno (por ejemplo, enzima). La fase sólida se lava después para eliminar el antígeno no unido. Después se une un anticuerpo marcado (por ejemplo, unido a enzima) al antígeno unido (si está presente) formando un sandwich anticuerpo-antígeno-anticuerpo. Los ejemplos de enzimas que se pueden unir al anticuerpo son fosfatasa alcalina, peroxidasa de rábano, luciferasa, ureasa, y B- galactosidasa. El anticuerpo unido a enzima reacciona con un sustrato para generar un producto de reacción coloreado que se puede medir.In an "ELISA sandwich", an antibody (for example anti-enzyme) is bound to a solid phase (ie, to a microtiter plate) and is exposed to a biological sample containing antigen (for example, enzyme). The solid phase is then washed to remove unbound antigen. A labeled antibody (for example, enzyme bound) is then attached to the bound antigen (if present) forming an antibody-antigen-antibody sandwich. Examples of enzymes that can bind to the antibody are alkaline phosphatase, horseradish peroxidase, luciferase, urease, and B-galactosidase. The enzyme-bound antibody reacts with a substrate to generate a colored reaction product that can be measured.
En un "ELISA competitivo", el anticuerpo se incuba con una muestra que contiene antígeno (es decir, enzima). La mezcla antígeno-anticuerpo se pone en contacto después con una fase sólida (por ejemplo, una placa de microtitulación) que está recubierta con antígeno (es decir, enzima). Cuanto más antígeno haya presente en la muestra, menos anticuerpo libre estará disponible para unirse a la fase sólida. Se añade después un anticuerpo secundario marcado (por ejemplo, unido a enzima) a la fase sólida para determinar la cantidad de anticuerpo primario unido a la fase sólida.In a "competitive ELISA", the antibody is incubated with a sample containing antigen (ie, enzyme). The antigen-antibody mixture is then contacted with a solid phase (for example, a microtiter plate) that is coated with antigen (i.e., enzyme). The more antigen present in the sample, the less free antibody will be available to bind to the solid phase. It is added after a labeled secondary antibody (eg, enzyme bound) to the solid phase to determine the amount of primary antibody bound to the solid phase.
En un "ensayo de inmunohistoquímica" se prueba una sección de tejido para proteínas específicas exponiendo el tejido a anticuerpos que son específicos para la proteína que se está ensayando. Los anticuerpos se visualizan después mediante cualquiera de un número de métodos para determinar la presencia y cantidad de la proteína presente. Los ejemplos de los métodos usados para visualizar anticuerpos son, por ejemplo, por medio de enzimas unidas a los anticuerpos (por ejemplo, luciferasa, fosfatasa alcalina, peroxidasa de rábano, o beta-galactosidasa), o métodos químicos (por ejemplo, cromógeno DAB/sustrato). La muestra se analiza después de forma microscópica, lo más preferiblemente mediante microscopía óptica de una muestra teñida con un colorante que se detecta en el espectro visible, usando cualquiera de una variedad de tales métodos y reactivos de tinción conocidos para el experto en la materia.In a "immunohistochemical assay" a section of tissue is tested for specific proteins by exposing the tissue to antibodies that are specific to the protein being tested. The antibodies are then visualized by any of a number of methods to determine the presence and amount of the protein present. Examples of the methods used to visualize antibodies are, for example, by means of enzymes bound to the antibodies (for example, luciferase, alkaline phosphatase, horseradish peroxidase, or beta-galactosidase), or chemical methods (for example, DAB chromogen /substratum). The sample is then analyzed microscopically, most preferably by optical microscopy of a sample stained with a dye that is detected in the visible spectrum, using any of a variety of such methods and staining reagents known to the person skilled in the art.
De forma alternativa, se pueden emplear "radioinmunoensayos". Un radioinmunoensayo es una técnica para detectar y medir la concentración de un antígeno usando una forma marcada (por ejemplo, marcada de forma radioactiva o fluorescente) del antígeno. Los ejemplos de marcas radioactivas para antígenos incluyen 3H, 14C, y 125I. Se mide la concentración de enzima antígeno en una muestra biológica haciendo que el antígeno en la muestra biológica compita con el antígeno marcado (por ejemplo radioactivamente) para la unión a un anticuerpo para el antígeno. Para asegurar la unión competitiva entre el antígeno marcado y el antígeno no marcado, el antígeno marcado está presente en una concentración suficiente para saturar los sitios de unión del anticuerpo. Cuanto mayor sea la concentración del antígeno en la muestra, menor será la concentración del antígeno marcado que se unirá al anticuerpo. En un radioinmunoensayo, para determinar la concentración del antígeno marcado unido al anticuerpo, el complejo antígeno-anticuerpo se debe separar del antígeno libre. Un método para separar el complejo antígeno-anticuerpo del antígeno libre es precipitar el complejo antígeno-anticuerpo con antisuero anti-isotipo. Otro método de separar el complejo antígeno-anticuerpo del antígeno libre es precipitar el complejo antígeno- anticuerpo con S. aureus aniquilada con formalina. Aún otro método para separar el complejo antígeno-anticuerpo del antígeno libre es realizar un "radioinmunoensayo de fase sólida" donde el anticuerpo está unido (por ejemplo, de forma covalente) a bolas de Sepharosa, pocilios de poliestireno, pocilios de policloruro de vinilo, o pocilios de microtitulación. Comparando la concentración de antígeno marcado unido al anticuerpo con una curva patrón basada en muestras que tienen una concentración de antígeno conocida, se puede determinar la concentración de antígeno en la muestra biológica.Alternatively, "radioimmunoassays" can be used. A radioimmunoassay is a technique to detect and measure the concentration of an antigen using a labeled form (for example, radioactively or fluorescently labeled) of the antigen. Examples of radioactive labels for antigens include 3 H, 14 C, and 125 I. The concentration of antigen enzyme in a biological sample is measured by causing the antigen in the biological sample to compete with the labeled antigen (eg radioactively) for binding. to an antibody to the antigen. To ensure competitive binding between the labeled antigen and the unlabeled antigen, the labeled antigen is present in a concentration sufficient to saturate the antibody binding sites. The higher the concentration of the antigen in the sample, the lower the concentration of the labeled antigen that will bind to the antibody. In a radioimmunoassay, to determine the concentration of the labeled antigen bound to the antibody, the antigen-antibody complex must be separated from the free antigen. One method of separating the antigen-antibody complex from the free antigen is to precipitate the antigen-antibody complex with anti-isotype antiserum. Another method of separating the antigen-antibody complex from the free antigen is to precipitate the antigen-antibody complex with S. aureus annihilated with formalin. Yet another method to separate the Antigen-antibody complex of the free antigen is to perform a "solid phase radioimmunoassay" where the antibody is bound (for example, covalently) to Sepharose beads, polystyrene wells, polyvinylchloride wells, or microtiter wells. By comparing the concentration of labeled antigen bound to the antibody with a standard curve based on samples having a known antigen concentration, the concentration of antigen in the biological sample can be determined.
Un "ensayo inmunorradiométrico" (IRMA) es un inmunoensayo en el que el reactivo anticuerpo está marcado radioactivamente. Un IRMA requiere la producción de un conjugado antigénico multivalente, mediante técnicas tales como conjugación a una proteína por ejemplo, seroalbúmina de conejo (RSA). El conjugado antigénico multivalente debe tener al menos 2 residuos de antígenos por molécula y los residuos de antígeno deben estar separados a una distancia suficiente para permitir la unión de al menos dos anticuerpos al antígeno. Por ejemplo, en un IRMA el conjugado antigénico multivalente puede estar unido a una superficie sólida tal como una esfera de plástico. Se añaden antígeno "muestra" no marcado y anticuerpo para el antígeno que está marcado radioactivamente a un tubo de ensayo que contiene la esfera recubierta con el conjugado antigénico multivalente. El antígeno en la muestra compite con el conjugado antigénico multivalente por los sitios de unión del anticuerpo al antígeno. Después de un periodo de incubación apropiado, los reactivos no unidos se eliminan lavando y se determina la cantidad de radioactividad en la fase sólida. La cantidad de anticuerpo radioactivo unido es inversamente proporcional a la concentración de antígeno en la muestra.An "immunoradiometric assay" (IRMA) is an immunoassay in which the antibody reagent is radioactively labeled. An IRMA requires the production of a multivalent antigen conjugate, by techniques such as conjugation to a protein, for example, rabbit serum albumin (RSA). The multivalent antigenic conjugate must have at least 2 antigen residues per molecule and the antigen residues must be separated at a sufficient distance to allow the binding of at least two antibodies to the antigen. For example, in an IRMA the multivalent antigen conjugate may be attached to a solid surface such as a plastic sphere. Unlabeled "sample" antigen and antibody for the antigen that is radioactively labeled are added to a test tube containing the sphere coated with the multivalent antigen conjugate. The antigen in the sample competes with the multivalent antigen conjugate for the binding sites of the antibody to the antigen. After an appropriate incubation period, unbound reagents are removed by washing and the amount of radioactivity in the solid phase is determined. The amount of radioactive antibody bound is inversely proportional to the concentration of antigen in the sample.
Se pueden usar otras técnicas para detectar los niveles de proteína de un biomarcador en una muestra biológica, se pueden realizar según las preferencias del que practica, y basado en la presente divulgación y el tipo de muestra biológica (es decir, plasma, orina, muestra de tejido etc.). Una de tales técnicas es la inmunotransferencia (Towbin et at., Proc. Nat. Acad. Sci. 76:4350 (1979)), en donde una muestra tratada de una forma adecuada se corre en un gel de SDS-PAGE antes de ser transferida a un soporte sólido, tal como un filtro de nitrocelulosa. Se pueden usar después anticuerpos anti-enzima marcados de forma detectable para evaluar los niveles de enzima, donde la intensidad de la señal de la marca detectable corresponde a la cantidad de enzima presente. Los niveles se pueden cuantificar, por ejemplo mediante densitometría.Other techniques can be used to detect a biomarker's protein levels in a biological sample, can be performed according to the preferences of the practitioner, and based on the present disclosure and the type of biological sample (i.e., plasma, urine, sample of tissue etc.). One such technique is immunoblotting (Towbin et al., Proc. Nat. Acad. Sci. 76: 4350 (1979)), where a sample treated in a suitable manner is run on an SDS-PAGE gel before being transferred to a solid support, such as a nitrocellulose filter. Detectable labeled anti-enzyme antibodies can then be used to assess enzyme levels, where the intensity of The detectable brand signal corresponds to the amount of enzyme present. The levels can be quantified, for example by densitometry.
En una forma de realización, los niveles de biomarcadores como se divulgan aquí, y/o de sus polipéptidos se pueden detectar en una muestra de tejido mediante espectrometría de masas tal como MALDI/TOF (tiempo de vuelo), SELDI/TOF, cromatografía liquida- espectrometría de masas (LC-MS), cromatografía de gases-espectrometría de masas (GC-MS), cromatografía líquida de alta resolución-espectrometría de masas (HPLC- MS), electroforesis capilar- espectrometría de masas, espectrometría de resonancia magnética nuclear, o espectrometría de masas en tándem (por ejemplo MS/MS, MS/MS/MS, ESI-MS/MS, etc.). Ver por ejemplo, las solicitudes de patente de EE.UU. Nos: 20030199001, 20030134304, 20030077616, que se incorporan aquí mediante referencia.In one embodiment, the levels of biomarkers as disclosed herein, and / or their polypeptides can be detected in a tissue sample by mass spectrometry such as MALDI / TOF (flight time), SELDI / TOF, liquid chromatography. - mass spectrometry (LC-MS), gas chromatography-mass spectrometry (GC-MS), high performance liquid chromatography-mass spectrometry (HPLC-MS), capillary electrophoresis-mass spectrometry, nuclear magnetic resonance spectrometry , or tandem mass spectrometry (for example MS / MS, MS / MS / MS, ESI-MS / MS, etc.). See for example, US patent applications. Nos: 20030199001, 20030134304, 20030077616, which are incorporated herein by reference.
Los métodos de espectrometría de masas son bien conocidos en la técnica y se han usado para cuantificar y/o identificar biomoléculas, tal como proteínas (ver, por ejemplo, Li et al. (2000) Tibtech 18:151-160; Rowley et al. (2000) Methods 20: 383- 397; y Kuster y Mann (1998) Curr. Opin. Structural Biol. 8: 393-400). Además, se han desarrollado técnicas de espectrometría de masas que permiten la secuenciación de novo al menos parcial de proteínas aisladas. Chait et al., Science 262:89-92 (1993); Keough et al, Proc. Nati. Acad. Sci. USA. 96:7131-6 (1999); revisado en Bergman, EXS 88:133-44 (2000).Mass spectrometry methods are well known in the art and have been used to quantify and / or identify biomolecules, such as proteins (see, for example, Li et al. (2000) Tibtech 18: 151-160; Rowley et al . (2000) Methods 20: 383-397; and Kuster and Mann (1998) Curr. Opin. Structural Biol. 8: 393-400). In addition, mass spectrometry techniques have been developed that allow at least partial de novo sequencing of isolated proteins. Chait et al., Science 262: 89-92 (1993); Keough et al, Proc. Nati Acad. Sci. USA. 96: 7131-6 (1999); reviewed in Bergman, EXS 88: 133-44 (2000).
En ciertas formas de realización, se usa un espectrómetro de iones en fase gaseosa. En otras formas de realización, se usa espectrometría de masas por desorción/ionización láser para analizar la muestra. Se puede llevar a cabo espectrometría de masas por desorción/ionización láser ("LDI-MS") moderna en dos variaciones principales: espectrometría de masas por desorción/ionización láser asistida por matriz ("MALDI") y desorción/ionización láser aumentada por superficie ("SELDI"). En MALDI, el analito se mezcla con una solución que contiene una matriz, y se coloca una gota de líquido en la superficie de un sustrato. La solución de la matriz co-cristaliza entonces con las moléculas biológicas. El sustrato se inserta en el espectrómetro de masas. La energía láser se dirige a la superficie del sustrato donde desorbe e ioniza las moléculas biológicas sin fragmentarlas significativamente. Ver, por ejemplo la patente de EE.UU. No. 5118937, y la patente de EE.UU. No. 5045694.In certain embodiments, a gas phase ion spectrometer is used. In other embodiments, laser desorption / ionization mass spectrometry is used to analyze the sample. Modern laser desorption / ionization ("LDI-MS") mass spectrometry can be carried out in two main variations: matrix-assisted laser desorption / ionization mass spectrometry ("MALDI") and surface-enhanced laser desorption / ionization ("SELDI"). In MALDI, the analyte is mixed with a solution containing a matrix, and a drop of liquid is placed on the surface of a substrate. The matrix solution then co-crystallizes with the biological molecules. The substrate is inserted into the mass spectrometer. The Laser energy is directed to the surface of the substrate where it desorbs and ionizes the biological molecules without significantly fragmenting them. See, for example, US Pat. No. 5118937, and U.S. Pat. No. 5045694.
En SELDI, la superficie del sustrato se modifica de modo que es un participante activo en el proceso de desorción. En una variante, la superficie se deriva con un adsorbente y/o reactivos de captura que se unen selectivamente a la proteína de interés. En otra variante, la superficie se deriva con moléculas que absorben energía que no se desorben cuando son alcanzadas por el láser. En otra variante, la superficie se deriva con moléculas que se unen a la proteína de interés y que contienen un enlace fotolítico que se rompe tras la aplicación del láser. En cada uno de estos métodos, el agente de derivación generalmente se localiza en una localización específica en la superficie del sustrato donde se aplica la muestra. Ver, por ejemplo la patente de EE.UU. No. 5719060 y WO 98/59361. Los dos métodos se pueden combinar mediante, por ejemplo, el uso de una superficie de afinidad SELDI para capturar un analito y adición de un líquido que contiene matriz al analito capturado para proporcionar el material absorbente de energía.In SELDI, the surface of the substrate is modified so that it is an active participant in the desorption process. In a variant, the surface is derived with an adsorbent and / or capture reagents that selectively bind to the protein of interest. In another variant, the surface is derived with molecules that absorb energy that are not desorbed when they are reached by the laser. In another variant, the surface is derived with molecules that bind to the protein of interest and that contain a photolytic bond that breaks after laser application. In each of these methods, the bypass agent is generally located at a specific location on the surface of the substrate where the sample is applied. See, for example, US Pat. No. 5719060 and WO 98/59361. The two methods can be combined by, for example, the use of a SELDI affinity surface to capture an analyte and adding a matrix containing liquid to the captured analyte to provide the energy absorbing material.
Para información adicional respecto a espectrómetros de masas, ver, por ejemplo, Principies of Instrumental Analysis, 3a edición., Skoog, Saunders College Publishing, Filadelfia, 1985; y Kirk-Othmer Encyclopedia of Chemical Technology, 4a ed. VoI. 15 (John Wiley AND Sons, Nueva York 1995), pp. 1071-1094.For additional information regarding mass spectrometers, see, e.g., Principles of Instrumental Analysis, 3rd edition, Skoog, Saunders College Publishing, Philadelphia, 1985.; and Kirk-Othmer Encyclopedia of Chemical Technology, 4 ed. VoI 15 (John Wiley AND Sons, New York 1995), pp. 1071-1094.
La detección de los niveles del biomarcador típicamente dependerá de la detección de la intensidad de señal. Esto, de hecho, puede reflejar la cantidad y carácter de un polipéptido unido al sustrato. Por ejemplo, en ciertas formas de realización, se puede comparar la fuerza de la señal de valores pico de los espectros de una primera muestra y una segunda muestra (por ejemplo, visualmente, mediante análisis por ordenador, etc.), para determinar las cantidades relativas de biomoléculas particulares. Se pueden usar programas de software tal como el programa Biomarker Wizard (Ciphergen Biosystems, Inc., Fremont, Calif.) como ayuda en el análisis de los espectros de masa. Los espectrómetros de masa y sus técnicas son bien conocidos para los expertos en la materia. De forma alternativa, los niveles de un biomarcador polipéptido se pueden determinar obteniendo un espectro de espectrometría de masas por desorción/ionización láser aumentada por superficie tiempo de vuelo (SELDI-TOF MS) de la muestra.The detection of biomarker levels will typically depend on the detection of signal strength. This, in fact, may reflect the amount and character of a polypeptide bound to the substrate. For example, in certain embodiments, the strength of the peak value signal of the spectra of a first sample and a second sample (for example, visually, by computer analysis, etc.) can be compared, to determine the quantities relative of particular biomolecules. Software programs such as the Biomarker Wizard program (Ciphergen Biosystems, Inc., Fremont, Calif.) Can be used as an aid in the analysis of mass spectra. Mass spectrometers and their techniques are well known to those skilled in the art. Alternatively, the levels of a polypeptide biomarker can be determine obtaining a spectrometry spectrum of mass by laser desorption / ionization increased by surface flight time (SELDI-TOF MS) of the sample.
El experto en la materia apreciará que la detección, diagnóstico y evaluación se llevará a cabo detectando un descenso en la expresión de uno o más de los biomarcadores de la invención con respecto al valor de referencia. Un descenso en la expresión con el valor de referencia se considera en donde la diferencia entre la muestra de ensayo y la muestra de referencia de al menos 0,9 veces, 0,75 veces, 0,2 veces, 0,1 veces, 0,05 veces, 0,025 veces, 0,02 veces, 0,01 veces, 0,005 veces o incluso menos.The person skilled in the art will appreciate that the detection, diagnosis and evaluation will be carried out by detecting a decrease in the expression of one or more of the biomarkers of the invention with respect to the reference value. A decrease in expression with the reference value is considered where the difference between the test sample and the reference sample of at least 0.9 times, 0.75 times, 0.2 times, 0.1 times, 0 , 05 times, 0.025 times, 0.02 times, 0.01 times, 0.005 times or even less.
Si el método de la invención se lleva a cabo para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis, entonces el valor de referencia es el nivel de expresión del biomarcador en un muestra de referencia que se obtiene combinando cantidades iguales de muestras de una población de sujetos. En general, las muestras de referencia típicas se obtendrán de sujetos que están clínicamente bien documentados y que están libres de la enfermedad. En tales muestras, las concentraciones normales (de referencia) del biomarcador se pueden determinar, por ejemplo proporcionando la concentración media sobre la población de referencia. Al determinar la concentración de referencia del marcador se toman en cuenta varias consideraciones. Entre tales consideraciones están el tipo de muestra implicada (por ejemplo tejido o LCR), la edad, peso, sexo, estado físico general del paciente y similares. Por ejemplo, se toman como grupo de referencia cantidades iguales de un grupo de al menos 2, al menos 10, al menos 100 a preferiblemente más de 1000 sujetos, preferiblemente clasificados según las consideraciones anteriores, por ejemplo de varias categorías de edad.If the method of the invention is carried out for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis, then the reference value is the level of expression of the biomarker in a reference sample that is obtained by combining equal amounts of samples from a population of subjects. In general, typical reference samples will be obtained from subjects that are clinically well documented and free of the disease. In such samples, normal (reference) concentrations of the biomarker can be determined, for example by providing the average concentration over the reference population. When determining the reference concentration of the marker, several considerations are taken into account. Among such considerations are the type of sample involved (eg tissue or CSF), age, weight, sex, general physical condition of the patient and the like. For example, equal amounts of a group of at least 2, at least 10, at least 100 to preferably more than 1000 subjects are taken as reference group, preferably classified according to the above considerations, for example of various age categories.
Si el método de la invención se lleva a cabo para determinar la eficacia de una terapia para el tratamiento de fibrosis o de una enfermedad asociada con fibrosis, entonces el valor de referencia usado para determinar si una terapia es eficaz normalmente es el nivel de expresión del biomarcador o biomarcadores en consideración con una muestra del paciente antes del inicio de la terapia. Según este método de la invención, la variación con respecto al valor de referencia es un aumento en la expresión por encima del valor de referencia. El nivel de expresión se considera aumentado en donde la diferencia entre la muestra de ensayo y la muestra de referencia es al menos 1,1 veces,If the method of the invention is carried out to determine the efficacy of a therapy for the treatment of fibrosis or a disease associated with fibrosis, then the reference value used to determine whether a therapy is effective is usually the level of expression of the biomarker or biomarkers under consideration with a patient sample before the start of therapy. According to this method of the invention, the variation with respect to the reference value is an increase in the expression above of the reference value. The level of expression is considered to be increased where the difference between the test sample and the reference sample is at least 1.1 times,
1,5 veces, 5 veces, 10 veces, 20 veces, 30 veces, 40 veces, 50 veces, 60 veces, 70 veces,1.5 times, 5 times, 10 times, 20 times, 30 times, 40 times, 50 times, 60 times, 70 times,
80 veces, 90 veces, 100 veces o incluso más.80 times, 90 times, 100 times or even more.
Se entenderá que el método de la invención se puede llevar a cabo determinando el nivel de un número variable de los biomarcadores. Por ejemplo, el/los nivel(es) de un biomarcador, dos o más biomarcadores, tres o más biomarcadores o cuatro o más biomarcadores como se definen en la presente invención. La determinación de niveles de combinaciones de biomarcadores puede permitir mayor sensibilidad y especificidad en el diagnóstico de la fibrosis y/o una enfermedad asociada con fibrosis o puede permitir mejor diferenciación de fibrosis de otras enfermedades que pueden tener biomarcadores similares o solapantes. De esta manera, la presente invención comprende la determinación simultánea de los siguientes grupos de biomarcadores:It will be understood that the method of the invention can be carried out by determining the level of a variable number of biomarkers. For example, the level (s) of a biomarker, two or more biomarkers, three or more biomarkers or four or more biomarkers as defined in the present invention. The determination of levels of combinations of biomarkers may allow greater sensitivity and specificity in the diagnosis of fibrosis and / or a disease associated with fibrosis or may allow better differentiation of fibrosis from other diseases that may have similar or overlapping biomarkers. Thus, the present invention comprises the simultaneous determination of the following groups of biomarkers:
SLURPl y el fragmento SLURPl,SLURPl and the SLURPl fragment,
SLURPI y HAMP,SLURPI and HAMP,
SLURPl y Hepc25,SLURPl and Hepc25,
SLURPl y Hepc20, SLURPI y GSN;SLURPl and Hepc20, SLURPI and GSN;
SLURPI y APOD;SLURPI and APOD;
SLURPI y SPPl,SLURPI and SPPl,
SLURPI y DEFBl,SLURPI and DEFBl,
SLURPl y MASP2; fragmento SLURPl y HAMP, fragmento SLURPl y Hepc25, fragmento SLURPl y Hepc20, fragmento SLURPl y GSN, fragmento SLURPl y APOD, fragmento SLURP 1 y SPP 1 , fragmento SLURPl y DEFBl, fragmento SLURPl y MASP2; HAMP y Hepc25,SLURPl and MASP2; SLURPl and HAMP fragment, SLURPl and Hepc25 fragment, SLURPl and Hepc20 fragment, SLURPl and GSN fragment, SLURPl and APOD fragment, SLURP 1 and SPP 1 fragment, SLURPl and DEFBl fragment, SLURPl and MASP2 fragment; HAMP and Hepc25,
HAMP y Hepc20,HAMP and Hepc20,
HAMP y GSN,HAMP and GSN,
HAMP y APOD, HAMPySPPl,HAMP and APOD, HAMPySPPl,
HAMPyDEFBl,HAMPyDEFBl,
HAMP y MASP2;HAMP and MASP2;
Hepc25 y Hepc20,Hepc25 and Hepc20,
Hepc25 y GSN, Hepc25yAPOD,Hepc25 and GSN, Hepc25yAPOD,
Hepc25ySPPl,Hepc25ySPPl,
Hepc25yDEFBl,Hepc25yDEFBl,
Hepc25 y MASP2;Hepc25 and MASP2;
Hepc20 y GSN, Hepc20yAPOD,Hepc20 and GSN, Hepc20yAPOD,
Hepc20ySPPl,Hepc20ySPPl,
Hepc20yDEFBl,Hepc20yDEFBl,
Hepc20 y MASP2Hepc20 and MASP2
GSN y APOD; GSNySPPl,GSN and APOD; GSNySPPl,
GSNyDEFBl,GSNyDEFBl,
GSN y MASP2,GSN and MASP2,
APODySPPl,APODySPPl,
APODyDEFBl, APOD y MASP2,APODyDEFBl, APOD and MASP2,
SLURPl, fragmento SLURPl y HAMP;SLURPl, SLURPl and HAMP fragment;
SLURPl, fragmento SLURPl y Hepc25;SLURPl, SLURPl and Hepc25 fragment;
SLURPl, fragmento SLURPl y Hepc20;SLURPl, SLURPl and Hepc20 fragment;
SLURPl, fragmento SLURPl y GSN; SLURP 1 , fragmento SLURP 1 y APOD;SLURPl, SLURPl and GSN fragment; SLURP 1, SLURP 1 fragment and APOD;
SLURPl, fragmento SLURPl y SPPl;SLURPl, SLURPl and SPPl fragment;
SLURPl, fragmento SLURPl y DEFBl; SLURPl, fragmento SLURPl y MASP2;SLURPl, SLURPl and DEFBl fragment; SLURPl, SLURPl and MASP2 fragment;
SLURPl, HAMP y Hepc25;SLURPl, HAMP and Hepc25;
SLURPl, HAMP y Hepc20;SLURPl, HAMP and Hepc20;
SLURPl5 HAMP y GSN; SLURP 1 , HAMP y APOD;SLURPl 5 HAMP and GSN; SLURP 1, HAMP and APOD;
SLURPl5 HAMP y SPPlSLURPl 5 HAMP and SPPl
SLURPl, HAMP y DEFBlSLURPl, HAMP and DEFBl
SLURPl, HAMP y MASP2SLURPl, HAMP and MASP2
SLURPl, Hepc25 y Hepc20; SLURP 1 , Hepc25 y GSN;SLURPl, Hepc25 and Hepc20; SLURP 1, Hepc25 and GSN;
SLURPl, Hepc25 y APOD;SLURPl, Hepc25 and APOD;
SLURPl, Hepc25 y SPPlSLURPl, Hepc25 and SPPl
SLURPl, Hepc25 y DEFBlSLURPl, Hepc25 and DEFBl
SLURPl, Hepc25 y MASP2 SLURP 1 , Hepc20 y GSN;SLURPl, Hepc25 and MASP2 SLURP 1, Hepc20 and GSN;
SLURPl, Hepc20 y APOD;SLURPl, Hepc20 and APOD;
SLURPl, Hepc20 y SPPlSLURPl, Hepc20 and SPPl
SLURPl, Hepc20 y DEFBlSLURPl, Hepc20 and DEFBl
SLURPl, Hepc20 y MASP2 SLURP 1 , GSN y APOD;SLURPl, Hepc20 and MASP2 SLURP 1, GSN and APOD;
SLURPI5 GSN y SPPl;SLURPI 5 GSN and SPPl;
SLURPl5 GSN y DEFBl;SLURPl 5 GSN and DEFBl;
SLURPl5 GSN y MASP2; fragmento SLURPl5 HAMP y Hepc25; fragmento SLURP 1 , HAMP y Hepc20; fragmento SLURPl5 HAMP y GSN; fragmento SLURPl5 HAMP y APOD; fragmento SLURPl5 HAMP y SPPl; fragmento SLURPl5 HAMP y DEFBl; fragmento SLURP 1 , HAMP y MASP2; fragmento SLURPl5 Hepc25 y Hepc20; fragmento SLURPl5 Hepc25 y GSN; fragmento SLURPl, Hepc25 y APOD; fragmento SLURPl, Hepc25 y SPPl fragmento SLURPl, Hepc25 y DEFBl fragmento SLURPl, Hepc25 y MASP2 fragmento SLURP 1 , Hepc20 y GSN; fragmento SLURPl, Hepc20 y APOD; fragmento SLURPl, Hepc20 y SPPl fragmento SLURPl, Hepc20 y DEFBl fragmento SLURPl, Hepc20 y MASP2 fragmento SLURP 1 , GSN y APOD; fragmento SLURPl, GSN y SPPl; fragmento SLURPl, GSN y DEFBl; fragmento SLURPl, GSN y MASP2;SLURPl 5 GSN and MASP2; SLURPl 5 HAMP and Hepc25 fragment; SLURP 1, HAMP and Hepc20 fragment; SLURPl 5 HAMP and GSN fragment; SLURPl 5 HAMP and APOD fragment; SLURPl 5 HAMP and SPPl fragment; SLURPl 5 HAMP and DEFBl fragment; SLURP 1, HAMP and MASP2 fragment; SLURPl 5 Hepc25 and Hepc20 fragment; SLURPl 5 Hepc25 and GSN fragment; SLURPl, Hepc25 and APOD fragment; SLURPl fragment, Hepc25 and SPPl SLURPl fragment, Hepc25 and DEFBl SLURPl fragment, Hepc25 and MASP2 SLURP fragment 1, Hepc20 and GSN; SLURPl, Hepc20 and APOD fragment; SLURPl fragment, Hepc20 and SPPl SLURPl fragment, Hepc20 and DEFBl SLURPl fragment, Hepc20 and MASP2 SLURP fragment 1, GSN and APOD; SLURPl, GSN and SPPl fragment; SLURPl, GSN and DEFBl fragment; SLURPl, GSN and MASP2 fragment;
HAMP, Hepc25 y Hepc20; HAMP, Hepc25 y GSN;HAMP, Hepc25 and Hepc20; HAMP, Hepc25 and GSN;
HAMP, Hepc25 y APOD;HAMP, Hepc25 and APOD;
HAMP, Hepc25 y SPPl;HAMP, Hepc25 and SPPl;
HAMP, Hepc25 y DEFBl;HAMP, Hepc25 and DEFBl;
HAMP, Hepc25 y MASP2; HAMP, Hepc20 y GSN;HAMP, Hepc25 and MASP2; HAMP, Hepc20 and GSN;
HAMP, Hepc20 y APOD;HAMP, Hepc20 and APOD;
HAMP, Hepc20 y SPPl;HAMP, Hepc20 and SPPl;
HAMP, Hepc20 y DEFBl;HAMP, Hepc20 and DEFBl;
HAMP, Hepc20 y MASP2; HAMP, GSN y APOD;HAMP, Hepc20 and MASP2; HAMP, GSN and APOD;
HAMP5 GSN y SPPl;HAMP 5 GSN and SPPl;
HAMP5 GSN y DEFBl;HAMP 5 GSN and DEFBl;
HAMP, GSN y MASP2;HAMP, GSN and MASP2;
Hepc25, Hepc20 y GSN; Hepc25, Hepc20 y APOD;Hepc25, Hepc20 and GSN; Hepc25, Hepc20 and APOD;
Hepc25, Hepc20 y SPPl;Hepc25, Hepc20 and SPPl;
Hepc25, Hepc20 y DEFBl; Hepc25, Hepc20 y MASP2;Hepc25, Hepc20 and DEFBl; Hepc25, Hepc20 and MASP2;
Hepc25, GSN y APOD;Hepc25, GSN and APOD;
Hepc25, GSN y SPPl;Hepc25, GSN and SPPl;
Hepc25, GSN y DEFBl; Hepc25, GSN y MASP2;Hepc25, GSN and DEFBl; Hepc25, GSN and MASP2;
SLURPl, fragmento SLURPl, HAMP y Hepc25;SLURPl, SLURPl fragment, HAMP and Hepc25;
SLURPl, fragmento SLURPl, HAMP yHepc20;SLURPl, SLURPl fragment, HAMP and Hepc20;
SLURPl, fragmento SLURPl, HAMP y GSN;SLURPl, SLURPl fragment, HAMP and GSN;
SLURPl, fragmento SLURPl, HAMP y APOD; SLURP 1 , fragmento SLURP 1 , HAMP y SPP 1 ;SLURPl, SLURPl, HAMP and APOD fragment; SLURP 1, SLURP 1 fragment, HAMP and SPP 1;
SLURPl, fragmento SLURPl, HAMP y DEFBl;SLURPl, SLURPl, HAMP and DEFBl fragment;
SLURPl, fragmento SLURPl, HAMP y MASP2;SLURPl, SLURPl fragment, HAMP and MASP2;
SLURPl, fragmento SLURPl, Hepc25 y Hepc20;SLURPl, SLURPl fragment, Hepc25 and Hepc20;
SLURPl, fragmento SLURPl, Hepc25 y GSN; SLURP 1 , fragmento SLURP 1 , Hepc25 y APOD;SLURPl, SLURPl fragment, Hepc25 and GSN; SLURP 1, fragment SLURP 1, Hepc25 and APOD;
SLURPl, fragmento SLURPl, Hepc25 y SPPl;SLURPl, SLURPl, Hepc25 and SPPl fragment;
SLURPl, fragmento SLURPl, Hepc25 y DEFBl;SLURPl, SLURPl, Hepc25 and DEFBl fragment;
SLURPl, fragmento SLURPl, Hepc25 y MASP2;SLURPl, SLURPl, Hepc25 and MASP2 fragment;
SLURPl, fragmento SLURPl, Hepc20 y GSN; SLURP 1 , fragmento SLURP 1 , Hepc20 y APOD;SLURPl, SLURPl fragment, Hepc20 and GSN; SLURP 1, SLURP 1 fragment, Hepc20 and APOD;
SLURPl, fragmento SLURPl, Hepc20 y SPPl;SLURPl, SLURPl, Hepc20 and SPPl fragment;
SLURPl, fragmento SLURPl, Hepc20 y DEFBl;SLURPl, SLURPl, Hepc20 and DEFBl fragment;
SLURPl, fragmento SLURPl, Hepc20 y MASP2;SLURPl, SLURPl, Hepc20 and MASP2 fragment;
SLURPl, HAMP, Hepc25 y Hepc20; SLURP 1 , HAMP, Hepc25 y GSN;SLURPl, HAMP, Hepc25 and Hepc20; SLURP 1, HAMP, Hepc25 and GSN;
SLURPl, HAMP, Hepc25 y APOD;SLURPl, HAMP, Hepc25 and APOD;
SLURPl, HAMP, Hepc25 y SPPl;SLURPl, HAMP, Hepc25 and SPPl;
SLURPl, HAMP, Hepc25 y DEFBl;SLURPl, HAMP, Hepc25 and DEFBl;
SLURPl, HAMP, Hepc25 y MASP2; SLURPl, HAMP, Hepc20 y GSN;SLURPl, HAMP, Hepc25 and MASP2; SLURPl, HAMP, Hepc20 and GSN;
SLURPl, HAMP, Hepc20 y APOD;SLURPl, HAMP, Hepc20 and APOD;
SLURPl, HAMP, Hepc20 y SPPl; SLURPl, HAMP, Hepc20 y DEFBl;SLURPl, HAMP, Hepc20 and SPPl; SLURPl, HAMP, Hepc20 and DEFBl;
SLURPl, HAMP, Hepc20 y MASP2;SLURPl, HAMP, Hepc20 and MASP2;
SLURPl, HAMP, GSN y APOD;SLURPl, HAMP, GSN and APOD;
SLURPl, HAMP, GSN y SPPl; SLURP 1 , HAMP, GSN y DEFB 1 ;SLURPl, HAMP, GSN and SPPl; SLURP 1, HAMP, GSN and DEFB 1;
SLURPl, HAMP, GSN y MASP2; fragmento SLURPl, HAMP, Hepc25 y Hepc20; fragmento SLURPl, HAMP, Hepc25 y GSN; fragmento SLURPl, HAMP, Hepc25 y APOD; fragmento SLURP 1 , HAMP, Hepc25 y SPP 1 ; fragmento SLURPl, HAMP, Hepc25 y DEFBl; fragmento SLURPl, HAMP, Hepc25 y MASP2; fragmento SLURPl, HAMP, Hepc20 y GSN; fragmento SLURPl, HAMP, Hepc20 y APOD; fragmento SLURP 1 , HAMP, Hepc20 y SPP 1 ; fragmento SLURPl, HAMP, Hepc20 y DEFBl; fragmento SLURPl, HAMP, Hepc20 y MASP2; fragmento SLURPl, HAMP, GSN y APOD; fragmento SLURPl, HAMP, GSN y SPPl; fragmento SLURP 1 , HAMP, GSN y DEFB 1 ; fragmento SLURPl, HAMP, GSN y MASP2; fragmento SLURPl, Hepc25, Hepc20 y GSN; fragmento SLURPl, Hepc25, Hepc20 y APOD; fragmento SLURPl, Hepc25, Hepc20 y SPPl; fragmento SLURP 1 , Hepc25 , Hepc20 y DEFB 1 ; fragmento SLURPl, Hepc25, Hepc20 y MASP2; fragmento SLURPl, Hepc20, GSN y APOD; fragmento SLURPl, Hepc20, GSN y SPPl; fragmento SLURPl, Hepc20, GSN y DEFBl; fragmento SLURP 1 , Hepc20, GSN y MASP2;SLURPl, HAMP, GSN and MASP2; SLURP1, HAMP, Hepc25 and Hepc20 fragment; SLURPl, HAMP, Hepc25 and GSN fragment; SLURPl, HAMP, Hepc25 and APOD fragment; SLURP 1, HAMP, Hepc25 and SPP 1 fragment; SLURPl, HAMP, Hepc25 and DEFBl fragment; SLURPl, HAMP, Hepc25 and MASP2 fragment; SLURPl, HAMP, Hepc20 and GSN fragment; SLURPl, HAMP, Hepc20 and APOD fragment; SLURP 1, HAMP, Hepc20 and SPP 1 fragment; SLURPl, HAMP, Hepc20 and DEFBl fragment; SLURPl, HAMP, Hepc20 and MASP2 fragment; SLURPl, HAMP, GSN and APOD fragment; SLURPl, HAMP, GSN and SPPl fragment; SLURP 1, HAMP, GSN and DEFB 1 fragment; SLURPl, HAMP, GSN and MASP2 fragment; SLURP1, Hepc25, Hepc20 and GSN fragment; SLURPl, Hepc25, Hepc20 and APOD fragment; SLURPl, Hepc25, Hepc20 and SPPl fragment; SLURP 1, Hepc25, Hepc20 and DEFB 1 fragment; SLURPl, Hepc25, Hepc20 and MASP2 fragment; SLURPl, Hepc20, GSN and APOD fragment; SLURPl, Hepc20, GSN and SPPl fragment; SLURPl, Hepc20, GSN and DEFBl fragment; SLURP 1, Hepc20, GSN and MASP2 fragment;
HAMP, Hepc25, Hepc20 y GSN;HAMP, Hepc25, Hepc20 and GSN;
HAMP, Hepc25, Hepc20 y APOD; HAMP, Hepc25, Hepc20 y SPPl;HAMP, Hepc25, Hepc20 and APOD; HAMP, Hepc25, Hepc20 and SPPl;
HAMP, Hepc25, Hepc20 y DEFBl;HAMP, Hepc25, Hepc20 and DEFBl;
HAMP, Hepc25, Hepc20 y MASP2;HAMP, Hepc25, Hepc20 and MASP2;
Hepc25, Hepc20, GSN y APOD Hepc25, Hepc20, GSN y SPPlHepc25, Hepc20, GSN and APOD Hepc25, Hepc20, GSN and SPPl
Hepc25, Hepc20, GSN y DEFBlHepc25, Hepc20, GSN and DEFBl
Hepc25, Hepc20, GSN y MASP2Hepc25, Hepc20, GSN and MASP2
SLURPl, fragmento SLURPl, HAMP, Hepc25 y Hepc20;SLURPl, SLURPl fragment, HAMP, Hepc25 and Hepc20;
SLURPl, fragmento SLURPl, HAMP, Hepc25 y GSN; SLURP 1 , fragmento SLURP 1 , HAMP, Hepc25 y APOD;SLURPl, SLURPl fragment, HAMP, Hepc25 and GSN; SLURP 1, SLURP 1 fragment, HAMP, Hepc25 and APOD;
SLURPl, fragmento SLURPl, HAMP, Hepc25 y SPPl;SLURPl, SLURPl fragment, HAMP, Hepc25 and SPPl;
SLURPl, fragmento SLURPl, HAMP, Hepc25 y DEFBl;SLURPl, SLURPl, HAMP, Hepc25 and DEFBl fragment;
SLURPl, fragmento SLURPl, HAMP, Hepc25 y MASP2;SLURPl, SLURPl fragment, HAMP, Hepc25 and MASP2;
SLURPl, fragmento SLURPl, HAMP, Hepc20 y GSN; SLURP 1 , fragmento SLURP 1 , HAMP, Hepc20 y APOD;SLURPl, SLURPl, HAMP, Hepc20 and GSN fragment; SLURP 1, SLURP 1 fragment, HAMP, Hepc20 and APOD;
SLURPl, fragmento SLURPl, HAMP, Hepc20 y SPPl;SLURPl, SLURPl, HAMP, Hepc20 and SPPl fragment;
SLURPl, fragmento SLURPl, HAMP, Hepc20 y DEFBl;SLURPl, SLURPl, HAMP, Hepc20 and DEFBl fragment;
SLURPl, fragmento SLURPl, HAMP, Hepc20 y MASP2;SLURPl, SLURPl fragment, HAMP, Hepc20 and MASP2;
SLURPl, fragmento SLURPl, HAMP, GSN y APOD; SLURP 1 , fragmento SLURP 1 , HAMP, GSN y SPP 1 ;SLURPl, SLURPl, HAMP, GSN and APOD fragment; SLURP 1, SLURP 1 fragment, HAMP, GSN and SPP 1;
SLURPl, fragmento SLURPl, HAMP, GSN y DEFBl;SLURPl, SLURPl, HAMP, GSN and DEFBl fragment;
SLURPl, fragmento SLURPl, HAMP, GSN y MASP2;SLURPl, SLURPl, HAMP, GSN and MASP2 fragment;
SLURPl, fragmento SLURPl, Hepc25, Hepc20 y GSN;SLURPl, SLURPl, Hepc25, Hepc20 and GSN fragment;
SLURPl, fragmento SLURPl, Hepc25, Hepc20 y APOD; SLURP 1 , fragmento SLURP 1 , Hepc20, GSN y APOD; fragmento SLURPl, HAMP, Hepc25, Hepc20 y GSN, fragmento SLURPl, HAMP, Hepc25, Hepc20 y APOD; fragmento SLURPl, HAMP, Hepc25, Hepc20 y SPPl; fragmento SLURPl, HAMP, Hepc25, Hepc20 y DEFBl; fragmento SLURPl, HAMP, Hepc25, Hepc20 y MASP2; fragmento SLURPl, Hepc25, Hepc20, GSN y APOD; fragmento SLURPl, Hepc25, Hepc20, GSN y SPPl; fragmento SLURPl, Hepc25, Hepc20, GSN y DEFBl; fragmento SLURPl, Hepc25, Hepc20, GSN y MASP2SLURPl, SLURPl fragment, Hepc25, Hepc20 and APOD; SLURP 1, SLURP 1 fragment, Hepc20, GSN and APOD; SLURPl, HAMP, Hepc25, Hepc20 and GSN fragment, SLURPl, HAMP, Hepc25, Hepc20 and APOD fragment; SLURPl, HAMP, Hepc25, Hepc20 and SPPl fragment; SLURPl, HAMP, Hepc25, Hepc20 and DEFBl fragment; SLURP1, HAMP, Hepc25, Hepc20 and MASP2 fragment; SLURPl, Hepc25, Hepc20, GSN and APOD fragment; SLURPl, Hepc25, Hepc20, GSN and SPPl fragment; SLURPl, Hepc25, Hepc20, GSN and DEFBl fragment; SLURPl, Hepc25, Hepc20, GSN and MASP2 fragment
HAMP, Hepc25, Hepc20, GSN y APOD;HAMP, Hepc25, Hepc20, GSN and APOD;
HAMP, Hepc25, Hepc20, GSN y SPPl; HAMP, Hepc25, Hepc20, GSN y DEFBl;HAMP, Hepc25, Hepc20, GSN and SPPl; HAMP, Hepc25, Hepc20, GSN and DEFBl;
HAMP, Hepc25, Hepc20, GSN y MASP2;HAMP, Hepc25, Hepc20, GSN and MASP2;
HAMP, Hepc20, GSN y APOD;HAMP, Hepc20, GSN and APOD;
Hepc25, Hepc20, GSN y APOD;Hepc25, Hepc20, GSN and APOD;
Hepc25, Hepc20, GSN y SPPl; Hepc25, Hepc20, GSN y DEFBl;Hepc25, Hepc20, GSN and SPPl; Hepc25, Hepc20, GSN and DEFBl;
Hepc25, Hepc20, GSN y MASP2;Hepc25, Hepc20, GSN and MASP2;
SLURPl, fragmento SLURPl, HAMP, Hepc25, Hepc20 y GSN;SLURPl, SLURPl fragment, HAMP, Hepc25, Hepc20 and GSN;
SLURPl, fragmento SLURPl, HAMP, Hepc25, Hepc20 y APOD;SLURPl, SLURPl, HAMP, Hepc25, Hepc20 and APOD fragment;
SLURPl, fragmento SLURPl, HAMP, Hepc25, Hepc20 y SPPl; SLURP 1 , fragmento SLURP 1 , HAMP, Hepc25 , Hepc20 y DEFB 1 ;SLURPl, SLURPl, HAMP, Hepc25, Hepc20 and SPPl fragment; SLURP 1, fragment SLURP 1, HAMP, Hepc25, Hepc20 and DEFB 1;
SLURPl, fragmento SLURPl, HAMP, Hepc25, Hepc20 y MASP2;SLURPl, SLURPl, HAMP, Hepc25, Hepc20 and MASP2 fragment;
SLURPl, HAMP, Hepc25, Hepc20, GSN y APOD;SLURPl, HAMP, Hepc25, Hepc20, GSN and APOD;
SLURPl, HAMP, Hepc25, Hepc20, GSN y SPPl;SLURPl, HAMP, Hepc25, Hepc20, GSN and SPPl;
SLURPl, HAMP, Hepc25, Hepc20, GSN y DEFBl; SLURPl, HAMP, Hepc25, Hepc20, GSN y MASP2;SLURPl, HAMP, Hepc25, Hepc20, GSN and DEFBl; SLURPl, HAMP, Hepc25, Hepc20, GSN and MASP2;
SLURPl, fragmento SLURPl, Hepc25, Hepc20, GSN y APOD;SLURPl, SLURPl, Hepc25, Hepc20, GSN and APOD fragment;
SLURPl, fragmento SLURPl, Hepc25, Hepc20, GSN y SPPl;SLURPl, SLURPl fragment, Hepc25, Hepc20, GSN and SPPl;
SLURPl, fragmento SLURPl, Hepc25, Hepc20, GSN y DEFBl;SLURPl, SLURPl, Hepc25, Hepc20, GSN and DEFBl fragment;
SLURPl, fragmento SLURPl, Hepc25, Hepc20, GSN y MASP2; SLURP 1 , fragmento SLURP 1 , HAMP, Hepc20, GSN y APOD;SLURPl, SLURPl fragment, Hepc25, Hepc20, GSN and MASP2; SLURP 1, SLURP 1 fragment, HAMP, Hepc20, GSN and APOD;
SLURPl, fragmento SLURPl, HAMP, Hepc20, GSN y SPPl;SLURPl, SLURPl, HAMP, Hepc20, GSN and SPPl fragment;
SLURPl, fragmento SLURPl, HAMP, Hepc20, GSN y DEFBl;SLURPl, SLURPl fragment, HAMP, Hepc20, GSN and DEFBl;
SLURPl, fragmento SLURPl, HAMP, Hepc20, GSN y MASP2;SLURPl, SLURPl fragment, HAMP, Hepc20, GSN and MASP2;
Además, el experto en la materia apreciará que los métodos de la invención se pueden combinar con otros métodos conocidos en la técnica para la detección de fibrosis o de una enfermedad asociada a fibrosis y para la determinación de la eficacia de una terapia para fibrosis o una enfermedad asociada a fϊbrosis. En particular, los biomarcadores de la invención se pueden medir simultáneamente con la serie de 5 biomarcadores definida en WO0216949 (α2-macroglobulina, haptoglobulina, apolipoproteina Al, γ-glutamil transpeptidasa, y bilirrubina), con la serie de tres biomarcadores descrita en WO0373822 (α-MG, HA y TIMP-I), con las diferentes series de biomarcadores descritas en WO2005116901 (que comprenden α-2 macroglobulina, ácido hialurónico, apolipoproteina Al, propéptido N-terminal de colágeno de tipo III, γ- glutamiltranspeptidasa, bilirrubina, γ-globulinas, plaquetas, tiempo de protrombina, aspartato amino-transferasa, alanina aminotransferasa, urea, sodio, glucemia, triglicéridos, albúmina, fosfatasas alcalinas, YKL-40 (glicoproteína de cartílago humana 39), Inhibidor tisular de metaloproteinasa de matriz 1 (TIMP-I), metaloproteinasa de matriz 2 (MMP-2) y ferritina); con uno o más de los marcadores descritos en WO2007003670 (que incluye uromodulina, MAC2BP, AGPl y catepsina A), con uno o más de los biomarcadores de suero descritos en WO2008031051.In addition, the person skilled in the art will appreciate that the methods of the invention can be combined with other methods known in the art for the detection of fibrosis or a disease associated with fibrosis and for the determination of the efficacy of a therapy. for fibrosis or a disease associated with fibrosis. In particular, the biomarkers of the invention can be measured simultaneously with the series of 5 biomarkers defined in WO0216949 (α2-macroglobulin, haptoglobulin, apolipoprotein Al, γ-glutamyl transpeptidase, and bilirubin), with the series of three biomarkers described in WO0373822 ( α-MG, HA and TIMP-I), with the different series of biomarkers described in WO2005116901 (comprising α-2 macroglobulin, hyaluronic acid, apolipoprotein Al, N-terminal propeptide of type III collagen, γ-glutamyltranspeptidase, bilirubin, γ-globulins, platelets, prothrombin time, aspartate amino transferase, alanine aminotransferase, urea, sodium, glycemia, triglycerides, albumin, alkaline phosphatases, YKL-40 (human cartilage glycoprotein 39), Tissue matrix metalloproteinase inhibitor 1 ( TIMP-I), matrix 2 metalloproteinase (MMP-2) and ferritin); with one or more of the markers described in WO2007003670 (which includes uromodulin, MAC2BP, AGPl and cathepsin A), with one or more of the serum biomarkers described in WO2008031051.
Además, el método de la invención se puede usar junto con otros métodos para detectar fibrosis no basados en la determinación de los niveles de expresión de uno o más biomarcadores, tal como biopsia hepática y elastografía como se describe en Sandrin L et al. (Ultrasound Med. Biol. 2003; 29:1705-1711).In addition, the method of the invention can be used together with other methods to detect fibrosis not based on the determination of expression levels of one or more biomarkers, such as liver biopsy and elastography as described in Sandrin L et al. (Ultrasound Med. Biol. 2003; 29: 1705-1711).
La invención también proporciona kits que comprenden reactivos para detectar al menos dos o más biomarcadores seleccionados del grupo de SLURPl, HAMP, GSN,, APOD, SPPl, DEFBl, MASP2. Las diferentes combinaciones de biomarcadores que se pueden detectar usando el kit de la invención se han definido anteriormente. En una forma de realización preferida, los biomarcadores son polipéptidos y de esta manera, los reactivos que forman el kit pueden ser cualquier compuesto capaz de unirse con alta afinidad a los biomarcadores polipeptídicos anteriores. Los reactivos del kit pueden ser, sin limitación, cualquier tipo de anticuerpo o molécula de inmunoglobulina o fragmento de los mismos tal como anticuerpos policlonales, monoclonales, humanos o humanizados o recombinantes así como anticuerpos de cadena sencilla, por ejemplo construcciones scFv, o anticuerpos sintéticos como tal y que pueden pertenecer a cualquiera de las siguientes clases de inmunoglobulinas: IgG, IgM, IgE, IgA, y donde sea aplicable, una subclase de cualquiera de las clases mencionadas anteriormente, por ejemplo las subclases de la clase IgG tal como IgGl, IgG2, IgG2a, IgG2b, IgG3 o IgGM. Además, el kit, también puede comprender fragmentos de anticuerpo tal como fragmentos Fv, Fab o F(ab')2 o fragmentos de cadena sencilla tal como scFv. Los fragmentos de cadena doble tal como Fv, Fab o F(ab')2 son preferidos. Los fragmentos Fab y F(ab')2 no tienen fragmento Fc contenido en los anticuerpos intactos. Como consecuencia beneficiosa, tales fragmentos se transportan más rápido en el sistema circulatorio, y muestran menos unión no específica a tejido en comparación con las especies de anticuerpos completas. Tales fragmentos se pueden producir a partir de anticuerpos intactos mediante digestión proteo lítica usando proteasas tales como papaína (para la producción de fragmentos Fab) o pepsina (para la producción de fragmentos F(ab')2), u oxidación química.The invention also provides kits comprising reagents for detecting at least two or more biomarkers selected from the group of SLURPl, HAMP, GSN ,, APOD, SPPl, DEFBl, MASP2. The different combinations of biomarkers that can be detected using the kit of the invention have been defined above. In a preferred embodiment, the biomarkers are polypeptides and thus, the reagents that form the kit can be any compound capable of binding with high affinity to the above polypeptide biomarkers. The kit reagents can be, without limitation, any type of antibody or immunoglobulin molecule or fragment thereof such as polyclonal, monoclonal, human or humanized or recombinant antibodies as well as single chain antibodies, for example scFv constructs, or synthetic antibodies as such and that they can belong to any of the following classes of immunoglobulins: IgG, IgM, IgE, IgA, and where where applicable, a subclass of any of the aforementioned classes, for example subclasses of the IgG class such as IgGl, IgG2, IgG2a, IgG2b, IgG3 or IgGM. In addition, the kit may also comprise antibody fragments such as Fv, Fab or F (ab ') 2 fragments or single chain fragments such as scFv. Double chain fragments such as Fv, Fab or F (ab ') 2 are preferred. Fab and F (ab ') 2 fragments have no Fc fragment contained in intact antibodies. As a beneficial consequence, such fragments are transported faster in the circulatory system, and show less non-specific binding to tissue compared to complete antibody species. Such fragments can be produced from intact antibodies by proteolytic digestion using proteases such as papain (for the production of Fab fragments) or pepsin (for the production of F (ab ') 2 fragments), or chemical oxidation.
Los anticuerpos pueden estar unidos a un soporte sólido formando matrices de anticuerpos o chips de proteína. Las matrices de proteína son sistemas de ensayos unión de ligandos en fase sólida que usan proteínas inmovilizadas sobre superficies que incluyen vidrio, membranas, pocilios de microtitulación, placas de espectrómetro de masas, y bolas u otras partículas. Las matrices son altamente paralelas (multiplexadas) y con frecuencias miniaturizadas (micromatrices, chips de proteína). Sus ventajas incluyen que son rápidos y automatizables, capaces de gran sensibilidad, económicos en reactivos, y dan abundancia de datos para un solo experimento. El soporte bioinformático es importante; el manejo de datos demanda software sofisticado y análisis de comparación de datos. Sin embargo, el software se adapta del usado para matrices de ADN, como puede ser gran parte del hardware y sistemas de detección.The antibodies can be bound to a solid support forming antibody matrices or protein chips. Protein matrices are solid phase ligand binding assay systems that use immobilized proteins on surfaces that include glass, membranes, microtiter wells, mass spectrometer plates, and balls or other particles. The matrices are highly parallel (multiplexed) and with miniaturized frequencies (microarrays, protein chips). Its advantages include that they are fast and automatable, capable of great sensitivity, economical in reagents, and give plenty of data for a single experiment. Bioinformatic support is important; Data management demands sophisticated software and data comparison analysis. However, the software adapts to that used for DNA matrices, such as much of the hardware and detection systems.
Uno de los formatos principales es la matriz de captura, en el que se usan reactivos de unión a ligandos, que normalmente son anticuerpos pero que también pueden ser alternativamente esqueletos proteicos, péptidos o aptámeros de ácido nucleico, para detectar moléculas diana en mezclas tal como plasma o extractos de tejido. En diagnóstico, se usan las matrices de captura para llevar a cabo múltiples inmunoensayos en paralelo, probando tanto para varios analitos en sueros individuales por ejemplo como probando muchas muestras de suero de forma simultánea. En proteómica, las matrices de captura se usan para cuantificar y comparar los niveles de proteínas en diferentes muestras en salud y enfermedad, es decir perfil de expresión de proteínas. Se usan proteínas diferentes de las que unen los ligandos específicos en el formato de la matriz para cribados funcionales in vitro tal como proteína-proteína, proteína-ADN, proteína-fármaco, receptor-ligando, enzima- sustrato, etc. Los reactivos de captura mismos se seleccionan y criban contra muchas proteínas, opcionalmente en formato de matriz multiplex contra muchas proteínas diana.One of the main formats is the capture matrix, in which ligand binding reagents are used, which are normally antibodies but which can also alternatively be protein skeletons, peptides or nucleic acid aptamers, to detect target molecules in mixtures such as plasma or tissue extracts. In diagnosis, capture matrices are used to perform multiple immunoassays in parallel, testing both for several analytes in individual sera for example and testing many serum samples simultaneously. In proteomics, the Capture matrices are used to quantify and compare protein levels in different samples in health and disease, that is, protein expression profile. Different proteins are used than those that bind specific ligands in the matrix format for in vitro functional screening such as protein-protein, protein-DNA, protein-drug, receptor-ligand, enzyme-substrate, etc. The capture reagents themselves are selected and screened against many proteins, optionally in multiplex matrix format against many target proteins.
Para la construcción de matrices, las fuentes de proteínas incluyen sistemas de expresión basados en células para proteínas recombinantes, purificación de fuentes naturales, producción in vitro mediante sistemas de traducción libres de células, y métodos sintéticos para péptidos. Muchos de estos métodos están automatizados para producción de alto rendimiento. Para las matrices de captura y análisis de la función de proteínas, es importante que las proteínas estén correctamente plegadas y sean funcionales; este no es siempre el caso, por ejemplo, donde se extraen las proteínas recombinantes de bacterias en condiciones desnaturalizantes. Sin embargo, las matrices de proteínas desnaturalizadas son útiles en el cribado de anticuerpos para reactividad cruzada, identificación de autoanticuerpos y selección de proteínas que se unen a ligandos.For matrix construction, protein sources include cell-based expression systems for recombinant proteins, purification of natural sources, in vitro production by cell-free translation systems, and synthetic methods for peptides. Many of these methods are automated for high performance production. For capture matrices and protein function analysis, it is important that the proteins are correctly folded and functional; This is not always the case, for example, where recombinant bacteria proteins are extracted under denaturing conditions. However, denatured protein matrices are useful in the screening of antibodies for cross-reactivity, identification of autoantibodies and selection of ligand-binding proteins.
Se han diseñado matrices de proteínas como una miniaturización de métodos de inmunoensayo familiares tal como ELISA y transferencia en gota, utilizando con frecuencia lectores fluorescentes, y facilitada por robótica y sistemas de detección de alto rendimiento para permitir que se lleven a cabo múltiples ensayos en paralelo. Los soportes físicos incluyen portaobjetos de vidrio, silicona, micropocillos, membranas de nitrocelulosa o PVDF, y microbolas magnéticas y otras. Mientras que el formato más familiar son microgotas de proteína de que dejan sobre superficies planares, las arquitecturas alternativas incluyen dispositivos de centrifugación de CD basados en desarrollo en microfluidos (Gyros, Monmouth Junction, NJ) y diseños especializados de chip, tal como los microcanales construidos en una placa (por ejemplo, The Living Chip™, Biotrove, Woburn, MA) y postes 3D minúsculos sobre una superficie de silicona (Zyomyx, Hayward CA). También se usan partículas en suspensión como base para las matrices, siempre que estén codificadas para identificación; los sistemas incluyen códigos de colores para microbolas (Luminex, Austin, TX; Bio-Rad Laboratories), nanocristales semiconductores (por ejemplo, QDOTS™, Quantum Dot, Hayward, CA), código de barras para bolas (bolas ULTRAPLEX™, SmartBead Technologies Ltd, Babraham, Cambridge, RU) y microbastones multimetálicos (por ejemplo, partículas NANOBARCODES™, Nanoplex Technologies, Mountain View, CA). Las bolas se ensamblan opcionalmente en matrices planares en chips semiconductores (tecnología LEAPS™, BioArray Solutions, Warren, NJ).Protein matrices have been designed as a miniaturization of familiar immunoassay methods such as ELISA and drop transfer, often using fluorescent readers, and facilitated by robotics and high performance detection systems to allow multiple tests to be carried out in parallel. . Physical supports include glass slides, silicone, microwells, nitrocellulose or PVDF membranes, and magnetic microbeads and others. While the most familiar format is protein droplets that leave on planar surfaces, alternative architectures include microfluidic-based CD centrifugation devices (Gyros, Monmouth Junction, NJ) and specialized chip designs, such as built-in microchannels on a plate (for example, The Living Chip ™, Biotrove, Woburn, MA) and tiny 3D posts on a silicone surface (Zyomyx, Hayward CA). Suspended particles are also used as a base for matrices, provided they are coded for identification; The systems include color codes for microballs (Luminex, Austin, TX; Bio-Rad Laboratories), semiconductor nanocrystals (e.g., QDOTS ™, Quantum Dot, Hayward, CA), ball barcode (ULTRAPLEX ™ balls, SmartBead Technologies Ltd, Babraham, Cambridge, RU) and multimetallic microbeads (for example, NANOBARCODES ™ particles, Nanoplex Technologies, Mountain View, CA). The balls are optionally assembled in planar matrices on semiconductor chips (LEAPS ™ technology, BioArray Solutions, Warren, NJ).
La inmovilización de proteínas implica tanto el reactivo de acoplamiento como la naturaleza de la superficie a la que se acopla. Una superficie soporte de matriz de proteínas buena es químicamente estable antes y después de los procedimientos de acoplamiento, permite buena morfología de las manchas, muestra unión no específica mínima, no contribuye al fondo en sistemas de detección, y es compatible con diferentes sistemas de detección. El método de inmovilización usado es reproducible, aplicable a proteínas de diferentes propiedades (tamaño, hidrofílica, hidrofóbica), manejable a alto rendimiento y automatización, y compatible con la retención de actividad de proteína completamente funcional. La orientación de la proteína unida a la superficie se reconoce como un factor un importante al presentarla al ligando o sustrato en un estado activo; para las matrices de captura los resultados de unión más eficaces se obtienen con reactivos de captura orientados, que generalmente requieren mareaje específico de sitio de la proteína. Se usan tanto métodos covalentes como no covalentes de inmovilización de proteínas y tienen varios pros y contras. La adsorción pasiva a superficies es metodológicamente sencilla, pero permite poco control cuantitativo o de orientación. Puede alterar o no las propiedades funcionales de la proteína, y la reproducibilidad y eficacia son variables. Los métodos de acoplamiento covalente proporcionan una unión estable, se aplican a un rango de proteínas y tienen buena reproducibilidad. Sin embargo, la orientación es variable. Además, la derivación química puede alterar la función de la proteína y requiere una superficie interactiva estable. Los métodos de captura biológicos que utilizan una etiqueta en la proteína proporcionan una unión estable y se unen a la proteína específicamente y en orientación reproducible, pero los reactivos biológicos se deben inmovilizar primero de forma adecuada, y la matriz puede requerir manejo especial y tener estabilidad variable. Se han descrito varias químicas de inmovilización y etiquetas para la fabricación de matrices de proteína. Los sustratos para la unión covalente incluyen portaobjetos de vidrio recubiertos con reactivos de silanos que contienen amino o aldehido. En el sistema VERSALINX™ (Prolinx, Bothell, WA) se alcanza acoplamiento covalente reversible mediante interacción entre la proteína derivada con ácido fenildiborónico, y ácido salicilhidroxámico inmovilizado en la superficie del soporte. Ésto también tiene unión de fondo baja y fluorescencia intrínseca baja y permite que las proteínas inmovilizadas retengan la función. La unión no covalente de proteínas no modificadas sucede con estructuras porosas tal como HYDROGEL™ (PerkinElmer, Wellesley, MA), basado en un gel tridimensional de poliacrilamida; se ha descrito que este sustrato da un fondo particularmente bajo en micromatrices de vidrio, y gran capacidad y retención de función de las proteínas. Los métodos de acoplamiento biológico ampliamente usados son por medio de interacciones biotina/estreptavidina o hexahistidina/Ni, habiendo modificado la proteína de forma adecuada. La biotina se puede conjugar a un esqueleto de polilisina inmovilizado sobre una superficie como dióxido de titanio (Zyomyx, Inc., Hayward, CA) o pentóxido de tantalio (Zeptosens, Witterswil, Suiza). Los métodos de fabricación de matrices incluyen impresión de contacto robótica, inyección de tinta, punteo piezoeléctrico y fotolitografía. Están disponibles un número de fabricantes de matrices comerciales [por ejemplo, Packard Biosciences, Affymetrix Inc. y Genetix] así como equipo manual [por ejemplo, V and P Scientific]. Las colonias de bacterias opcionalmente se ponen en rejillas robóticamente sobre membranas de PVDF para inducción de la expresión de la proteína in situ. En el límite del tamaño de mancha y densidad están las nanomatrices, con manchas en la escala espacial de nanómetros, que permiten que se realicen miles de reacciones en un solo chip de menos de 1 mm cuadrado. BioForce Nanosciences Inc. y Nanolink Inc., por ejemplo, han desarrollado nanomatrices comercialmente disponibles.Protein immobilization involves both the coupling reagent and the nature of the surface to which it is attached. A good protein matrix support surface is chemically stable before and after coupling procedures, allows good spot morphology, shows minimal non-specific binding, does not contribute to the background in detection systems, and is compatible with different detection systems . The immobilization method used is reproducible, applicable to proteins of different properties (size, hydrophilic, hydrophobic), manageable at high performance and automation, and compatible with the retention of fully functional protein activity. The orientation of the surface bound protein is recognized as an important factor when presenting it to the ligand or substrate in an active state; for capture matrices the most effective binding results are obtained with oriented capture reagents, which generally require site specific protein marking. Both covalent and non-covalent methods of protein immobilization are used and have several pros and cons. Passive adsorption to surfaces is methodologically simple, but allows little quantitative or orientation control. It may or may not alter the functional properties of the protein, and the reproducibility and efficacy are variable. Covalent coupling methods provide stable binding, apply to a range of proteins and have good reproducibility. However, the orientation is variable. In addition, chemical shunt can alter protein function and requires a stable interactive surface. Biological capture methods that use a tag on the protein provide stable binding and bind to the protein specifically and in reproducible orientation, but the biological reagents must first be properly immobilized, and the matrix can require special handling and have variable stability. Several immobilization chemicals and labels for the manufacture of protein matrices have been described. Substrates for covalent bonding include glass slides coated with silane reagents containing amino or aldehyde. In the VERSALINX ™ system (Prolinx, Bothell, WA), reversible covalent coupling is achieved by interaction between the protein derived with phenyldiboronic acid and salicylic hydroxamic acid immobilized on the support surface. This also has low background binding and low intrinsic fluorescence and allows immobilized proteins to retain function. Non-covalent binding of unmodified proteins occurs with porous structures such as HYDROGEL ™ (PerkinElmer, Wellesley, MA), based on a three-dimensional polyacrylamide gel; It has been described that this substrate gives a particularly low background in glass microarrays, and high capacity and retention of protein function. The widely used biological coupling methods are through biotin / streptavidin or hexahistidine / Ni interactions, having modified the protein appropriately. Biotin can be conjugated to a polylysine skeleton immobilized on a surface such as titanium dioxide (Zyomyx, Inc., Hayward, CA) or tantalum pentoxide (Zeptosens, Witterswil, Switzerland). Matrix manufacturing methods include robotic contact printing, inkjet, piezoelectric tapping and photolithography. A number of commercial matrix manufacturers are available [for example, Packard Biosciences, Affymetrix Inc. and Genetix] as well as manual equipment [for example, V and P Scientific]. Bacteria colonies are optionally placed in robotic grids on PVDF membranes for induction of protein expression in situ. In the limit of the spot size and density are the nanomatrices, with spots on the nanometer space scale, which allow thousands of reactions to be carried out on a single chip of less than 1 mm square. BioForce Nanosciences Inc. and Nanolink Inc., for example, have developed commercially available nanomatrices.
Los métodos de mareaje y detección con fluorescencia se usan ampliamente. Los mismos instrumentos que se usan para la lectura de micromatrices de ADN son aplicables para matrices de proteínas. Para la presentación diferencial, las matrices de captura (por ejemplo, anticuerpos) se prueban con proteínas marcadas fluorescentemente de dos estados celulares diferentes, en las que los usados celulares se conjugan directamente con fluoróforos diferentes (por ejemplo, Cy-3, Cy-5) y se mezclan, de modo que el color actúa como un lector para cambios en la abundancia de la diana. La sensibilidad de lectura fluorescente se amplifica 10-100 veces mediante amplificación de señal de tiramida (TSA) (PerkinElmer Lifesciences). La tecnología planar de ondas guiadas (Zeptosens) permite detección fluorescente ultrasensible, con la ventaja adicional de no tener procedimientos de lavado intermedios. La alta sensibilidad se alcanza con la suspensión de bolas y partículas, usando ficoeritrina como marca (Luminex) o las propiedades de nanocristales semiconductores (Quantum Dot). Se han desarrollado un número de lectores alternativos, especialmente en el área de la biotecnología comercial. Estos incluyen adaptaciones de resonancia de plasmón nuclear (HTS Biosystems, Intrinsic Bioprobes, Tempe, AZ), amplificación de ADN en círculo rodante (Molecular Staging, New Haven, CT), espectrometría de masas (Intrinsic Bioprobes; Ciphergen, Fremont, CA), dispersión de luz por resonancia (Genicon Sciences, San Diego, CA) y microscopía de fuerza atómica [BioForce Laboratories].The methods of marking and fluorescence detection are widely used. The same instruments that are used for reading DNA microarrays are applicable for protein matrices. For differential presentation, capture matrices (for example, antibodies) are tested with fluorescently labeled proteins from two different cell states, in which the used cell phones are they conjugate directly with different fluorophores (for example, Cy-3, Cy-5) and mix, so that the color acts as a reader for changes in the abundance of the target. The fluorescent reading sensitivity is amplified 10-100 times by tyramide signal amplification (TSA) (PerkinElmer Lifesciences). Planar guided wave technology (Zeptosens) allows ultrasensitive fluorescent detection, with the additional advantage of not having intermediate washing procedures. High sensitivity is achieved with the suspension of balls and particles, using phycoerythrin as a brand (Luminex) or the properties of semiconductor nanocrystals (Quantum Dot). A number of alternative readers have been developed, especially in the area of commercial biotechnology. These include nuclear plasmon resonance adaptations (HTS Biosystems, Intrinsic Bioprobes, Tempe, AZ), rolling circle DNA amplification (Molecular Staging, New Haven, CT), mass spectrometry (Intrinsic Bioprobes; Ciphergen, Fremont, CA), resonance light scattering (Genicon Sciences, San Diego, CA) and atomic force microscopy [BioForce Laboratories].
Las matrices de captura forman la base de los chips de diagnóstico para el perfil de expresión. Emplean reactivos de captura de alta afinidad, tal como anticuerpos convencionales, dominios individuales, esqueletos construidos, péptidos o aptámeros de ácidos nucleicos, para unirse y detectar ligandos dianas específicos en una manera de alto rendimiento.Capture matrices form the basis of diagnostic chips for the expression profile. They employ high affinity capture reagents, such as conventional antibodies, individual domains, constructed skeletons, peptides or nucleic acid aptamers, to bind and detect specific target ligands in a high yield manner.
Las matrices de anticuerpos tienen las propiedades requeridas de especificidad y fondo aceptable, y algunas están comercialmente disponibles (BD Biosciences, San José, CA; Clontech, Mountain View, CA; BioRad; Sigma, St. Louis, MO). Los anticuerpos para las matrices de captura se producen bien mediante inmunización convencional (sueros policlonales e hibridomas), o como fragmentos recombinantes, normalmente expresados en E. coli, después de la selección de bibliotecas de presentación en fagos o ribosomas (Cambridge Antibody Technology, Cambridge, RU; Biolnvent, Lund, Suecia; Affitech, Walnut Creek, CA; Biosite, San Diego, CA). Además de los anticuerpos convencionales, los fragmentos Fab y scFv, dominios V individuales de camélidos o equivalentes humanos construidos (Domantis, Waltham, MA) son opcionalmente útiles en matrices. El término esqueleto se refiere a dominios de proteína de unión a ligando, que se construyen en múltiples variantes capaces de unirse a diversas moléculas diana con propiedades de especificidad y afinidad similares a anticuerpo. Las variantes se producen en formato de biblioteca genética y se seleccionan contra dianas individuales mediante presentación en fago, bacteria o ribosoma. Tales esqueletos o marcos que se unen a ligando incluyen Affybodies basados en proteína A de S. aureus (Affibody, Bromma, Suecia), Trinectins basado en fibronectinas (Phylos, Lexington, MA) y Anticalins basado en la estructura de la lipocalina (Pieris Proteolab, Freising- Weihenstephan, Alemania). Éstos se usan en matrices de captura de una manera similar a los anticuerpos y tienen las ventajas de la consistencia y facilidad de producción.Antibody matrices have the required specificity and acceptable background properties, and some are commercially available (BD Biosciences, San Jose, CA; Clontech, Mountain View, CA; BioRad; Sigma, St. Louis, MO). Antibodies to capture matrices are produced either by conventional immunization (polyclonal sera and hybridomas), or as recombinant fragments, normally expressed in E. coli, after selection of phage or ribosome display libraries (Cambridge Antibody Technology, Cambridge , RU; Biolnvent, Lund, Sweden; Affitech, Walnut Creek, CA; Biosite, San Diego, CA). In addition to conventional antibodies, Fab and scFv fragments, individual V domains of camelids or constructed human equivalents (Domantis, Waltham, MA) are optionally useful in matrices. The term "skeleton" refers to ligand-binding protein domains, which are constructed in multiple variants capable of binding to various target molecules with specificity and affinity properties similar to antibody. Variants are produced in a genetic library format and selected against individual targets by phage, bacteria or ribosome presentation. Such skeletons or ligand-binding frameworks include Affybodies based on S. aureus protein A (Affibody, Bromma, Sweden), Trronectins based on fibronectins (Phylos, Lexington, MA) and Anticalins based on lipocalin structure (Pieris Proteolab , Freising- Weihenstephan, Germany). These are used in capture matrices in a manner similar to antibodies and have the advantages of consistency and ease of production.
Las moléculas de captura no proteicas, notablemente los aptámeros de ácido nucleico de cadena sencilla que se unen a ligandos proteicos con alta especificidad y afinidad, también se usan en matrices (SomaLogic, Boulder, CO). Los aptámeros se seleccionan de bibliotecas de oligonucleótidos mediante el procedimiento Selex™ (SomaLogic, Boulder, CO) y su interacción con proteínas aumenta mediante unión covalente, por medio de la incorporación de desoxiuridina brominada y entrecruzamiento activado por UV (fotoaptámeros). El fotoentrecruzamiento a ligandos reduce la reactividad cruzada de los aptámeros debido a los requerimientos estéricos específicos. Los aptámeros tienen las ventajas de facilidad de producción mediante síntesis automatizada de oligonucleótidos y la estabilidad y consistencia del ADN; en las matrices de fotoaptámeros, se usan colorantes de proteínas fluorescentes universales para detectar la unión.Non-protein capture molecules, notably single chain nucleic acid aptamers that bind protein ligands with high specificity and affinity, are also used in matrices (SomaLogic, Boulder, CO). The aptamers are selected from oligonucleotide libraries by the Selex ™ method (SomaLogic, Boulder, CO) and their interaction with proteins is increased by covalent binding, by the incorporation of brominated deoxyuridine and UV-activated cross-linking (photoaptamers). Photo-cross-linking to ligands reduces cross-reactivity of aptamers due to specific steric requirements. Aptamers have the advantages of ease of production by automated oligonucleotide synthesis and DNA stability and consistency; in the photoaptamer matrices, universal fluorescent protein dyes are used to detect binding.
Los analitos proteicos que se unen a matrices de anticuerpos se detectan de forma directa o indirecta, por ejemplo, a través de un anticuerpo secundario. Se usa el mareaje directo para la comparación de diferentes muestras con diferentes colores. Donde están disponibles pares de anticuerpos dirigidos al mismo ligando proteico, los inmunoensayos en sandwich proporcionan alta especificidad y sensibilidad y por lo tanto son el método de elección para proteínas de baja abundancia tal como citoquinas; también dan la posibilidad de detección de modificaciones de proteína. Los métodos de detección sin marca, incluyendo espectrometría de masas, resonancia de plasmón de superficie y microscopía de fuerza atómica, evitan la alteración del ligando. Lo que se requiere de cualquier método es sensibilidad y especificidad óptimas, con fondo bajo para dar una señal a ruido alta. Puesto que las concentraciones de analito cubren un amplio rango, la sensibilidad tiene que adaptarse apropiadamente. La dilución en serie de la muestra o el uso de anticuerpos de diferentes afinidades son soluciones para este problema. Las proteínas de interés con frecuencia son aquellas en concentraciones bajas en los líquidos corporales y extractos, que requieren detección en el intervalo de pg o menor, tal como citoquinas o los productos de baja expresión en células. Una alternativa a una matriz de moléculas de captura es una hecha por medio de la tecnología de huella molecular, en la cual se usan péptidos (por ejemplo, de las regiones C-terminales de las proteínas) como moldes para generar cavidades estructuralmente complementarias, específicas de secuencia en una matriz polimerizable; las cavidades pueden después capturar específicamente proteínas (desnaturalizadas) que tienen la secuencia primaria de aminoácidos apropiada (ProteinPrint™, Aspira Biosystems, Burlingame, CA).Protein analytes that bind to antibody matrices are detected directly or indirectly, for example, through a secondary antibody. Direct marking is used to compare different samples with different colors. Where pairs of antibodies directed to the same protein ligand are available, sandwich immunoassays provide high specificity and sensitivity and are therefore the method of choice for low abundance proteins such as cytokines; They also give the possibility of detection of protein modifications. The methods of Unmarked detection, including mass spectrometry, surface plasmon resonance and atomic force microscopy, prevents ligand alteration. What is required of any method is optimal sensitivity and specificity, with a low background to give a signal at high noise. Since analyte concentrations cover a wide range, sensitivity has to be adapted appropriately. Serial dilution of the sample or the use of antibodies of different affinities are solutions to this problem. The proteins of interest are often those at low concentrations in body fluids and extracts, which require detection in the range of pg or less, such as cytokines or low-expression products in cells. An alternative to a matrix of capture molecules is one made by means of molecular fingerprint technology, in which peptides (for example, from C-terminal regions of proteins) are used as templates to generate structurally complementary, specific cavities sequence in a polymerizable matrix; the cavities can then specifically capture (denatured) proteins that have the appropriate primary amino acid sequence (ProteinPrint ™, Aspira Biosystems, Burlingame, CA).
Otra metodología que es útil diagnósticamente y en el perfil de expresión es la matriz ProteinChip® (Ciphergen, Fremont, CA), en la que superficies cromatográficas en fase sólida se unen a proteínas con características similares de carga o hidrofobicidad de mezclas tales como plasma o extractos de tumores, y se usa espectrometría de masas SELDI-TOF para la detección de las proteínas retenidas.Another methodology that is useful diagnostically and in the expression profile is the ProteinChip® matrix (Ciphergen, Fremont, CA), in which solid-phase chromatographic surfaces bind to proteins with similar loading or hydrophobicity characteristics of mixtures such as plasma or tumor extracts, and SELDI-TOF mass spectrometry is used for the detection of retained proteins.
Se han construido chips funcionales a gran escala inmovilizando grandes números de proteínas purificadas y se usan para ensayar un amplio intervalo de funciones biológicas, tal como interacciones de proteínas con otras proteínas, interacciones fármaco-diana, enzima-sustrato, etc. En general requieren una biblioteca de expresión, clonada en E. coli, levadura o similar a partir de la cual se purifican las proteínas expresadas, por ejemplo, a través de una etiqueta de His y se inmovilizan. La transcripción/traducción de proteínas libre de células es una alternativa viable para la síntesis de proteínas que no se expresan bien en sistemas bacterianos u otros sistemas in vivo. Para detectar interacciones proteína-proteína, las matrices de proteína son alternativas in vitro al sistema de los dos híbridos de levadura basado en célula y son útiles donde el último es deficiente, tal como interacciones que implican proteínas secretadas o proteínas con puentes disulfúro. Se ha descrito el análisis de alto rendimiento de actividades bioquímicas en matrices para proteínas quinasas de levadura y para varias funciones (interacciones proteína-proteína y proteína-lípido) del proteoma de levadura, donde se expresó una gran proporción de todos los marcos abiertos de lectura de levadura y se inmovilizó en una micromatriz. Los chips de proteoma a gran escala también son útiles en la identificación de interacciones funcionales, cribado de fármacos, etc. (Proteometrix, Branford, CT).Large-scale functional chips have been constructed by immobilizing large numbers of purified proteins and are used to test a wide range of biological functions, such as protein interactions with other proteins, drug-target, enzyme-substrate interactions, etc. They generally require an expression library, cloned in E. coli, yeast or the like from which the expressed proteins are purified, for example, through a His tag and immobilized. Transcription / translation of cell-free proteins is a viable alternative for the synthesis of proteins that do not express well in bacterial systems or other systems in vivo. To detect protein-protein interactions, protein matrices are alternatives in vitro to the system of the two cell-based yeast hybrids and are useful where the latter is deficient, such as interactions involving secreted proteins or proteins with disulfuric bonds. The high-performance analysis of biochemical activities in matrices for yeast protein kinases and for various functions (protein-protein and protein-lipid interactions) of yeast proteome has been described, where a large proportion of all open reading frames was expressed of yeast and immobilized in a microarray. Large-scale proteome chips are also useful in identifying functional interactions, drug screening, etc. (Proteometrix, Branford, CT).
Como una presentación bidimensional de elementos individuales, se usa una matriz de proteínas para cribar bibliotecas de presentación en fagos o ribosomas, para seleccionar compañeros de unión, incluyendo anticuerpos, esqueletos sintéticos, péptidos y aptámeros. De esta manera, se lleva a cabo un cribado de biblioteca contra biblioteca. El cribado de fármacos candidatos en bibliotecas químicas combinatorias contra una matriz de proteínas diana identificadas de proyectos genómicos es otra aplicación de esta aproximación.As a two-dimensional presentation of individual elements, a protein matrix is used to screen phage display libraries or ribosomes, to select binding partners, including antibodies, synthetic skeletons, peptides and aptamers. In this way, a library vs. library screening is carried out. The screening of candidate drugs in combinatorial chemical libraries against a matrix of target proteins identified from genomic projects is another application of this approach.
Los ensayos multiplexados con bolas usan una serie de partículas espectralmente discretas que se usan para capturar y cuantificar analitos solubles. El analito se mide después mediante detección de una emisión basada en fluorescencia y análisis por citometría de flujo. Los ensayos multiplexados con bolas generan datos que son comparables a ensayos basados en ELISA, pero en una forma multiplexada o simultánea. Se calcula la concentración de desconocidos para la matriz citométrica de bolas como con cualquier ensayo en formato sandwich, es decir, por medio del uso de estándares conocidos y representando los desconocidos contra la curva patrón. Además, los ensayos multiplexados con bolas permiten la cuantificación de analitos solubles en muestras nunca antes consideradas debido a las limitaciones de volumen de la muestra. Además de los datos cuantitativos, se generan imágenes visuales poderosas que revelan perfiles únicos o firmas que proporcionan al usuario información adicional de un vistazo. El kit de la invención puede contener adicionalmente instrucciones para el uso en determinar los niveles de proteína de los biomarcadores presentes en la muestra. Dichas instrucciones se pueden encontrar en forma de material impreso o en forma de soporte electrónico que puede almacenar instrucciones de modo que puedan ser leídas por un sujeto, tales como medios electrónicos de almacenamiento (discos ópticos, cintas y similares), medios ópticos (CD-ROM, DVD) y similares. Los medios pueden contener de forma adicional o alternativa páginas de Internet que proporcionan dichas instrucciones.Multiplexed ball assays use a series of spectrally discrete particles that are used to capture and quantify soluble analytes. The analyte is then measured by detection of a fluorescence based emission and flow cytometric analysis. Multiplexed ball assays generate data that is comparable to ELISA based assays, but in a multiplexed or simultaneous manner. The concentration of strangers for the cytometric ball matrix is calculated as with any sandwich test, that is, through the use of known standards and representing the unknown ones against the standard curve. In addition, multiplexed ball assays allow quantification of soluble analytes in samples never before considered due to sample volume limitations. In addition to quantitative data, powerful visual images are generated that reveal unique profiles or signatures that provide the user with additional information at a glance. The kit of the invention may additionally contain instructions for use in determining the protein levels of the biomarkers present in the sample. These instructions can be found in the form of printed material or in the form of electronic support that can store instructions so that they can be read by a subject, such as electronic storage media (optical discs, tapes and the like), optical media (CD- ROM, DVD) and the like. The media may additionally or alternatively contain Internet pages that provide such instructions.
La invención se describe aquí en más detalle como los siguientes ejemplos que se deben considerar como meramente ilustrativos y no limitantes del ámbito de la invención.The invention is described here in more detail as the following examples that should be considered as merely illustrative and not limiting the scope of the invention.
EJEMPLOSEXAMPLES
MATERIALES Y MÉTODOSMATERIALS AND METHODS
MUESTRASSAMPLES
Se obtuvieron un total de 80 muestras de orina de la Clínica Universitaria de Navarra. La recogida de muestras fue aprobada por el Comité de Ética para Ensayos Clínicos local de la Clínica Universitaria de Navarra, y todos los pacientes dieron consentimiento informado para participar. Las muestras de orina eran de 35 pacientes con cirrosis F4 (índice de Metavir), 10 pacientes con fibrosis Fl (índice de Metavir), y 35 controles sanos. El criterio usado para clasificar el daño en el hígado fue la biopsia hepática para los pacientes con cirrosis y fibrosis, y el análisis de sangre estándar para los individuos control. El 81,5% de los pacientes eran hombres y el 18,5% mujeres y todos tenían entre 38 y 75 años de edad.A total of 80 urine samples were obtained from the University Clinic of Navarra. Sample collection was approved by the local Ethics Committee for Clinical Trials of the University Clinic of Navarra, and all patients gave informed consent to participate. Urine samples were from 35 patients with F4 cirrhosis (Metavir index), 10 patients with Fl fibrosis (Metavir index), and 35 healthy controls. The criteria used to classify liver damage was liver biopsy for patients with cirrhosis and fibrosis, and standard blood tests for control individuals. 81.5% of the patients were men and 18.5% women and all were between 38 and 75 years old.
DIGESay
Las muestras de orina se centrifugaron a 4000xg durante 5 minutos a 40C, y los sobrenadantes de 30 mi se concentraron usando dispositivos de filtros de centrifugaciónUrine samples were centrifuged at 4000xg for 5 minutes at 4 0 C, and 30 ml supernatants were concentrated using centrifugal filter devices
Amicon Ultra-15 (5000 NMWL) de Millipore. Las proteínas concentradas se precipitaron con el kit ReadyPrep 2-D Cleanup (Bio-Rad) y los precipitados resultantes se resuspendieron en urea 7 M, tiourea 2 M, CHAPS al 4%, Tris/HCl 30 mM pH 8. La concentración de proteína se determinó usando el kit del ensayo de Bradford (BioRad), usando seroalbúmina bovina como proteína estándar. Se marcaron las muestras de proteína (50 μg) con 400 pmoles de colorantes fluorescentes Cy3 y Cy5 en N ,N- dimetilformamida. Se preparó una muestra de referencia mezclando 25 μg de proteína de cada condición experimental y el conjunto resultante se marcó posteriormente con Cy2. Se realizaron análisis en triplicado alternando el mareaje fluorescente de las muestras del estudio. La primera dimensión se realizó en un Ettan IPGphor (GE Healthcare) usando Immobiline DryStrips pH3-l l NL, 24 cm (GE Healthcare). Las muestras se cargaron en el extremo ácido de la tira con un dispositivo de carga de copa. Las condiciones de isoelectroenfoque fueron las recomendadas por el fabricante para el tipo de tira usada. El SDS-PAGE (12,5%) se desarrolló en un Ettan DALTsix (GE Healthcare) a 250C y lW/gel durante 12 horas. Las imágenes de los geles se capturaron con un Typhoon Trio (GE Healthcare) a una resolución de 100 μm con λex/λem de 488/520, 532/580, y 633/670 nm para Cy2, Cy3, and Cy5 respectivamente. El análisis de imágenes y estadística se realizaron con el software DeCyder Differential versión 6.5 (GE Healthcare) usando el módulo Biological Variation Analysis (BVA). Sólo se aceptaron diferencias con p<0,05 (prueba t).Amicon Ultra-15 (5000 NMWL) from Millipore. The concentrated proteins were precipitated with the ReadyPrep 2-D Cleanup (Bio-Rad) kit and the resulting precipitates were resuspended in 7 M urea, 2 M thiourea, 4% CHAPS, 30 mM Tris / HCl pH 8. Protein concentration was determined using the Bradford assay kit (BioRad), using bovine serum albumin as a standard protein. Protein samples (50 μg) were labeled with 400 pmoles of Cy3 and Cy5 fluorescent dyes in N, N-dimethylformamide. A reference sample was prepared by mixing 25 μg of protein from each experimental condition and the resulting set was subsequently labeled with Cy2. Triplicate analyzes were performed alternating fluorescent tide of study samples. The first dimension was performed on an Ettan IPGphor (GE Healthcare) using Immobiline DryStrips pH3-l l NL, 24 cm (GE Healthcare). The samples were loaded at the acid end of the strip with a cup loading device. The isoelectric focusing conditions were those recommended by the manufacturer for the type of strip used. The SDS-PAGE (12.5%) was developed in an Ettan DALTsix (GE Healthcare) at 25 0 C and lW / gel for 12 hours. The images of the gels were captured with a Typhoon Trio (GE Healthcare) at a resolution of 100 μm with λex / λem of 488/520, 532/580, and 633/670 nm for Cy2, Cy3, and Cy5 respectively. Image analysis and statistics were performed with DeCyder Differential version 6.5 software (GE Healthcare) using the Biological Variation Analysis (BVA) module. Only differences with p <0.05 (t test) were accepted.
IDENTIFICACIÓN DE MARCADORES POR DIGEIDENTIFICATION OF MARKERS BY SAY
Los geles preparativos se corrieron con 400 μg de proteína siguiendo el mismo procedimiento indicado anteriormente. Las proteínas se visualizaron mediante tinción con tinción de gel de proteínas SYPRO Ruby (Bio-Rad) y las imágenes se adquirieron con un Typhoon Trio usando λex/λem 532/560 nm. Las manchas diferencialmente representadas se cortaron de forma manual y las muestras de gel se procesaron con una estación MassPrep (Waters). La digestión tríptica en el gel se realizó con 12,5 ng/μl de tripsina en bicarbonato de amonio 50 mM durante 12 horas a 370C. Se realizó CL en fase reversa microcapilar con un sistema capilar CapLC™ (Waters). La separación en fase reversa de los digeridos trípticos se realizó con una columna capilar de sílice fusionada Atlantis, C 18, 3 μm, 75 μmx lO cm Nano Ease™ (Waters) equilibrada en acetonitrilo al 5%, ácido fórmico al 0,2%. Tras la inyección de 6 μl de muestra, la columna se lavó durante 5 minutos con el mismo tampón y los péptidos se eluyeron usando un gradiente lineal de acetonitrilo del 5-50% en 45 minutos a una velocidad de flujo constante de 0,2 μl/min. La columna se acopló en línea a un Q-TOF Micro (Waters) usando una fuente de ionización por nanospray PicoTip (Waters). La temperatura del capilar calentado fue de 8O0C y la diferencia de potencial del spray fue de 1,8-2,2 kV. Los datos de MS/MS se recogieron en un modo automatizado dependiente de los datos. Los tres iones más intensos en cada estudio se fraccionaron secuencialmente mediante disociación inducida por colisión (CID) usando una anchura de aislamiento de 2,0 y una energía relativa de colisión del 35%. El procesamiento de datos se realizó con MassLynx 4.0. La búsqueda en las bases de datos se hizo con Phenyx 2.2 (GeneBio, Ginebra, Suiza) contra la base de conocimiento Uniprot Reléase 12.3 que consiste en UniprotKB/Swiss-Prot Reléase 54.3 y UniprotKB/TrEMBL Reléase 37.3 con las entradas 285.335 y 4.932.421, respectivamente. La búsqueda se restringió enzimáticamente para tripsina y se permitió un sitio de corte fallado. Los parámetros de búsqueda adicionales fueron como sigue: sin restricción en el peso molecular y punto isoeléctrico; modificación fija, carbamidometilación de cisterna; modificación variable, oxidación de metionina.The preparative gels were run with 400 μg of protein following the same procedure indicated above. The proteins were visualized by staining with SYPRO Ruby protein gel staining (Bio-Rad) and the images were acquired with a Typhoon Trio using λex / λem 532/560 nm. Differentially represented spots were cut manually and gel samples were processed with a MassPrep (Waters) station. Triptych digestion in the gel was performed with 12.5 ng / µl trypsin in 50 mM ammonium bicarbonate for 12 hours at 37 ° C. Microcapillary reverse phase CL was performed with a CapLC ™ (Waters) capillary system. Reverse phase separation of the tryptic digested was performed with a fused silica capillary column Atlantis, C 18, 3 μm, 75 μm x 10 cm Nano Ease ™ (Waters) balanced in 5% acetonitrile, 0.2% formic acid . After injection of 6 μl of sample, the column was washed for 5 minutes with the same buffer and the peptides were eluted using a linear gradient of 5-50% acetonitrile in 45 minutes at a constant flow rate of 0.2 μl / min. The column was coupled in line to a Q-TOF Micro (Waters) using a PicoTip nanospray ionization source (Waters). The temperature of the heated capillary was 8O 0 C and the potential difference of the spray was 1.8-2.2 kV. MS / MS data was collected in an automated mode dependent on the data. The three most intense ions in each study were sequentially fractionated by collision-induced dissociation (CID) using an isolation width of 2.0 and a relative collision energy of 35%. Data processing was performed with MassLynx 4.0. The database search was done with Phenyx 2.2 (GeneBio, Geneva, Switzerland) against the Uniprot Relay 12.3 knowledge base consisting of UniprotKB / Swiss-Prot Relay 54.3 and UniprotKB / TrEMBL Relay 37.3 with entries 285.335 and 4.932. 421, respectively. The search was enzymatically restricted for trypsin and a failed cut site was allowed. Additional search parameters were as follows: no restriction on molecular weight and isoelectric point; fixed modification, cistern carbamidomethylation; variable modification, methionine oxidation.
SELDI TOFSELDI TOF
En el método SELDI, se aplican soluciones de proteína a las manchas de matrices de ProteinChip, que se han derivado con químicas cromatográficas planares. Las proteínas interaccionan activamente con la superficie de la matriz cromatográfica, y son secuestradas según su potencial de interacción de superficie así como separadas de sales y otras contaminantes de la muestra mediante lavado posterior en la mancha con las soluciones tampones adecuadas. Como en la técnica MALDI, las proteínas a ser analizadas se cocristalizan con componentes que absorben UV y se volatilizan mediante un haz de láser UV pulsado. Las proteínas ionizadas se aceleran después en un campo eléctrico, y las relaciones de la masa a la carga de las diferentes especies iónicas de las proteínas se pueden deducir a partir de su velocidad.In the SELDI method, protein solutions are applied to ProteinChip matrix stains, which have been derived with planar chromatographic chemicals. The proteins actively interact with the surface of the chromatographic matrix, and are sequestered according to their potential for surface interaction as well as separated from salts and other contaminants in the sample by subsequent washing in the stain with the appropriate buffer solutions. As in the MALDI technique, the proteins to be analyzed are co-crystallized with UV absorbing components and volatilized by a pulsed UV laser beam. The ionized proteins are then accelerated in an electric field, and the mass to charge ratios of the different ionic species of the proteins can be deduced from their velocity.
Las muestras se procesaron y analizaron en duplicado. Las muestras de orina se centrifugaron brevemente a 10000 g durante 5 minutos a 40C para eliminar restos y se diluyeron 2:3 en urea 9 M, CHAPS al 2%. Todas las muestras de orina desnaturalizadas se analizaron sin más fraccionamiento mediante dilución 1 :10 con los tampones de unión correspondientes antes de aplicarlas a la superficie del chip. Las proteínas se analizaron usando chips CMlO (intercambio catiónico), IMAC30 (afinidad a metal), QlO (intercambio aniónico) y H5 (interacción hidrofóbica) siguiendo las instrucciones del fabricante. Se usaron los siguientes tampones de unión para cada tipo específico de chip: tampón de CMlO poco fuerte (acetato de sodio 0,1 M, pH 4,0); tampón de H50 (la superficie se cargó inicialmente con ACN al 50% y la unión se realizo en ACN al 10%, TFA al 0,1%); tampón de QlO (Tris HCl 100 mM pH 9); tampón de IMAC30 (la superficie se cargó inicialmente con sulfato cúprico 0,1 M, se neutralizó con acetato de sodio 0,1 M pH 4,0, y la unión se realizó en fosfato de sodio 0,1 M, cloruro de sodio 0,5 M pH 7,0). El perfil de SELDI-TOF se realizó como sigue: los chips se equilibraron dos veces con 150 μl del tampón de unión apropiado durante 5 minutos y se cargaron dos veces (30 minutos cada carga) con 25 μl de muestra mezclada con 175 μl de tampón de unión. Las proteínas no unidas se eliminaron lavando los chips tres veces con 150 μl de tampón de unión y tres veces con 200 μl de agua desionizada. Después de secar la superficie cargada, se aplicó 1 μl de ácido sinapínico (Bio-Rad, Hercules, CA) disuelto en ACN al 50% y TFA al 0,5% dos veces y se dejó secar. El análisis de las proteínas/péptidos unidos se realizó usando ProteinChip System, Series 4000 (Bio-Rad, Hercules, CA). Los espectros se recogieron en modo de ion positivo entre el intervalo de masas de 1-100 kDa con una masa foco de 48 kDa y 8 kDa. Los espectros obtenidos se procesaron, normalizaron y compararon con el Ciphergen Express, un programa de análisis de grupos de datos, usando los parámetros por defecto. El análisis estadístico elegido fue la prueba univariante no paramétrica U-Mann-Whitney. Además, se usaron modelos lineales usando el paquete LIMMA de Bioconductor que permitió la selección de péptidos usando una corrección del valor de p basado en FDR o la estadística B (logaritmo del cociente probabilidad de que un péptido se exprese diferencialmente/probabilidad de que no sea diferencial). La capacidad discriminadora de los péptidos seleccionados se evaluó con la curva ROC (sensibilidad frente a especificidad) y particularmente con el valor AUC (área bajo la curva). Una vez establecidos los marcadores individuales más fiables, se realizó un análisis multivariante para evaluar la combinación que proporciona la clasificación más eficaz de las muestras. INMUNOTRANSFERENCIASamples were processed and analyzed in duplicate. Urine samples were briefly centrifuged at 10,000 g for 5 minutes at 4 0 C to remove debris and diluted 2: 3 in 9 M urea, 2% CHAPS. All denatured urine samples they were analyzed without further fractionation by dilution 1: 10 with the corresponding binding buffers before applying them to the chip surface. Proteins were analyzed using CMlO (cation exchange), IMAC30 (metal affinity), QlO (anion exchange) and H5 (hydrophobic interaction) chips following the manufacturer's instructions. The following binding buffers were used for each specific type of chip: shallow CMlO buffer (0.1 M sodium acetate, pH 4.0); H50 buffer (the surface was initially loaded with 50% ACN and binding was performed in 10% ACN, 0.1% TFA); QlO buffer (100 mM Tris HCl pH 9); IMAC30 buffer (the surface was initially loaded with 0.1 M cupric sulfate, neutralized with 0.1 M sodium acetate pH 4.0, and the binding was performed in 0.1 M sodium phosphate, 0 sodium chloride , 5 M pH 7.0). The SELDI-TOF profile was performed as follows: the chips were equilibrated twice with 150 μl of the appropriate binding buffer for 5 minutes and loaded twice (30 minutes each load) with 25 μl of sample mixed with 175 μl of buffer of Union. Unbound proteins were removed by washing the chips three times with 150 µl of binding buffer and three times with 200 µl of deionized water. After drying the charged surface, 1 µl of synapine acid (Bio-Rad, Hercules, CA) dissolved in 50% ACN and 0.5% TFA was applied twice and allowed to dry. The analysis of bound proteins / peptides was performed using ProteinChip System, Series 4000 (Bio-Rad, Hercules, CA). The spectra were collected in positive ion mode between the mass range of 1-100 kDa with a focus mass of 48 kDa and 8 kDa. The obtained spectra were processed, normalized and compared with the Ciphergen Express, a data group analysis program, using the default parameters. The statistical analysis chosen was the univariate non-parametric U-Mann-Whitney test. In addition, linear models were used using the LIMMA Bioconductor package that allowed the selection of peptides using a correction of the p-value based on FDR or the B statistic (logarithm of the quotient probability that a peptide is differentially expressed / probability of not being differential). The discriminant capacity of the selected peptides was evaluated with the ROC curve (sensitivity versus specificity) and particularly with the AUC value (area under the curve). Once the most reliable individual markers were established, a multivariate analysis was performed to evaluate the combination that provides the most effective classification of the samples. IMMUNOTRANSFERENCE
Las muestras de orina se mezclaron con tampón de carga 5x (Tris-HCl 250 mM pH 6,8, SDS al 10%, glicerol al 50%, mercaptoetanol al 5%, EDTA 62,5 mM, azul de bromo fenol al 0,1%) y se cargaron 1,2 μg de proteína y se corrieron en un gel de glicina-dodecilsulfato de sodio-poliacrilamida al 12,5%. Después de la electroforesis, las proteínas se transfirieron a una membrana de nitrocelulosa mediante transferencia húmeda (100 V a 40C durante 1 hora). Después de bloquear en leche al 5% en solución salina tamponada con fosfato más Tween (PBST), las membranas se incubaron con un anticuerpo policlonal de conejo anti-gelsolina (Santa Cruz Biotechnology, Santa Cruz, California), (diluido 1 :1000) y anticuerpo policlonal de conejo apolipoproteína D (Santa Cruz Biotechnology, Santa Cruz, California), en PBST que contenía leche al 1%, a 40C durante la noche. Después de lavar en PBST las bandas de proteínas inmunoreactivas se visualizaron incubando las membranas con anticuerpo anti-conejo conjugado a peroxidasa (diluido 1 :5000 en leche al 5% en PBST) durante 1 hora a temperatura ambiente. Usando un sistema de análisis ECL Western blotting, las bandas inmunoreactivas se representaron en una película quimioluminiscente mediante revelado de película normal.Urine samples were mixed with 5x loading buffer (250 mM Tris-HCl pH 6.8, 10% SDS, 50% glycerol, 5% mercaptoethanol, 62.5 mM EDTA, 0 bromine phenol blue, 1%) and 1.2 μg of protein were loaded and run on a 12.5% sodium glycine-dodecyl sulfate-polyacrylamide gel. After electrophoresis, the proteins were transferred to a nitrocellulose membrane by wet transfer (100 V at 4 0 C for 1 hour). After blocking in 5% milk in phosphate buffered saline plus Tween (PBST), the membranes were incubated with a polyclonal rabbit anti-gelsolin antibody (Santa Cruz Biotechnology, Santa Cruz, California), (diluted 1: 1000) and rabbit polyclonal antibody apolipoprotein D (Santa Cruz Biotechnology, Santa Cruz, California) in PBST containing 1% milk, 4 0 C overnight. After washing in PBST the immunoreactive protein bands were visualized by incubating the membranes with anti-rabbit antibody conjugated to peroxidase (diluted 1: 5000 in 5% milk in PBST) for 1 hour at room temperature. Using a Western blotting ECL analysis system, immunoreactive bands were represented on a chemiluminescent film by normal film development.
IDENTIFICACIÓN DE MARCADORES POR SELDI-TOFIDENTIFICATION OF MARKERS BY SELDI-TOF
Cromatografía de retenido (perfil de SELDI): Para ensayar las proteínas de interés durante la purificación, se hizo el perfil de las fracciones en matrices de ProteinChip CMlO y/o QlO. Las matrices se equilibraron primero en tampón de unión (Tris 50 mM, pH 9,0 para matrices QlO y acetato de Na 100 mM, pH 4,0 para matrices CMlO). Se diluyeron alícuotas de cada muestra 10 veces en tampón de unión y se dejaron unir a la superficie de la matriz durante 30 minutos. La matriz se lavó tres veces con 150 μl de tampón de unión, se aclaró con agua desionizada, y se dejó secar antes de la adición de matriz. Para evaluar contenido y pureza total de proteína, alícuotas de 1-2 μL también se unieron a matrices NP20 y se dejaron secar antes de añadir la matriz. Se usaron SPA o CHCA como matrices. Fraccionamiento de intercambio aniónico: Se desnaturalizaron 10 mL de orina con 16 mi de urea 9 M Tris 50 mM, pH 9,0. Se equilibró resina Q HyperD® F (PaIl Corporation) con tampón Ul (urea 1 M, CHAPS al 0,2%, Tris-HCl 50 mM, pH 9) y se incubó con cada muestra de orina. Las muestras de orina desnaturalizadas se incubaron por lotes durante 1 hora a temperatura ambiente con 800 μL de resina Q HyperD® F. Se recogió la fracción no unida y la resina se lavó con 1 mL de Tris 50 mM, pH 9 en OGP al 0.1%. Las proteínas unidas se eluyeron sucesivamente con 1 mL de tampones de pH 7, 6, 5, 4, y 3 en OGP al 0,1% y por último con una solución de isopropanol al 33%/ ACN al 17%/ TFA al 0,1%. Se hizo el perfil de una alícuota de cada fracción sobre matrices de ProteinChip CMlO y/o QlO para ensayar para las proteínas de interés.Retained chromatography (SELDI profile): To test the proteins of interest during purification, the fractions profile was made in ProteinChip CMlO and / or QlO matrices. The matrices were first equilibrated in binding buffer (50 mM Tris, pH 9.0 for QlO matrices and 100 mM Na acetate, pH 4.0 for CMlO matrices). Aliquots of each sample were diluted 10 times in binding buffer and allowed to bind to the surface of the matrix for 30 minutes. The matrix was washed three times with 150 µl of binding buffer, rinsed with deionized water, and allowed to dry before adding matrix. To assess total protein content and purity, 1-2 μL aliquots were also bound to NP20 matrices and allowed to dry before adding the matrix. SPA or CHCA were used as matrices. Anion exchange fractionation: 10 mL of urine were denatured with 16 ml of 50 mM 9M Tris urea, pH 9.0. Q HyperD® F resin (PaIl Corporation) was equilibrated with Ul buffer (1 M urea, 0.2% CHAPS, 50 mM Tris-HCl, pH 9) and incubated with each urine sample. Denatured urine samples were incubated in batches for 1 hour at room temperature with 800 μL of Q HyperD® F resin. The unbound fraction was collected and the resin was washed with 1 mL of 50 mM Tris, pH 9 in 0.1 OGP %. The bound proteins were eluted successively with 1 mL of buffers of pH 7, 6, 5, 4, and 3 in 0.1% OGP and finally with a solution of 33% isopropanol / 17% ACN / 0% TFA ,one%. The profile of an aliquot of each fraction was made on ProteinChip CMlO and / or QlO matrices to be tested for the proteins of interest.
Fraccionamiento en fase reversa: Se equilibraron 100 μL de bolas de RPC Poly-BioReverse phase fractionation: 100 μL of Poly-Bio RPC balls were balanced
(BioSepra) con ACN al 5% /TFA al 0.5%. Para la purificación e identificación de los marcadores de 2193 y 2793 Da, la reducción y alquilación fue antes que la cromatografía de fase reversa (ver la sección de reducción y alquilación más adelante). Para la cromatografía en fase reversa, se ajustó un volumen apropiado de cada muestra a una concentración final de ACN al 5% /TFA al 0.5% y se mezcló durante 1 hora con las bolas de RPC a temperatura ambiente. El tubo se centrifugó y el sobrenadante se eliminó mediante aspiración. Las proteínas unidas se eluyeron sucesivamente con 400 μL de ACN al 20%, (al 25%), al 30%, (al 35%), al 40% y al 50% en TFA al 0,1% (ACN al 25% y al 35% se usaron sólo para la purificación de los péptidos de 2193 y 2793 Da). Se hizo el perfil de una alícuota de 2 μL de cada fracción en una matriz de ProteinChip NP20 para determinar el patrón de elución de los marcadores de interés. También se hizo el perfil de alícuotas de las fracciones seleccionadas en una matriz de ProteinChip CMlO.(BioSepra) with 5% ACN / 0.5% TFA. For the purification and identification of the 2193 and 2793 Da markers, the reduction and alkylation was before the reverse phase chromatography (see the reduction and alkylation section below). For reverse phase chromatography, an appropriate volume of each sample was adjusted to a final concentration of 5% ACN / 0.5% TFA and mixed for 1 hour with the RPC balls at room temperature. The tube was centrifuged and the supernatant was removed by aspiration. The bound proteins were eluted successively with 400 μL of 20% ACN, (25%), 30%, (35%), 40% and 50% in 0.1% TFA (25% ACN and 35% were used only for the purification of the 2193 and 2793 Da peptides). The profile of a 2 μL aliquot of each fraction was made in a ProteinChip NP20 matrix to determine the elution pattern of the markers of interest. The aliquot profile of the selected fractions was also made in a ProteinChip CMlO matrix.
Fraccionamiento mediante sistema proteominer: Para enriquecer el pico 4.6 kDa antes de su purificación, se utilizó como paso preliminar, el fraccionamiento mediante el sistema proteominer: Se equilibró 1 mi de resina Proteominer en PBS, se añadió 40 mi de orina y se incubó a 4o C toda la noche en rotación. La fracción no unida se recogió y la resina se transfirió a una columna para su elución. La fracción unida se eluyó sucesivamente en 1 mi de 1) 2.2 M tiourea, 7.7 M urea, 4.4% CHAPS, 2) 9 M urea, 25 mM ácido cítrico, pH 3.8 and 3) 33.3% alcohol isopropilo, 16.7% acetonitrilo, 0.5% ácido trifluoroacético. La columna se incubó 10 minutos a temperatura ambiente en rotación antes de recoger cada elución para asegurar la máxima eficacia.Fractionation using a proteominer system: To enrich the 4.6 kDa peak before purification, fractionation was used as a preliminary step using the proteominer system: 1 ml of Proteominer resin in PBS was equilibrated, 40 ml of urine was added and incubated at 4 or C overnight in rotation. The unbound fraction was collected and the resin was transferred to a column for elution. The bound fraction was eluted successively in 1 ml of 1) 2.2 M thiourea, 7.7 M urea, 4.4% CHAPS, 2) 9 M urea, 25 mM citric acid, pH 3.8 and 3) 33.3% isopropyl alcohol, 16.7% acetonitrile, 0.5% trifluoroacetic acid. The column was incubated 10 minutes at room temperature in rotation before collecting each elution to ensure maximum efficiency.
Fraccionamiento IMAC-Cu: La resina IMAC HyperCel se cargó con 100 mM de sulfato de cobre y se equilibró en PBS, 0.5 M NaCl. Las muestras se diluyeron con PBS/0.5 M NaCl, y se incubaron con la resina a 4o C durante toda la noche en rotación. Para la purificación del pico 4.6 kDa, la fracción no unida del proteominer se diluyó 1 :3 en PBS/0.5 M NaCl y se incubó con 1.5 mi de resina cargada con cobre. Para la purificación de los picos 5.8 y 7.6/7.7 kDa, la orina fue diluida 1 :2 en PBS/0.5 M NaCl. Las muestras de pacientes se juntaron (25 mi) y se incubaron en 1.25 mi de resina IMAC-Cu y la muestra control (25 mi) se incubó con 2 mi de resina IMAC-Cu. La fracción no unida se recogió y las resinas se transfirieron a dos columnas, donde se realizó un lavado con PBS/0.5 M NaCl. Las proteínas unidas se eluyeron con 1.5 volúmenes de columna de 5, 10, 20, 30, 40, 50, 100, y 200 mM imidazol en 50 mM Hepes, pH 7.0. Una alícuota de cada elución se diluyó 1 :10 y sus perfiles peptídicos se analizaron en chips CMlO y QlO. Las fracciones que contenían los picos 7.6/7.7 kDa (20 y 30 mM imidazol) se juntaron y se diluyeron 1 :2 en 50 mM Tris-HCl pH 9, y se incubaron con 0.25 mi de resina Q durante toda la noche a 4o C. La elución de las proteínas unidas se realizó tal y como se ha indicado en el apartado de fraccionamiento de intercambio aniónico.IMAC-Cu fractionation: IMAC HyperCel resin was loaded with 100 mM copper sulfate and equilibrated in PBS, 0.5 M NaCl. The samples were diluted with PBS / 0.5 M NaCl, and incubated with the resin at 4 ° C overnight in rotation. For the purification of the 4.6 kDa peak, the unbound fraction of the proteominer was diluted 1: 3 in PBS / 0.5 M NaCl and incubated with 1.5 ml of copper-charged resin. For the purification of peaks 5.8 and 7.6 / 7.7 kDa, the urine was diluted 1: 2 in PBS / 0.5 M NaCl. Patient samples were pooled (25 ml) and incubated in 1.25 ml of IMAC-Cu resin and the control sample (25 ml) was incubated with 2 ml of IMAC-Cu resin. The unbound fraction was collected and the resins were transferred to two columns, where a wash with PBS / 0.5 M NaCl was performed. Bound proteins were eluted with 1.5 column volumes of 5, 10, 20, 30, 40, 50, 100, and 200 mM imidazole in 50 mM Hepes, pH 7.0. An aliquot of each elution was diluted 1: 10 and its peptide profiles were analyzed on CMlO and QlO chips. Fractions containing 7.6 / 7.7 kDa peaks (20 and 30 mM imidazole) were pooled and diluted 1: 2 in 50 mM Tris-HCl pH 9, and incubated with 0.25 ml of Q resin overnight at 4 o C. Elution of bound proteins was performed as indicated in the anion exchange fractionation section.
Purificación en resina 18: La purificación del péptido 5.8 kDa incluye un paso de purificación mediante intercambio hidrofóbico en resina 18. Esta resina se equilibró en tampón de unión ((150 mM NaCl in 100 mM Acetato sódico pH 5) y se incubó durante toda la noche a 4o C. La orina de pacientes y la orina control se incubó con 100 ul y 200 ul de resina 18 respectivamente. La fracción no unida se recogió y las proteínas unidas a la resina se eluyeron sucesivamente con 1.5 volúmenes de los siguientes tampones: El) 4.5 M Urea, E2) 9 M urea, E3) 9 M urea, 0.6% hidróxido amónico, E4) 9 M urea, 1.2% hidróxido amónico, and E5) 9 M urea, 2.4% hidróxido amónico. Las fracciones provenientes de la elución con hidróxido amónico se neutralizaron con ácido acético 5 M. Una alícuota de cada fracción se diluyó 1 :10 y sus perfiles peptídicos se analizaron en chips CMlO. Electroforesis en gel: Una vez que se estimó que la proteína de interés estaba lo suficientemente enriquecida y purificada, las fracciones se concentraron en un evaporador de centrifugación CentriVap (Labconco) y se redisolvieron en 20 μL del tampón de carga de gel apropiado antes de la electroforesis en gel. Se usaron geles de acrilamida al 18%/Tris SDS-PAGE (Bio-Rad). Se incluyeron estándares de proteína Precisión Plus y/o estándares preteñidos Kaleidoscope (Bio-Rad) en cada gel con el fin de determinar el tamaño de las bandas de proteína. Los geles de proteína se tiñeron usando el kit de tinción Colloidal Blue siguiendo el protocolo del fabricante (Invitrogen).Resin purification 18: Purification of the 5.8 kDa peptide includes a purification step by hydrophobic exchange in resin 18. This resin was equilibrated in binding buffer ((150 mM NaCl in 100 mM Sodium acetate pH 5) and incubated throughout the overnight at 4 o C. Patient urine and control urine was incubated with 100 ul and 200 ul of resin respectively 18. The unbound fraction was collected and the resin bound proteins were eluted successively with 1.5 volumes of the following buffers : El) 4.5 M Urea, E2) 9 M urea, E3) 9 M urea, 0.6% ammonium hydroxide, E4) 9 M urea, 1.2% ammonium hydroxide, and E5) 9 M urea, 2.4% ammonium hydroxide. Fractions from elution with ammonium hydroxide were neutralized with 5M acetic acid. An aliquot of each fraction was diluted 1: 10 and its peptide profiles were analyzed on CMlO chips. Gel electrophoresis: Once it was estimated that the protein of interest was sufficiently enriched and purified, the fractions were concentrated in a CentriVap centrifuge evaporator (Labconco) and redissolved in 20 μL of the appropriate gel loading buffer before gel electrophoresis. 18% acrylamide / Tris SDS-PAGE (Bio-Rad) gels were used. Precision Plus protein standards and / or intended Kaleidoscope (Bio-Rad) standards were included in each gel in order to determine the size of the protein bands. Protein gels were stained using the Colloidal Blue staining kit following the manufacturer's protocol (Invitrogen).
Extracción analítica: Las bandas de proteínas seleccionadas se cortaron del gel de poliacrilamida con una pipeta pasteur y se extrajeron para confirmar la m/z de la proteína y preparar la muestra para digestión. Los trozos de geles se lavaron dos veces con 200 μL de metanol al 50% /ácido acético al 10% durante 20 minutos, se deshidrataron con 100 μL de ACN durante 15 minutos, y se extrajeron con 70 μL de ácido fórmico al 50% /ACN al 25% /IPA al 15% incubando 2 horas a temperatura ambiente con agitación vigorosa. Se hizo el perfil de una alícuota de 1-2 μL del extracto en una matriz de ProteinChip NP20.Analytical extraction: The selected protein bands were cut from the polyacrylamide gel with a pasteur pipette and extracted to confirm the m / z of the protein and prepare the sample for digestion. The pieces of gels were washed twice with 200 μL of 50% methanol / 10% acetic acid for 20 minutes, dehydrated with 100 μL of ACN for 15 minutes, and extracted with 70 μL of 50% formic acid / 25% ACN / 15% IPA incubating 2 hours at room temperature with vigorous stirring. The profile of a 1-2 μL aliquot of the extract was made on a ProteinChip NP20 matrix.
Identificación de proteínas:Protein Identification:
Identificación de los picos 8.85, 2.19 y 2.79 kDa: Los trozos de geles se lavaron dos veces con 200 μL de metanol al 50% /ácido acético al 10% durante 20 minutos, se deshidrataron con 100 μL de ACN durante 15 minutos, y se extrajeron con 70 μL de ácido fórmico al 50% /ACN al 25% /IPA al 15% incubando 2 horas a temperatura ambiente con agitación vigorosa. Se hizo el perfil de una alícuota de 1-2 μL del extracto en una matriz de ProteinChip NP20.Identification of peaks 8.85, 2.19 and 2.79 kDa: The pieces of gels were washed twice with 200 μL of 50% methanol / 10% acetic acid for 20 minutes, dehydrated with 100 μL of ACN for 15 minutes, and extracted with 70 μL of 50% formic acid / 25% ACN / 15% IPA by incubating 2 hours at room temperature with vigorous stirring. The profile of a 1-2 μL aliquot of the extract was made on a ProteinChip NP20 matrix.
El marcador de 8,85 kDa se redujo y alquiló después de la extracción del gel. El extracto analítico se secó en un concentrador de centrifugación CentriVap (Labconco), se enjuagó dos veces con hidróxido de amonio, y se secó. El precipitado se resuspendió después en 10 μL de DTT 2,5 mM, bicarbonato de amonio 50 mM pH 8 y se incubó a 600C durante 15 minutos. Se añadió entonces un exceso de yodoacetamida a la reacción y se incubó durante 15 minutos en la oscuridad. La reacción de alquilación se extinguió con un exceso de DTT durante 15 minutos en la oscuridad. Antes de la digestión con tripsina, esta muestra se secó de nuevo en el CentriVap. Para el enriquecimiento de los marcadores de 2193 y 2793 Da, las muestras se redujeron y alquilaron antes de la purificación. Se añadió DTT a las muestras a una concentración de 5 mM y se incubaron durante 1 hora a 220C. Se añadió entonces un exceso de yodoacetamida a la reacción y se incubó durante 15 minutos en la oscuridad. La reacción de alquilación se extinguió con un exceso de DTT durante 15 minutos en la oscuridad. La digestión en solución para los digeridos de tripsina, los extractos de geles secos alquilados se rehidrataron con 20 μL de bicarbonato de amonio 50 mM (pH 8) que contenía 5 ng/μL de tripsina modificada (Roche Applied Science) y se incubaron durante 3 horas a 370C. La identificación de las proteínas mediante fragmentación de péptidos usando un espectrómetro de masas en tándem. Los espectros MS individual y MS/MS se adquirieron en un espectrómetro de masas en tándem. Se mezclaron volúmenes iguales de digeridos de tripsina y CHCA saturado y se dirigieron a la diana de MALDI. Los digeridos también se unieron a μC18 Zip-Tips (Millipore), se eluyeron en ACN al 50%/TFA al 0,1%, se mezclaron con un volumen igual de y CHCA saturado y se dirigieron a la diana de MALDI. Se recogieron los espectros de 600 a 4000 Da en modo MS individual. Después de revisar los espectros, se seleccionaron iones específicos para análisis de MS/MS. Los espectros de MS/MS se enviaron a la herramienta de extracción de base de datos Mascot (Matrix Sciences) para su identificación.The 8.85 kDa marker was reduced and rented after gel extraction. The analytical extract was dried in a CentriVap centrifuge concentrator (Labconco), rinsed twice with ammonium hydroxide, and dried. The precipitate was then resuspended in 10 .mu.l of 2.5 mM DTT, 50 mM ammonium bicarbonate pH 8 and incubated at 60 0 C for 15 minutes. An excess of iodoacetamide was then added to the reaction. and incubated for 15 minutes in the dark. The alkylation reaction was quenched with an excess of DTT for 15 minutes in the dark. Before digestion with trypsin, this sample was dried again in the CentriVap. For the enrichment of the 2193 and 2793 Da markers, the samples were reduced and rented before purification. DTT was added to the samples at a concentration of 5 mM and incubated for 1 hour at 22 ° C. An excess of iodoacetamide was then added to the reaction and incubated for 15 minutes in the dark. The alkylation reaction was quenched with an excess of DTT for 15 minutes in the dark. Digestion in solution for trypsin digested, alkylated dried gels extracts were rehydrated with 20 μL of 50 mM ammonium bicarbonate (pH 8) containing 5 ng / μL of modified trypsin (Roche Applied Science) and incubated for 3 hours at 37 0 C. Identification of proteins by peptide fragmentation using a tandem mass spectrometer. The individual MS and MS / MS spectra were acquired on a tandem mass spectrometer. Equal volumes of trypsin digested and saturated CHCA were mixed and directed to the MALDI target. The digested were also linked to μC18 Zip-Tips (Millipore), eluted in 50% ACN / 0.1% TFA, mixed with an equal volume of and saturated CHCA and directed to the MALDI target. The spectra of 600 to 4000 Da were collected in individual MS mode. After reviewing the spectra, specific ions were selected for MS / MS analysis. The MS / MS spectra were sent to the Mascot database extraction tool (Matrix Sciences) for identification.
Identificación de los picos 7.7, 7.6, 5.8 y 4.6 kDa: Las bandas correspondientes de los respectivos geles se recortaron y se les añadieron 100 μl de DTT 5 mM, y se incubaron durante 30 minutos a 37 0C. Seguidamente se retiró el DTT, se añadieron 100 μl de iodoacetamida y se incubaron otros 30 minutos a temperatura ambiente en oscuridad. La bandas de gel se lavaron dos veces con bicarbonato amónico pH8 50 mM y se secaron en un evaporador CentriVap, para su posterior digestión.Identification of peaks 7.7, 7.6, 5.8 and 4.6 kDa: The corresponding bands of the respective gels were trimmed and 100 µl of 5 mM DTT was added, and they were incubated for 30 minutes at 37 0 C. Then the DTT was removed, 100 µl of iodoacetamide was added and incubated another 30 minutes at room temperature in the dark. The gel bands were washed twice with 50 mM pH8 ammonium bicarbonate and dried on a CentriVap evaporator, for subsequent digestion.
Las bandas cortadas se trataron para eliminar la tinción de Coomassie y el SDS, incubando sucesivamente con 400 μl de metanol 50% / ácido acético 10% (una hora para la primera incubación y 30 minutos para la segunda) y con 400 μl de acetonitrilo 50% / bicarbonato amónico pH8 100 mM durante 30 minutos, y con 100 μl de acetronitrilo durante 10 minutos. Las bandas de gel se secaron en un evaporador CentriVap. Para la digestión, las bandas secas se rehidrataron en 20 μl de bicarbonato de amonio pH8 50 mM con, 5 ng/μl de tripsina o 10 ng /μl de AspN. Algunas digestiones se unieron a Zip/Tips (Millipore), se lavaron dos veces con acetonitrilo 5% / trifluoroacético 0.1% y se eluyeron en acetonitrilo 50% / trifluoroacético 0.1%. La identificación se realizó mediante fragmentación peptídica en un espectrómetro de masas en tándem. Las muestras se aplicaron en el espectrómetro MALDI utilizando el siguiente método: se añadió 0.5 μl de CHCA saturada al 25% en la placa, seguidamente se aplicó 1-2 μl de muestra y otros 0.5 μl de CHCA saturada al 25%. Los espectros se recogieron en el modo reflectron de 500 a 6000 Da. Los iones específicos se seleccionaron para el análisis MS/MS. Los espectros de fragmentación se enviaron a la herramienta de extracción de base de datos Mascot (Matrix Sciences) para su identificación.The cut bands were treated to remove Coomassie stain and SDS, incubating successively with 400 µl of 50% methanol / 10% acetic acid (one hour for the first incubation and 30 minutes for the second) and with 400 µl of acetonitrile 50% / 100 mM pH8 ammonium bicarbonate for 30 minutes, and with 100 μl of acetronitrile for 10 minutes. The gel bands were dried on a CentriVap evaporator. For digestion, the dried bands were rehydrated in 20 µl of 50 mM pH8 ammonium bicarbonate with, 5 ng / µl trypsin or 10 ng / µl AspN. Some digestions were attached to Zip / Tips (Millipore), washed twice with 5% acetonitrile / 0.1% trifluoroacetic acid and eluted in 50% acetonitrile / 0.1% trifluoroacetic acid. Identification was performed by peptide fragmentation in a tandem mass spectrometer. Samples were applied on the MALDI spectrometer using the following method: 0.5 µl of 25% saturated CHCA was added to the plate, then 1-2 µl of sample and another 0.5 µl of 25% saturated CHCA were applied. The spectra were collected in the reflectron mode of 500 to 6000 Da. Specific ions were selected for the MS / MS analysis. Fragmentation spectra were sent to the Mascot (Matrix Sciences) database extraction tool for identification.
RESULTADOSRESULTS
DIGESay
Después del procesamiento de las muestras, se obtuvieron los geles DIGE de cinco controles sanos y cinco pacientes cirróticos (Fig. 1). Las manchas se detectaron y compararon usando el software DeCyder con el análisis estadístico prueba T independiente. En este análisis se encontraron dieciocho manchas diferenciales con un valor de P menor de 0,05 y un cociente medio mayor de 1,5. Diecisiete de estas proteínas disminuían en los pacientes cirróticos y sólo una de ellas aumentaba en los pacientes. Para identificar las proteínas diferenciales, se hizo un gel preparativo juntando varias muestras control, y las proteínas se tiñeron con la tinción Sypro RubyAfter sample processing, DIGE gels were obtained from five healthy controls and five cirrhotic patients (Fig. 1). The spots were detected and compared using the DeCyder software with the independent T-test statistical analysis. In this analysis, eighteen differential spots were found with a P value of less than 0.05 and an average ratio greater than 1.5. Seventeen of these proteins decreased in cirrhotic patients and only one of them increased in patients. To identify the differential proteins, a preparative gel was made by combining several control samples, and the proteins were stained with the Sypro Ruby stain
(Biorad). Las manchas se cogieron, se digirieron con tripsina y se analizaron mediante(Biorad). The spots were taken, digested with trypsin and analyzed by
ESI-MS/MS. No fue posible coger algunas de las proteínas diferenciales, debido a la baja sensibilidad del gel preparativo. Los perfiles peptídicos y las secuencias de cada mancha se analizaron con las máquinas de búsqueda tanto Phenix 2.3 comoESI-MS / MS. It was not possible to take some of the differential proteins, due to the low sensitivity of the preparative gel. The peptide profiles and sequences of each spot were analyzed with both Phenix 2.3 and search engines.
ProteinLynx Global Server 2.3 en la base de datos Uniprot Global Server. Se identificaron dos proteínas: El precursor de apolipoproteína D se produce en varios tejidos y se secreta al plasma, está implicado en la unión y transporte de bilina.ProteinLynx Global Server 2.3 in the Uniprot Global Server database. Two proteins were identified: The apolipoprotein D precursor is produced in various tissues and is secreted to plasma, is involved in the binding and transport of biline.
El precursor de gelsolina despolimerizante de plasma f es una proteína moduladora de actina secretada al plasma que se une a actina y fϊbronectina. Puede fomentar el ensamblaje de monómeros en filamentos así como separar filamentos ya formados.The plasma depolymerizing gelsolin precursor f is a plasma secreted actin modulating protein that binds actin and f ybronectin. It can promote the assembly of monomers in filaments as well as separate filaments already formed.
Las muestras usadas en el estudio de 2D-DIGE se sometieron a análisis de inmunotransferencia con anticuerpos comerciales para estas dos proteínas, confirmando que los datos obtenidos en análisis por DIGE eran debidos a una caída en los niveles totales de proteína en los pacientes cirróticos (Fig. 2).The samples used in the 2D-DIGE study were subjected to immunoblot analysis with commercial antibodies for these two proteins, confirming that the data obtained in DIGE analysis were due to a drop in total protein levels in cirrhotic patients (Fig . 2).
SELDI TOF Se realizaron dos estudios usando la técnica SELDI-TOF. El primero se realizó en Centro Nacional de Biotecnología (CNB) español, y el segundo en el Centro de Investigación de Biomarcadores en los laboratorios de Bio-Rad.SELDI TOF Two studies were performed using the SELDI-TOF technique. The first was carried out in the Spanish National Biotechnology Center (CNB), and the second in the Biomarker Research Center in the Bio-Rad laboratories.
ESTUDIO DEL CNB En este estudio, se compararon 15 muestras de fϊbrosis hepática con 15 casos control en los chips CMlO (intercambio catiónico), e IMAC-Cu (afinidad a metal). El análisis de los espectros dio cinco péptidos significativamente diferentes (P <0,01). Uno de estos marcadores era el péptido de 8855 Da con un valor de P = 5,7 x 10"4 y un ROC de 0,74.CNB STUDY In this study, 15 samples of liver fibrosis were compared with 15 control cases in CMlO chips (cation exchange), and IMAC-Cu (metal affinity). Spectra analysis gave five significantly different peptides (P <0.01). One of these markers was the 8855 Da peptide with a value of P = 5.7 x 10 "4 and an ROC of 0.74.
ESTUDIO DE BIORADBIORAD STUDY
El propósito de este estudio era obtener información adicional, aumentando tanto el número de muestras como los tipos de chips. El número de muestras se aumentó a 35 pacientes con cirrosis y 35 controles sanos. Además, se añadió un grupo de 10 pacientes con fϊbrosis baja (índice de Metavir Fl). El número de chips usados también aumentó a cuatro: CMlO (intercambio catiónico), IMAC30 (afinidad a metal), QlO (intercambio aniónico) y H5 (intercambio hidrofóbico). El análisis de los datos produjo 61 picos que mostraron diferencias estadísticamente significativas (P < 0,01) entre pacientes cirróticos y controles sanos. El grupo de fibrosis era demasiado pequeño para un análisis estadístico consistente, pero proporcionó alguna información sobre la especificidad de los marcadores, mostrando un comportamiento intermedio entre las muestras cirróticas y controles (Figs. 3, 5 y 6).The purpose of this study was to obtain additional information, increasing both the number of samples and the types of chips. The number of samples was increased to 35 patients with cirrhosis and 35 healthy controls. In addition, a group of 10 patients with low fibrosis (Metavir Fl index) was added. The number of chips used also increased to four: CMlO (cation exchange), IMAC30 (metal affinity), QlO (anion exchange) and H5 (hydrophobic exchange). The data analysis produced 61 peaks that showed statistically significant differences (P <0.01) between cirrhotic patients and healthy controls. The fibrosis group was too small for a consistent statistical analysis, but provided some information on the specificity of the markers, showing an intermediate behavior between the cirrhotic samples and controls (Figs. 3, 5 and 6).
Este segundo estudio proporcionó la confirmación de cuatro marcadores detectados en el análisis del CNB. Uno de estos marcadores confirmados es el péptido de 8855 Da, detectado con un valor de P = l,lx 10" y un valor ROC = 0,74.This second study provided confirmation of four markers detected in the CNB analysis. One of these confirmed markers is the 8855 Da peptide, detected with a value of P = 1, 10 x 10 " and an ROC value = 0.74.
En conclusión, el análisis de péptidos realizado en los laboratorios de Bio-Rad proporciona, primero, confirmación de uno de los péptidos identificados en el estudio previo, y segundo, nuevos marcadores candidatos identificados por medio del chip QlO.In conclusion, the peptide analysis performed in the Bio-Rad laboratories provides, first, confirmation of one of the peptides identified in the previous study, and second, new candidate markers identified by means of the QlO chip.
La prevalencia de los marcadores negativos encontrados en el estudio de DIGE fue confirmada ya que la mayoría de los marcadores candidatos (57 de 61) disminuían en pacientes con cirrosis.The prevalence of negative markers found in the DIGE study was confirmed as most of the candidate markers (57 of 61) decreased in patients with cirrhosis.
Además, se aplicó otro modelo estadístico a los datos: el modelo lineal. En este caso, la prueba aplicada era el paquete LIMMA de Bioconductor. En este análisis, se encontraron dieciséis péptidos significativamente disminuidos en las muestras cirróticas (B>0).In addition, another statistical model was applied to the data: the linear model. In this case, the test applied was the LIMMA Bioconductor package. In this analysis, sixteen significantly decreased peptides were found in cirrhotic samples (B> 0).
Los péptidos que se consideraron significativamente diferentes en ambos estudios estadísticos univariantes se eligieron para realizar el análisis estadístico multivariante para mejorar el éxito de la clasificación de las muestras combinando marcadores individuales.Peptides that were considered significantly different in both univariate statistical studies were chosen to perform multivariate statistical analysis to improve the success of the classification of the samples by combining individual markers.
La Tabla 1 recoge aquellos péptidos identificados en los dos estudios estadísticos univariantes que mostraban niveles de expresión significativamente diferentes entre controles y pacientes cirróticos. La tabal indica la proteína que se ha asignado a cada péptido y los valores p y ROC obtenidos a la hora de discriminar entre pacientes controles y cirróticos. Tabla 1Table 1 lists those peptides identified in the two univariate statistical studies that showed significantly different levels of expression between controls and cirrhotic patients. The tabal indicates the protein that has been assigned to each peptide and the ROC p values obtained when discriminating between control and cirrhotic patients. Table 1
Figure imgf000049_0001
Figure imgf000049_0001
Las muestras usadas en el estudio de SELDI se sometieron a análisis de inmunotransferencia con anticuerpos comerciales para la proteína SLURPl, confirmando que los datos obtenidos en análisis por SELDI eran debidos a una caída en los niveles totales de proteína en los pacientes cirróticos. La figura 4 muestra la disminución de los niveles de la proteína SLURPl en muestras de orina de pacientes cirróticos con respecto a los controles detectada mediante Western Blot.The samples used in the SELDI study were subjected to immunoblot analysis with commercial antibodies to the SLURPl protein, confirming that the data obtained in SELDI analysis were due to a drop in total protein levels in cirrhotic patients. Figure 4 shows the decrease in SLURPl protein levels in urine samples from cirrhotic patients with respect to the controls detected by Western Blot.
Una herramienta útil para evaluar la capacidad diagnóstica de una prueba cuantitativa, es la denominada curva ROC. También nos servirá para comparar diferentes pruebas. Para obtener la curva ROC, se calcula la sensibilidad (proporción de verdaderos positivos) y especificidad (proporción de verdaderos negativos) para cada uno de los diferentes valores observados en nuestros datos y se representan en una gráfica, con la Sensibilidad en el eje de las Y, (1 -Especificidad) en el eje de las X. Por lo tanto, cuanto más desplazada esté la curva ROC hacia el vértice superior izquierdo, mejor es la capacidad discriminatoria de la prueba. Precisamente una forma de evaluar de manera global esa capacidad de discriminación consiste en calcular el área del polígono que queda debajo de la curva ROC, y se denomina área bajo la curva, sirviendo como índice de comparación entre pruebas diagnósticas, cuanto mayor es el área mejor es la capacidad diagnóstica.A useful tool to evaluate the diagnostic capacity of a quantitative test is the so-called ROC curve. It will also help us compare different tests. To obtain the ROC curve, the sensitivity (proportion of true positives) and specificity (proportion of true negatives) are calculated for each of the different values observed in our data and are represented in a graph, with the Sensitivity in the axis of the And, (1 -Specificity) on the X axis. Therefore, the more displaced the ROC curve towards the upper left vertex, the better the discriminatory capacity of the test. Precisely one way to assess this discrimination capacity in a global way is to calculate the area of the polygon that is below the ROC curve, and is called the area under the curve, serving as an index Comparison between diagnostic tests, the larger the area, the better the diagnostic capacity.
El análisis estadístico multivariante de la combinación de los marcadores 4623 / 7655 / 7735 / 8853 / 2193 / 2793 / 5804 produjo un patrón para diferenciar individuos cirróticos de sanos, con un valor ROC de 0,91.Multivariate statistical analysis of the combination of markers 4623/7655/7735/8853/2193/2793/5804 produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.91.
El análisis estadístico multivariante de la combinación de los marcadores 4623 / 7655 / / 8853 / 2193 / 2793 / 5804 produjo un patrón para diferenciar individuos cirróticos de sanos, con un valor ROC de 0,87.Multivariate statistical analysis of the combination of markers 4623/7655 / / 8853/2193/2793/5804 produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.87.
El análisis estadístico multivariante de la combinación de los marcadores 4623 / 7655 / 7735 / 8853 / 2193 / 2793 / produjo un patrón para diferenciar individuos cirróticos de sanos, con un valor ROC de 0,90.Multivariate statistical analysis of the combination of markers 4623/7655/7735/8853/2193/2793 / produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.90.
El análisis estadístico multivariante de la combinación de los marcadores 4623 / 7655 / 8853 / 2793 produjo un patrón para diferenciar individuos cirróticos de sanos, con un valor ROC de 0,87.Multivariate statistical analysis of the combination of markers 4623/7655/8853/2793 produced a pattern to differentiate healthy cirrhotic individuals, with an ROC value of 0.87.
La hepcidina se traduce como un péptido de 84 aminoácidos y se procesa para obtener la hepcidina-25 (residuos C-terminales 60-84 del péptido) o la hepcidina-20 (residuos C-terminales 65-84). Se ha demostrado que la hepcidina está implicada en el mantenimiento de la homeostasis de hierro. Regula tanto la absorción intestinal de hierro como el almacenamiento de hierro en macrófagos y hepatocitos. La hepcidina aumenta en respuesta a niveles aumentados de hierro e inflamación, y disminuye en respuesta a hipoxia, anemia y estrés oxidativo. Además, la hepcidina también tiene una actividad antimicrobiana fuerte contra varios microorganismos.Hepcidin is translated as an 84 amino acid peptide and processed to obtain hepcidin-25 (C-terminal residues 60-84 of the peptide) or hepcidin-20 (C-terminal residues 65-84). Hepcidin has been shown to be involved in the maintenance of iron homeostasis. Regulates both intestinal iron absorption and iron storage in macrophages and hepatocytes. Hepcidin increases in response to increased iron levels and inflammation, and decreases in response to hypoxia, anemia and oxidative stress. In addition, hepcidin also has a strong antimicrobial activity against several microorganisms.
Se ha encontrado que SLURP-I es un marcador de la diferenciación tardía de la piel y se ha mostrado que tiene actividad antitumoral. Está implicado en el mantenimiento de la integridad fisiológica y estructural de las capas de queratinocitos de la piel, defectos en SLURP-I son una causa del Mal de Meleda. La proteína MASP2 es una serina proteasa que se une a la lectina de unión a la mañosa (MBL), produciendo la activación de la vía del complemento. La MBL es capaz de unirse con estructuras repetidas de azúcares presentes en una amplia variedad de bacterias y otros microorganismos promoviendo su eliminación. Stengaard et al. (New Eng. J. Med. 349: 554-560, 2003) han descrito que la deficiencia congénita en el gen que codifica MASP2 produce una mayor susceptibilidad a infecciones y a enfermedades inmunes.SLURP-I has been found to be a marker of late skin differentiation and has been shown to have antitumor activity. It is involved in the maintenance of the physiological and structural integrity of the keratinocyte layers of the skin, defects in SLURP-I are a cause of Meleda Mal. The MASP2 protein is a serine protease that binds to the malignant binding lectin (MBL), causing activation of the complement pathway. MBL is able to bind with repeated structures of sugars present in a wide variety of bacteria and other microorganisms promoting their elimination. Stengaard et al. (New Eng. J. Med. 349: 554-560, 2003) have described that congenital deficiency in the gene encoding MASP2 results in a greater susceptibility to infections and immune diseases.
Las defensinas funcionan como antibióticos naturales que se hallan en la superficie de la piel. Son activas contra bacterias, hongos y virus con envuelta. Las beta-Defensinas tienen de 36 a 42 aminoácidos. La beta-Defensina humana 1 (hBD-1) ó DEFBl se expresa constitutivamente a nivel del tracto génito urinario y respiratorio . Se trata de un péptido antimicrobiano implicado en la resistencia de las superficies epiteliales a la colonización microbiana. Estos péptidos interactúan con varios receptores de las CDi y linfocitos, con lo cual se activan los mecanismos de la inmunidad adaptativa.Defensins function as natural antibiotics that are found on the surface of the skin. They are active against bacteria, fungi and enveloped viruses. Beta-Defensins have 36 to 42 amino acids. Human beta-Defensin 1 (hBD-1) or DEFBl is constitutively expressed at the level of the urinary and respiratory genital tract. It is an antimicrobial peptide involved in the resistance of epithelial surfaces to microbial colonization. These peptides interact with several CDi and lymphocyte receptors, thereby activating adaptive immunity mechanisms.
La osteopontina (SPPl) es una glicoproteína multifuncional con efecto estimulante sobre los fibroblastos y la síntesis de matriz extracelular. La Osteopontina se encuentra en diversos tejidos y tiene diversas funciones como la biomineralización del tejido óseo y la reparación de tejidos, fibrosis y calcificaciones distróficas tras lesiones inmunológicas, también participa en el crecimiento tumoral, en el desarrollo de cáncer y metástasis. Participa también en la iniciación de la respuesta inmune en la activación temprana de linfocitos T y macró fagos. Osteopontin (SPPl) is a multifunctional glycoprotein with a stimulating effect on fibroblasts and extracellular matrix synthesis. Osteopontin is found in various tissues and has various functions such as the biomineralization of bone tissue and tissue repair, fibrosis and dystrophic calcifications after immunological lesions, it also participates in tumor growth, in the development of cancer and metastasis. It also participates in the initiation of the immune response in the early activation of T lymphocytes and macrophages.

Claims

REIVINDICACIONES
1. Uso de al menos un biomarcador para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis o para seguir la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis en un sujeto en donde dicho biomarcador se selecciona del grupo que consiste en SLURPl, HAMP, GSN, APOD, SPPl, DEFBl y MASP2.1. Use of at least one biomarker for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or to follow the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis in a subject where said biomarker is selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
2. Uso según la reivindicación 1 en donde la enfermedad asociada con fibrosis es cirrosis.2. Use according to claim 1 wherein the disease associated with fibrosis is cirrhosis.
3. Uso según la reivindicación 1 en donde la fibrosis es fibrosis hepática.3. Use according to claim 1 wherein the fibrosis is hepatic fibrosis.
4. Un método para la detección, diagnóstico y evaluación de fibrosis y/o de una enfermedad asociada con fibrosis o para seguir la eficacia de un tratamiento para fibrosis y/o de una enfermedad asociada con fibrosis que comprende comparar la expresión de uno o más biomarcadores en una muestra de un sujeto con un estándar predeterminado de cada uno de dicho uno o más biomarcadores; en donde dicho uno o más biomarcadores se selecciona del grupo que consiste en SLURPl, HAMP, GSN, APOD, SPP l , DEFB l y MASP2; y en donde una diferencia significativa en la expresión de dicho uno o más biomarcadores en dicha muestra comparada con el estándar predeterminado de cada dicho uno o más biomarcadores es indicativa de inicio, fase, evolución de fibrosis y/o enfermedad asociada con fibrosis o eficacia de un tratamiento para la fibrosis y/o enfermedad asociada con fibrosis.4. A method for the detection, diagnosis and evaluation of fibrosis and / or a disease associated with fibrosis or for monitoring the efficacy of a treatment for fibrosis and / or a disease associated with fibrosis comprising comparing the expression of one or more biomarkers in a sample of a subject with a predetermined standard of each of said one or more biomarkers; wherein said one or more biomarkers is selected from the group consisting of SLURPl, HAMP, GSN, APOD, SPP l, DEFB l and MASP2; and wherein a significant difference in the expression of said one or more biomarkers in said sample compared to the predetermined standard of each said one or more biomarkers is indicative of onset, phase, evolution of fibrosis and / or disease associated with fibrosis or efficacy of a treatment for fibrosis and / or disease associated with fibrosis.
5. Un método según la reivindicación 4 en donde un descenso en la expresión de uno o más biomarcadores es indicativo de que el sujeto padece fibrosis y/o una enfermedad asociada con fibrosis.5. A method according to claim 4 wherein a decrease in the expression of one or more biomarkers is indicative that the subject suffers from fibrosis and / or a disease associated with fibrosis.
6. Un método según las reivindicaciones 4 ó 5 en donde un aumento en la expresión en uno o más biomarcadores es indicativo que el tratamiento para la fibrosis y/o la enfermedad asociada con fibrosis es eficaz. 6. A method according to claims 4 or 5 wherein an increase in expression in one or more biomarkers is indicative that the treatment for fibrosis and / or the disease associated with fibrosis is effective.
7. Un método como se define en cualquiera de las reivindicaciones 5 a 6 en donde la enfermedad asociada con fibrosis es cirrosis.7. A method as defined in any of claims 5 to 6 wherein the disease associated with fibrosis is cirrhosis.
8. Un método como se define en cualquiera de las reivindicaciones 5 a 7 en donde la fibrosis es fibrosis hepática.8. A method as defined in any of claims 5 to 7 wherein the fibrosis is liver fibrosis.
9. Un método como se define en cualquiera de las reivindicaciones 5 a 8 en donde el estándar predeterminado es el nivel de expresión en una muestra de referencia obtenida combinando cantidades iguales de muestras de una población de sujetos.9. A method as defined in any of claims 5 to 8 wherein the predetermined standard is the level of expression in a reference sample obtained by combining equal amounts of samples from a population of subjects.
10. Un método como se define en cualquiera de las reivindicaciones 5 a 9 en donde el estándar predeterminado es el nivel del biomarcador(es) en una muestra de referencia obtenida del mismo sujeto en el que se va a evaluar la eficacia de la terapia previa a la iniciación de la terapia.10. A method as defined in any of claims 5 to 9 wherein the predetermined standard is the level of the biomarker (s) in a reference sample obtained from the same subject in which the efficacy of the previous therapy is to be evaluated. to the initiation of therapy.
11. Un método como se define en cualquiera de las reivindicaciones 5 a 10 en donde la muestra es orina.11. A method as defined in any of claims 5 to 10 wherein the sample is urine.
12. Un método como se define en la reivindicación 1 1 en donde la expresión del biomarcador o bio marcadores se determina mediante la cuantificación de los niveles de la proteína o proteínas correspondientes.12. A method as defined in claim 1 wherein the expression of the biomarker or bio markers is determined by quantifying the levels of the corresponding protein or proteins.
13. Un método como se define en la reivindicación 12 en donde la determinación de los niveles de la proteína o proteínas se lleva a cabo mediante inmunotransferencia.13. A method as defined in claim 12 wherein the determination of the levels of the protein or proteins is carried out by immunoblotting.
14. Un kit que comprende reactivos para detectar al menos dos o más biomarcadores seleccionados del grupo de SLURPl, HAMP, GSN, APOD, SPPl , DEFB l Y MASP2.14. A kit comprising reagents for detecting at least two or more biomarkers selected from the group of SLURPl, HAMP, GSN, APOD, SPPl, DEFBl and MASP2.
15. Un kit como se define en la reivindicación 14 en donde los biomarcadores son polipéptidos. 15. A kit as defined in claim 14 wherein the biomarkers are polypeptides.
PCT/ES2010/070007 2009-01-09 2010-01-07 Bio-markers for diagnosing fibrosis WO2010079253A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ESP200900056 2009-01-09
ES200900056 2009-01-09

Publications (2)

Publication Number Publication Date
WO2010079253A2 true WO2010079253A2 (en) 2010-07-15
WO2010079253A3 WO2010079253A3 (en) 2010-10-14

Family

ID=42232658

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/ES2010/070007 WO2010079253A2 (en) 2009-01-09 2010-01-07 Bio-markers for diagnosing fibrosis

Country Status (1)

Country Link
WO (1) WO2010079253A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010133967A1 (en) * 2009-05-14 2010-11-25 University Of Oxford Clinical diagnosis of hepatic fibrosis using a novel panel of low abundant human plasma protein biomarkers
US9012162B2 (en) 2006-09-08 2015-04-21 The Chancellor, Masters And Scholars Of The University Of Oxford Clinical diagnosis of hepatic fibrosis using a novel panel of human serum protein biomarkers
US11366124B2 (en) 2016-08-24 2022-06-21 ShOx Science Limited Clinical diagnosis of non-alcoholic fatty liver disease using a panel of human blood protein biomarkers

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5045694A (en) 1989-09-27 1991-09-03 The Rockefeller University Instrument and method for the laser desorption of ions in mass spectrometry
US5118937A (en) 1989-08-22 1992-06-02 Finnigan Mat Gmbh Process and device for the laser desorption of an analyte molecular ions, especially of biomolecules
US5719060A (en) 1993-05-28 1998-02-17 Baylor College Of Medicine Method and apparatus for desorption and ionization of analytes
WO1998059361A1 (en) 1997-06-20 1998-12-30 Ciphergen Biosystems, Inc. Retentate chromatography and protein chip arrays with applications in biology and medicine
WO2002016949A1 (en) 2000-08-21 2002-02-28 Assistance Publique - Hopitaux De Paris Diagnosis method of fibrotic disease using biochemical markers
US20030077616A1 (en) 2001-04-19 2003-04-24 Ciphergen Biosystems, Inc. Biomolecule characterization using mass spectrometry and affinity tags
US20030134304A1 (en) 2001-08-13 2003-07-17 Jan Van Der Greef Method and system for profiling biological systems
WO2003073822A2 (en) 2002-02-28 2003-09-12 Prometheus Laboratories, Inc. Methods of diagnosing liver fibrosis
US20030199001A1 (en) 2002-04-23 2003-10-23 Pitt Aldo M. Sample preparation of biological fluids for proteomic applications
WO2005116901A2 (en) 2004-05-14 2005-12-08 Universite D'angers Method of diagnosing the presence and/or severity of a hepatic pathology in an individual and/or of monitoring the effectiveness of a treatment for one such pathology
WO2007003670A1 (en) 2005-07-01 2007-01-11 Proyecto De Biomedicina Cima, S.L. Fibrosis markers
EP1811041A1 (en) 2006-01-24 2007-07-25 Industrial Technology Research Institute Biomarkers for liver fibrotic injury
WO2008031051A2 (en) 2006-09-08 2008-03-13 United Therapeutics Corporation Clinical diagnosis of hepatic fibrosis using a novel panel of human serum protein biomarkers

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008123948A1 (en) * 2007-03-27 2008-10-16 Vermillion, Inc. Biomarkers for ovarian cancer

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5118937A (en) 1989-08-22 1992-06-02 Finnigan Mat Gmbh Process and device for the laser desorption of an analyte molecular ions, especially of biomolecules
US5045694A (en) 1989-09-27 1991-09-03 The Rockefeller University Instrument and method for the laser desorption of ions in mass spectrometry
US5719060A (en) 1993-05-28 1998-02-17 Baylor College Of Medicine Method and apparatus for desorption and ionization of analytes
WO1998059361A1 (en) 1997-06-20 1998-12-30 Ciphergen Biosystems, Inc. Retentate chromatography and protein chip arrays with applications in biology and medicine
WO2002016949A1 (en) 2000-08-21 2002-02-28 Assistance Publique - Hopitaux De Paris Diagnosis method of fibrotic disease using biochemical markers
US20030077616A1 (en) 2001-04-19 2003-04-24 Ciphergen Biosystems, Inc. Biomolecule characterization using mass spectrometry and affinity tags
US20030134304A1 (en) 2001-08-13 2003-07-17 Jan Van Der Greef Method and system for profiling biological systems
WO2003073822A2 (en) 2002-02-28 2003-09-12 Prometheus Laboratories, Inc. Methods of diagnosing liver fibrosis
US20030199001A1 (en) 2002-04-23 2003-10-23 Pitt Aldo M. Sample preparation of biological fluids for proteomic applications
WO2005116901A2 (en) 2004-05-14 2005-12-08 Universite D'angers Method of diagnosing the presence and/or severity of a hepatic pathology in an individual and/or of monitoring the effectiveness of a treatment for one such pathology
WO2007003670A1 (en) 2005-07-01 2007-01-11 Proyecto De Biomedicina Cima, S.L. Fibrosis markers
EP1811041A1 (en) 2006-01-24 2007-07-25 Industrial Technology Research Institute Biomarkers for liver fibrotic injury
WO2008031051A2 (en) 2006-09-08 2008-03-13 United Therapeutics Corporation Clinical diagnosis of hepatic fibrosis using a novel panel of human serum protein biomarkers

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
BERGMAN, EXS, vol. 88, 2000, pages 133 - 44
CAMPBELL ET AL.: "Methods and Immunology", 1964, W. A. BENJAMIN, INC.
CHAIT ET AL., SCIENCE, vol. 262, 1993, pages 89 - 92
DOWDY; WEARDEN: "Statistics for Research", 1983, JOHN WILEY & SONS
GANGADHARAN ET AL., CLINICAL CHEMISTRY, vol. 53, 2007, pages 1792 - 1799
IMBERT-BISMUT ET AL., LANCET, vol. 357, 2001, pages 1069 - 1075
KEOUGH ET AL., PROC. NATL. ACAD. SCI. USA., vol. 96, 1999, pages 7131 - 6
KIRK-OTHMER: "Encyclopedia of Chemical Technology, 4th ed.", vol. 15, 1995, JOHN WILEY AND SONS, pages: 1071 - 1094
KUSTER; MANN, CURR. OPIN. STRUCTURAL BIOL., vol. 8, 1998, pages 393 - 400
LI ET AL., TIBTECH, vol. 18, 2000, pages 151 - 160
OELLERICH, M., J. CLIN. CHEM. CLIN. BIOCHEM., vol. 22, 1984, pages 895 - 904
ROSE AND BIGAZZI,: "Methods in Immunodiagnosis, 2nd Edition,", 1980, JOHN WILEY AND SONS
ROWLEY ET AL., METHODS, vol. 20, 2000, pages 383 - 397
SANDRIN L ET AL., ULTRASOUND MED. BIOL., vol. 29, 2003, pages 1705 - 1711
SKOOG,: "Principles of Instrumental Analysis, 3rd edition.,", 1985, SKOOG, SAUNDERS COLLEGE PUBLISHING
STENGAARD ET AL., NEW ENG. J. MED., vol. 349, 2003, pages 554 - 560
TOWBIN, PROC. NAT. ACAD. SCI., vol. 76, 1979, pages 4350

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9012162B2 (en) 2006-09-08 2015-04-21 The Chancellor, Masters And Scholars Of The University Of Oxford Clinical diagnosis of hepatic fibrosis using a novel panel of human serum protein biomarkers
WO2010133967A1 (en) * 2009-05-14 2010-11-25 University Of Oxford Clinical diagnosis of hepatic fibrosis using a novel panel of low abundant human plasma protein biomarkers
US8889364B2 (en) 2009-05-14 2014-11-18 The Chancellor, Masters And Scholars Of The University Of Oxford Clinical diagnosis of hepatic fibrosis using a novel panel of low abundant human plasma protein biomarkers
US11366124B2 (en) 2016-08-24 2022-06-21 ShOx Science Limited Clinical diagnosis of non-alcoholic fatty liver disease using a panel of human blood protein biomarkers

Also Published As

Publication number Publication date
WO2010079253A3 (en) 2010-10-14

Similar Documents

Publication Publication Date Title
US9933439B2 (en) Method of predicting acute appendicitis
JP6198752B2 (en) Biomarkers for gastric cancer and uses thereof
JP5006802B2 (en) Cyr61 as a biomarker for cancer diagnosis and prognosis derived from epithelium
JP2008529008A (en) Methods for diagnosis and prognosis of epithelial cancer
JP2008527351A (en) Apolipoprotein A-II isoform as a biomarker for prostate cancer
EP2239576A1 (en) Composition and method for diagnosis or detection of gastric cancer
JP2004536278A (en) Method and apparatus for quantitative detection of prostate-specific membrane antigen and other prostate markers
ES2282780T3 (en) METHOD FOR THE DIAGNOSIS OF HEPATIC FIBROSIS.
ES2361808B1 (en) METHOD OF OBTAINING USEFUL DATA FOR THE DIAGNOSIS OR FORECAST OF THE COLORRECTAL CENDER NCER.
Zhang et al. Comparative analysis of the human urinary proteome by 1D SDS-PAGE and chip-HPLC-MS/MS identification of the AACT putative urinary biomarker
WO2010079253A2 (en) Bio-markers for diagnosing fibrosis
US20090220994A1 (en) Methods for diagnosis of chronic prostatitis/chronic pelvic pain syndrome
ES2303925T3 (en) METHOD FOR DIAGNOSING HEPATIC FIBROSIS.
WO2019242741A1 (en) Biomarkers for urothelial carcinoma and applications thereof
US20130023428A1 (en) Biomarkers for neurological conditions
ES2364169A1 (en) Use of apo j isoforms as tissue lesion biomarkers
ES2819055T3 (en) Method for the diagnosis / prognosis of colorectal cancer
ES2506615T3 (en) Markers for transformed epithelium and potential targets for cancer therapy of the gingiva-buccal complex
ES2323927B1 (en) IN VITRO NON-INVASIVE METHOD TO DETECT TRANSITIONAL CARCINOMA OF BLADDER.
ES2713098T3 (en) Diagnosis of heart failure
WO2018154171A9 (en) Biomarkers for diagnosing growth hormone deficiency, method and kit
ES2370671B1 (en) METHOD OF FORECAST AND DIAGNOSIS OF THE AORTIC STENOSIS THAT INCLUDES ALPHA-1-ANTICHEMIOTRIPSINE AS A MARKER.
ES2764379T3 (en) Procedure for detecting and / or evaluating annexin A3 of a mammal in the blood or in at least one of its derivatives
ES2925148A1 (en) METHOD OF DIAGNOSIS, PROGNOSIS OR PREDICTION OF THE EVOLUTION OF AORTIC STENOSIS (Machine-translation by Google Translate, not legally binding)
AU2015200306A1 (en) Method of predicting acute appendicitis

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: 10705618

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 09/12/2011)

122 Ep: pct application non-entry in european phase

Ref document number: 10705618

Country of ref document: EP

Kind code of ref document: A2