WO2009087190A1 - Means and methods for assessing the risk of patients presenting to emergency units based on gdf-15 - Google Patents

Means and methods for assessing the risk of patients presenting to emergency units based on gdf-15 Download PDF

Info

Publication number
WO2009087190A1
WO2009087190A1 PCT/EP2009/050170 EP2009050170W WO2009087190A1 WO 2009087190 A1 WO2009087190 A1 WO 2009087190A1 EP 2009050170 W EP2009050170 W EP 2009050170W WO 2009087190 A1 WO2009087190 A1 WO 2009087190A1
Authority
WO
WIPO (PCT)
Prior art keywords
subject
amount
gdf
hospital
admitted
Prior art date
Application number
PCT/EP2009/050170
Other languages
French (fr)
Inventor
Eberhard Spanuth
Original Assignee
Roche Diagnostics Gmbh
F. Hoffmann-La Roche Ag
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 Roche Diagnostics Gmbh, F. Hoffmann-La Roche Ag filed Critical Roche Diagnostics Gmbh
Priority to EP09700201A priority Critical patent/EP2227696A1/en
Priority to JP2010541072A priority patent/JP2011509403A/en
Publication of WO2009087190A1 publication Critical patent/WO2009087190A1/en
Priority to US12/822,690 priority patent/US20100261284A1/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
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5302Apparatus specially adapted for immunological test procedures
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/324Coronary artery diseases, e.g. angina pectoris, myocardial infarction

Definitions

  • the present invention relates to a method of risk stratification in a subject.
  • the method according to the present invention permits identifying if a subject is to be admitted to the hospital or intensive care unit or can be discharged to home. In most cases, the subject presents to the emergency unit.
  • the method is based on the determination of Growth-Differentiation Factor- 15 (GDF- 15) in a sample of the said subject.
  • GDF- 15 Growth-Differentiation Factor- 15
  • devices and kits for carrying out the aforementioned methods are also encompassed by the present invention.
  • Cardiovascular complications particularly heart diseases, are the leading cause of morbidity and mortality in the Western hemisphere. Cardiovascular complications can remain asymptomatic for long periods of time. However, they may have severe consequences once an acute cardiovascular event, such as myocardial infarction, as a cause of the cardiovascular complication occurs. Therefore, guidelines exist for the rapid diagnosis of patients presenting to a physician, generally in an emergency unit, and being suspected of suffering from an acute coronary syndrom ACS (i.e. unstable angina pectoris UAP or myocardial infarction MI), see J.Am.Coll.Cardiol.
  • acute coronary syndrom ACS i.e. unstable angina pectoris UAP or myocardial infarction MI
  • the composition of the subjects presenting to an emergency unit is heterogenous, comprising about 35% of subjects suffering from cardiovascular complications (including both acute, i.e. ischemic complications and non- ischemic complications), 10% of subjects suffering from pulmonary complications and 55 % or 56% subjects suffering from other complications, e.g.
  • so-called molecular markers permit to establish rapid and sufficiently accurate diagnosis of the pathological state of a subject.
  • a prominent example is troponin T and/or troponin I for the diagnosis of MI, as mentioned beforehand, or natriuretic peptides, in particular NT-proBNP for various non-ischemic heart diseases, e.g. heart failure.
  • GDF- 15 has been suggested to be an indicator for cardiovascular complications, too (US2003/0232385; Kempf 2006, Circ Res 98: 351-360).
  • Growth-differentiation factor- 15 (GDF- 15) is a member of the transforming growth factor- ⁇ cytokine superfamily. GDF- 15 was first identified as macrophage-inhibitory cytokine- 1 (MIC-I), and later also named placental transforming growth factor- ⁇ (Bootcov 1997, Proc Natl Acad Sci 94:11514- 11519; Tan 2000, Proc Natl Acad Sci 97: 109-114).
  • the invention pertains to a method for predicting the risk of mortality or a further acute cardiovascular event for a subject suffering from a cardiovascular complication based on the determination of GDF- 15 and a natriuretic peptide and/or a cardiac Troponin in a sample the said subject.
  • the technical problem underlying the present invention can be seen as the provision of means and methods for complying with the aforementioned needs.
  • the present invention relates to a method of identifying if a subject is to be admitted to the hospital, the method comprising
  • a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount, whereby a subject to be admitted to the hospital is to be identified.
  • the subject presents to a physician, often a general practitioner.
  • the subject is a subject presenting to an emergency unit, preferably in a hospital, even more preferably an internistic emergency unit.
  • the term "emergency unit” refers to any location where individuals feeling uncomfortable present, in order to consult a person having a medical background, preferably a physician, to have an analysis of their pathological state and the cause underlying their discomfort. Typical examples are emergency departments or emergency rooms in hospitals, emergency ambulances, doctor's offices and other institutions suitable for treatement of critical ill patients.
  • hospital refers to any location where individuals which have a disease or are suspected of having a disease are taken up or kept, in order to be further diagnosed, watched, treated or taken care of. "Hospital” thus refers to hospitals as such, and includes any units therein, including normal care units.
  • the hospital unit is an intensive care unit. Therefore, in one embodiment, the method according to the present invention allows to identify subjects which are to be admitted to the intensive care unit, as a consequence of their physiological condition requiring more than a normal care unit
  • the method of the present invention preferably, is an in vitro method. Moreover, it may comprise steps in addition to those explicitly mentioned above. For example, further steps may relate to sample pre-treatments or evaluation of the results obtained by the method.
  • the method may be carried out manually or assisted by automation.
  • step (a) and/or (b) may in total or in part be assisted by automation, e.g., by a suitable robotic and sensory equipment for the determination in step (a) or a computer-implemented comparison in step (b).
  • identifying means assessing whether a subject is in a pathological state necessitating admission to the hospital for further examination, and/or intensive care treatment, or intervention. As will be understood by those skilled in the art, such an assessment is usually not intended to be correct for all (i.e. 100%) of the subjects to be identified. The term, however, requires that a statistically significant portion of subjects can be identified (e.g. a cohort in a cohort study). Whether a portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p- value determination, Student's t-test, Mann- Whitney test etc..
  • Preferred confidence intervals are at least 90%, at least 95%, at least 97%, at least 98% or at least 99 %.
  • the p-values are, preferably, 0.1, 0.05, 0.01, 0.005, or 0.0001. More preferably, at least
  • the term "subject" as used herein relates to animals, preferably mammals, and, more preferably, humans.
  • the subject suffers from discomfort and/or a pathological state unknown to him and which he cannot diagnose on his own, and feels that a physician should be consulted.
  • the subject is an average subject, i.e. belonging to the average subject population as disclosed beforehand (about 35 % suffering from cardiovascular complications, 10 % suffering from pulmonary complications and 55 % or 56 % suffering from other complications, e.g. tumors, and may additionally suffer from cardiovascular complications).
  • the subject shall not be suffering from a complication which can be diagnosed by following a standard diagnosis protocol, preferably an acute cardiovascular event as defined by the American College of Cardiology (see above), e.g. chest discomfort, dyspnoea, ECG changes and others as described above. More preferably, the subject shall not exhibit one or more episodes of angina lasting at least 5 min within the preceding 24 h, and not have either a positive cardiac troponin T or I test or at least 0-5 mm of transient or persistent ST-segment depression not known to be preexisting and not attributable to coexisting disorders.
  • a standard diagnosis protocol preferably an acute cardiovascular event as defined by the American College of Cardiology (see above), e.g. chest discomfort, dyspnoea, ECG changes and others as described above. More preferably, the subject shall not exhibit one or more episodes of angina lasting at least 5 min within the preceding 24 h, and not have either a positive cardiac troponin T or I test or at least 0-5 mm of transient
  • the subject shall not exhibit symptoms of ischaemia that were increasing or occurring at rest, or that warranted the suspicion of acute myocardial infarction, with the last episode within the preceding 48 h.
  • Acute cardiovascular events are, preferably, acute coronary syndromes (ACS).
  • ACS patients can show unstable angina pectoris (UAP) or myocardial infarction (MI).
  • MI can be an ST-elevation MI (STEMI) or a non-ST-elevated MI (NSTEMI).
  • ST-elevation MI ST-elevation MI
  • NSTEMI non-ST-elevated MI
  • the occurring of an ACS can be followed by a left ventricular dysfunction (LVD) and symptoms of heart failure.
  • LDD left ventricular dysfunction
  • the method of the present invention can also be described as being a method of diagnosing or assessing if an individual having a disease or being suspected of having a disease has to be further diagnosed, watched, treated or taken care of, preferably in a hospital.
  • sample refers to a sample of a body fluid, to a sample of separated cells or to a sample from a tissue or an organ.
  • Samples of body fluids can be obtained by well known techniques and include, preferably, samples of blood, plasma, serum, or urine, more preferably, samples of blood, plasma or serum.
  • Tissue or organ samples may be obtained from any tissue or organ by, e.g., biopsy.
  • Separated cells may be obtained from the body fluids or the tissues or organs by separating techniques such as centrifugation or cell sorting.
  • cell-, tissue- or organ samples are obtained from those cells, tissues or organs which express or produce the peptides referred to herein.
  • GDF- 15 relates to a polypeptide being a member of the transforming growth factor (TGF)- ⁇ cytokine superfamily.
  • TGF transforming growth factor
  • GDF- 15 was originally cloned as macrophage-inhibitory cytokine- 1 and later also identified as placental transforming growth factor- ⁇ , placental bone morphogenetic protein, non-steroidal anti- inflammatory drug-activated gene-1, and prostate-derived factor (Bootcov loc cit; Hromas, 1997 Biochim Biophys Acta 1354:40-44; Lawton 1997, Gene 203:17-26; Yokoyama-Kobayashi 1997, J Biochem (Tokyo), 122:622-626; Paralkar 1998, J Biol Chem 273 : 13760-13767).
  • GDF- 15 is synthesized as an inactive precursor protein, which undergoes disul fide-linked homodimerization. Upon proteolytic cleavage of the N-terminal pro-peptide, GDF- 15 is secreted as a ⁇ 28 kDa dimeric protein (Bauskin 2000, Embo J 19:2212-2220). Amino acid sequences for GDF-15 are disclosed in WO99/06445, WO00/70051, WO2005/113585, Bottner 1999, Gene 237: 105-111, Bootcov loc. cit, Tan loc.
  • GDF-15 as used herein encompasses also variants of the aforementioned specific GDF-15 polypeptides. Such variants have at least the same essential biological and immunological properties as the specific GDF-15 polypeptides. In particular, they share the same essential biological and immunological properties if they are detectable by the same specific assays referred to in this specification, e.g., by ELISA assays using polyclonal or monoclonal antibodies specifically recognizing the said GDF-15 polypeptides.
  • a variant as referred to in accordance with the present invention shall have an amino acid sequence which differs due to at least one amino acid substitution, deletion and/or addition wherein the amino acid sequence of the variant is still, preferably, at least 50%, 60%, 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% identical with the amino sequence of the specific GDF-15 polypeptides.
  • the degree of identity between two amino acid sequences can be determined by algorithms well known in the art.
  • the degree of identity is to be determined by comparing two optimally aligned sequences over a comparison window, where the fragment of amino acid sequence in the comparison window may comprise additions or deletions (e.g., gaps or overhangs) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment.
  • the percentage is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman Add. APL. Math.
  • GAP Garnier et al. (1981), by the homology alignment algorithm of Needleman and Wunsch J. MoI. Biol. 48:443 (1970), by the search for similarity method of Pearson and Lipman Proc. Natl. Acad Sci. (USA) 85: 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, PASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, WI), or by visual inspection. Given that two sequences have been identified for comparison, GAP and BESTFIT are preferably employed to determine their optimal alignment and, thus, the degree of identity. Preferably, the default values of 5.00 for gap weight and 0.30 for gap weight length are used.
  • variants referred to above may be allelic variants or any other species specific homo logs, paralogs, or orthologs.
  • variants referred to herein include fragments of the specific GDF- 15 polypeptides or the aforementioned types of variants as long as these fragments have the essential immunological and biological properties as referred to above. Such fragments may be, e.g., degradation products of the GDF- 15 polypeptides. Further included are variants which differ due to posttranslational modifications such as phosphorylation or myristylation.
  • Determining the amount of GDF- 15 or any other peptide or polypeptide referred to in this specification relates to measuring the amount or concentration, preferably semi- quantitatively or quantitatively. Measuring can be done directly or indirectly.
  • Direct measuring relates to measuring the amount or concentration of the peptide or polypeptide based on a signal which is obtained from the peptide or polypeptide itself and the intensity of which directly correlates with the number of molecules of the peptide present in the sample.
  • a signal sometimes referred to herein as intensity signal -may be obtained, e.g., by measuring an intensity value of a specific physical or chemical property of the peptide or polypeptide.
  • Indirect measuring includes measuring of a signal obtained from a secondary component (i.e.
  • determining the amount of a peptide or polypeptide can be achieved by all known means for determining the amount of a peptide in a sample.
  • Said means comprise immunoassay devices and methods which may utilize labeled molecules in various sandwich, competition, or other assay formats.
  • Said assays will develop a signal which is indicative for the presence or absence of the peptide or polypeptide.
  • the signal strength can, preferably, be correlated directly or indirectly (e.g.
  • suitable methods comprise measuring a physical or chemical property specific for the peptide or polypeptide such as its precise molecular mass or NMR spectrum.
  • Said methods comprise, preferably, biosensors, optical devices coupled to immunoassays, biochips, analytical devices such as mass- spectrometers, NMR- analyzers, or chromatography devices.
  • methods include micro-plate ELISA-based methods, fully-automated or robotic immunoassays (available for example on ElecsysTM analyzers), CBA (an enzymatic Cobalt Binding Assay, available for example on Roche-HitachiTM analyzers), and latex agglutination assays (available for example on Roche-HitachiTM analyzers).
  • determining the amount of a peptide or polypeptide comprises the steps of (a) contacting a cell capable of eliciting a cellular response the intensity of which is indicative of the amount of the peptide or polypeptide with the said peptide or polypeptide for an adequate period of time, (b) measuring the cellular response.
  • the sample or processed sample is, preferably, added to a cell culture and an internal or external cellular response is measured.
  • the cellular response may include the measurable expression of a reporter gene or the secretion of a substance, e.g. a peptide, polypeptide, or a small molecule.
  • the expression or substance shall generate an intensity signal which correlates to the amount of the peptide or polypeptide.
  • determining the amount of a peptide or polypeptide comprises the step of measuring a specific intensity signal obtainable from the peptide or polypeptide in the sample.
  • a specific intensity signal may be the signal intensity observed at an m/z variable specific for the peptide or polypeptide observed in mass spectra or a NMR spectrum specific for the peptide or polypeptide.
  • Determining the amount of a peptide or polypeptide may, preferably, comprises the steps of (a) contacting the peptide with a specific ligand, (b) (optionally) removing non-bound ligand, (c) measuring the amount of bound ligand.
  • the bound ligand will generate an intensity signal.
  • Binding according to the present invention includes both covalent and non-covalent binding.
  • a ligand according to the present invention can be any compound, e.g., a peptide, polypeptide, nucleic acid, or small molecule, binding to the peptide or polypeptide described herein.
  • Preferred ligands include antibodies, nucleic acids, peptides or polypeptides such as receptors or binding partners for the peptide or polypeptide and fragments thereof comprising the binding domains for the peptides, and aptamers, e.g. nucleic acid or peptide aptamers.
  • Methods to prepare such ligands are well-known in the art. For example, identification and production of suitable antibodies or aptamers is also offered by commercial suppliers. The person skilled in the art is familiar with methods to develop derivatives of such ligands with higher affinity or specificity. For example, random mutations can be introduced into the nucleic acids, peptides or polypeptides.
  • Antibodies as referred to herein include both polyclonal and monoclonal antibodies, as well as fragments thereof, such as Fv, Fab and F(ab) 2 fragments that are capable of binding antigen or hapten.
  • the present invention also includes single chain antibodies and humanized hybrid antibodies wherein amino acid sequences of a non- human donor antibody exhibiting a desired antigen-specificity are combined with sequences of a human acceptor antibody.
  • the donor sequences will usually include at least the antigen-binding amino acid residues of the donor but may comprise other structurally and/or functionally relevant amino acid residues of the donor antibody as well.
  • the ligand or agent binds specifically to the peptide or polypeptide.
  • Specific binding according to the present invention means that the ligand or agent should not bind substantially to ("cross- react" with) another peptide, polypeptide or substance present in the sample to be analyzed.
  • the specifically bound peptide or polypeptide should be bound with at least 3 times higher, more preferably at least 10 times higher and even more preferably at least 50 times higher affinity than any other relevant peptide or polypeptide.
  • Nonspecific binding may be tolerable, if it can still be distinguished and measured unequivocally, e.g. according to its size on a Western Blot, or by its relatively higher abundance in the sample. Binding of the ligand can be measured by any method known in the art. Preferably, said method is semi-quantitative or quantitative. Suitable methods are described in the following.
  • binding of a ligand may be measured directly, e.g. by NMR or surface plasmon resonance.
  • an enzymatic reaction product may be measured (e.g. the amount of a protease can be measured by measuring the amount of cleaved substrate, e.g. on a Western Blot).
  • the ligand may exhibit enzymatic properties itself and the "ligand/peptide or polypeptide" complex or the ligand which was bound by the peptide or polypeptide, respectively, may be contacted with a suitable substrate allowing detection by the generation of an intensity signal.
  • the amount of substrate is saturating.
  • the substrate may also be labeled with a detectable lable prior to the reaction.
  • the sample is contacted with the substrate for an adequate period of time.
  • An adequate period of time refers to the time necessary for an detectable, preferably measurable, amount of product to be produced. Instead of measuring the amount of product, the time necessary for appearance of a given (e.g. detectable) amount of product can be measured.
  • the ligand may be coupled covalently or non-covalently to a label allowing detection and measurement of the ligand.
  • Labeling may be done by direct or indirect methods. Direct labeling involves coupling of the label directly (covalently or non-covalently) to the ligand. Indirect labeling involves binding (covalently or non-covalently) of a secondary ligand to the first ligand. The secondary ligand should specifically bind to the first ligand. Said secondary ligand may be coupled with a suitable label and/or be the target (receptor) of tertiary ligand binding to the secondary ligand. The use of secondary, tertiary or even higher order ligands is often used to increase the signal.
  • Suitable secondary and higher order ligands may include antibodies, secondary antibodies, and the well-known streptavidin-biotin system (Vector Laboratories, Inc.).
  • the ligand or substrate may also be "tagged" with one or more tags as known in the art. Such tags may then be targets for higher order ligands.
  • Suitable tags include biotin, digoxygenin, His-Tag, Glutathion-S- Transferase, FLAG, GFP, myc-tag, influenza A virus haemagglutinin (HA), maltose binding protein, and the like.
  • the tag is preferably at the N-terminus and/or C-terminus.
  • Suitable labels are any labels detectable by an appropriate detection method.
  • Typical labels include gold particles, latex beads, acridan ester, luminol, ruthenium, enzymatically active labels, radioactive labels, magnetic labels ("e.g. magnetic beads", including paramagnetic and superparamagnetic labels), and fluorescent labels.
  • Enzymatically active labels include e.g. horseradish peroxidase, alkaline phosphatase, beta-Galactosidase, Luciferase, and derivatives thereof.
  • Suitable substrates for detection include di-amino-benzidine (DAB), 3,3'-5,5'-tetramethylbenzidine, NBT- BCIP (4-nitro blue tetrazolium chloride and 5-bromo-4-chloro-3-indolyl-phosphate, available as ready-made stock solution from Roche Diagnostics), CDP-StarTM (Amersham Biosciences), ECFTM (Amersham Biosciences).
  • a suitable enzyme-substrate combination may result in a colored reaction product, fluorescence or chemoluminescence, which can be measured according to methods known in the art (e.g. using a light-sensitive film or a suitable camera system). As for measuring the enyzmatic reaction, the criteria given above apply analogously.
  • Typical fluorescent labels include fluorescent proteins (such as GFP and its derivatives), Cy3, Cy5, Texas Red, Fluorescein, and the Alexa dyes (e.g. Alexa 568). Further fluorescent labels are available e.g. from Molecular Probes (Oregon). Also the use of quantum dots as fluorescent labels is contemplated.
  • Typical radioactive labels include 35 S, 125 I, 32 P, 33 P and the like. A radioactive label can be detected by any method known and appropriate, e.g. a light-sensitive film or a phosphor imager.
  • Suitable measurement methods according the present invention also include precipitation (particularly immunoprecipitation), electrochemiluminescence (electro-generated chemiluminescence), RIA (radioimmunoassay), ELISA (enzyme- linked immunosorbent assay), sandwich enzyme immune tests, electrochemiluminescence sandwich immunoassays (ECLIA), dissociation-enhanced lanthanide fluoro immuno assay (DELFIA), scintillation proximity assay (SPA), turbidimetry, nephelometry, latex- enhanced turbidimetry or nephelometry, or solid phase immune tests.
  • the amount of a peptide or polypeptide may be, also preferably, determined as follows: (a) contacting a solid support comprising a ligand for the peptide or polypeptide as specified above with a sample comprising the peptide or polypeptide and (b) measuring the amount peptide or polypeptide which is bound to the support.
  • the ligand preferably chosen from the group consisting of nucleic acids, peptides, polypeptides, antibodies and aptamers, is preferably present on a solid support in immobilized form.
  • Materials for manufacturing solid supports include, inter alia, commercially available column materials, polystyrene beads, latex beads, magnetic beads, colloid metal particles, glass and/or silicon chips and surfaces, nitrocellulose strips, membranes, sheets, duracytes, wells and walls of reaction trays, plastic tubes etc.
  • the ligand or agent may be bound to many different carriers. Examples of well-known carriers include glass, polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amyloses, natural and modified celluloses, polyacrylamides, agaroses, and magnetite.
  • the nature of the carrier can be either soluble or insoluble for the purposes of the invention.
  • Suitable methods for fixing/immobilizing said ligand are well known and include, but are not limited to ionic, hydrophobic, covalent interactions and the like. It is also contemplated to use "suspension arrays" as arrays according to the present invention (Nolan 2002, Trends Biotechnol. 20(l):9-12).
  • the carrier e.g. a microbead or microsphere
  • the array consists of different microbeads or microspheres, possibly labeled, carrying different ligands.
  • Methods of producing such arrays for example based on solid-phase chemistry and photo-labile protective groups, are generally known (US 5,744,305).
  • amount encompasses the absolute amount of a polypeptide or peptide, the relative amount or concentration of the said polypeptide or peptide as well as any value or parameter which correlates thereto or can be derived therefrom.
  • values or parameters comprise intensity signal values from all specific physical or chemical properties obtained from the said peptides by direct measurements, e.g., intensity values in mass spectra or NMR spectra.
  • values or parameters which are obtained by indirect measurements specified elsewhere in this description e.g., response levels determined from biological read out systems in response to the peptides or intensity signals obtained from specifically bound ligands. It is to be understood that values correlating to the aforementioned amounts or parameters can also be obtained by all standard mathematical operations.
  • comparing encompasses comparing the amount of the peptide or polypeptide comprised by the sample to be analyzed with an amount of a suitable reference source specified elsewhere in this description. It is to be understood that comparing as used herein refers to a comparison of corresponding parameters or values, e.g., an absolute amount is compared to an absolute reference amount while a concentration is compared to a reference concentration or an intensity signal obtained from a test sample is compared to the same type of intensity signal of a reference sample.
  • the comparison referred to in step (b) of the method of the present invention may be carried out manually or computer assisted. For a computer assisted comparison, the value of the determined amount may be compared to values corresponding to suitable references which are stored in a database by a computer program.
  • the computer program may further evaluate the result of the comparison, i.e. automatically provide the desired assessment in a suitable output format. Based on the comparison of the amount determined in step a) and the reference amount, it is possible to assess whether a subject is susceptible for a cardiac intervention, i.e. belonging to the group of subjects which can be successfully treated by the cardiac intervention. Therefore, the reference amount is to be chosen so that either a difference or a similarity in the compared amounts allows identifying those the test subject which belong into the group of subjects susceptible for cardiac intervention or identifying those test subjects which are not susceptible for a cardiac intervention.
  • the term "reference amount” as used herein refers to an amount which allows assessing whether a subject is to be admitted to the hospital or can be discharged to home. Accordingly, the reference may e.g. be derived from (i) a subject known to have been successfully admitted to the hospital, i.e. who has been subject to further examination and subsequent or intensive care treatment based on the results of the further investigation without the occurrence of adverse effects such as mortality or side effects caused by unadapted treatment regimen, or (ii) a subject known to have not been admitted to the hospital and which died or developed side effects caused by unadapted treatment regimen.
  • the reference amount may define a threshold amount, whereby an amount larger than the threshold shall be indicative for a subject which should be admitted to the hospital for further examination and/or (intensive) treatment, while an amount lower than the threshold amount shall be an indicator for a subject which can be discharged to home.
  • the reference amount applicable for an individual subject may vary depending on various physiological parameters such as age, gender, or subpopulation, as well as on the means used for the determination of the polypeptide or peptide referred to herein.
  • a suitable reference amount may be determined from a reference sample to be analyzed together, i.e. simultaneously or subsequently, with the test sample.
  • a preferred reference amount serving as a threshold may be derived from the upper limit of normal (ULN), i.e.
  • the ULN for a given population of subjects can be determined by various well known techniques. A suitable technique may be to determine the median of the population for the peptide or polypeptide amounts to be determined in the method of the present invention.
  • a preferred threshold (i.e. reference amount) for GDF- 15 is at least one to two times the ULN.
  • the ULN referred to in this context is, preferably, 1800 pg/ml
  • the reference amount defining a threshold amount for GDF- 15 as referred to in accordance with the present invention is 1200 pg/ml, preferably 1560 pg/ml and, most preferably, 8210 pg/ml (representing a negative predictive value of 99,3%) for admission to hospital.
  • An amount of GDF- 15 larger than the reference amount is, more preferably, indicative for a subject which should be admitted to hospital.
  • the above method of the present invention may be used to identify a subject susceptible to intensive care treatment in an intensive care unit.
  • the intensive care treatment may comprise additional diagnostic procedures and therapeutic interventions.
  • a therapeutic intervention may be a drug based therapy or comprises all kinds of surgical interventions.
  • Additional diagnostic procedures include: transthoracal echocardiography; transesophageal echocardiography; abdominal sonographie; CT (thorax); x-ray (thorax); spiral CT; invasive cardiac catherization diagnostic (left, right, combined); lung scintigraphic (inhalation and perfusion); compression sonographie of legs; stress echocardiography; bronchoscopy; phlebography; angiography.
  • Drugs which may be administered include: oral anticoagulants, unfractionated heparins and other antithrombins and fibrinolytic agents; ASS; clopidrogel; loop diuretics and other diuretics; beta blockers; ACE inhibitors; AT blockers; digitalis; calcium antagonists; nitrates; steroids (oral and inhalation); theophyllin; beta sympathomimetics and other bronchodilators; opiates; antibiotics.
  • the present invention further relates to a method of deciding about admitting a subject to the hospital, the method comprising
  • the subject is a subject presenting to the emergency unit.
  • the subject may be admitted to the intensive care unit.
  • An interventional therapy as referred to herein is a therapy which is based on physical interventions with the subject, e.g., by surgery.
  • the subject in this embodiment of the invention is the same subject as defined in the earlier embodiments, in particular a subject not suffering from an acute cardiovascular event.
  • the present invention furthermore, relates to a method for predicting the risk of mortality for a subject comprising
  • a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount; and c) predicting the risk of mortality based on the result of steps a) and b).
  • the subject is a subject presenting to the emergency unit.
  • the subject may be admitted to the intensive care unit.
  • the subject in this embodiment of the invention is the same subject as defined in the earlier embodiments, in particular a subject not suffering from an acute cardiovascular event.
  • the term "predicting" used herein refers to assessing the probability according to which a subject suffering from a disease will die within a defined time window (predictive window) in the future.
  • the predictive window is an interval in which the subject will die according to the predicted probability.
  • the predictive window may be the entire remaining lifespan of the subject upon analysis by the method of the present invention.
  • the predictive window is an interval of one month, six months or one, two, three, four, five or ten years after appearance of the cardiovascular complication (more preferably and precisely, after the sample to be analyzed by the method of the present invention has been obtained).
  • an assessment is usually not intended to be correct for 100% of the subjects to be analyzed.
  • the probability envisaged by the present invention allows that the prediction will be correct for at least 60%, at least 70%, at least 80%, or at least 90% of the subjects of a given cohort.
  • the term "mortality” as used herein relates to mortality which is caused by.cardiovascular complications, lung diseases, lung embolism, thrombosis, thromboembolic complications, stroke, malignant diseases, sepsis, septic shock, bleeding disorders, organ failure, acute kidney disease, and others.
  • a chronic disorder of the cardiovascular system as used herein encompasses coronary heart diseases, stable angina pectoris (SAP) or heart failure, preferably chronic heart failure .
  • Acute cardiovascular events are, preferably, acute coronary syndromes (ACS).
  • ACS patients can show unstable angina pectoris (UAP) or myocardial infarction (MI).
  • MI can be an ST- elevation MI (STEMI) or a non-ST-elevation MI (NSTEMI).
  • NSTE-ACS as used herein encompasses UAP and NSTEMI. The occurring of an MI can be followed by a left ventricular dysfunction (LVD) or development of heart failure.
  • Further preferred cardiovascular complications encompass cardiac brady- or tachyarrhythmias including sudden cardiac death and stroke (cerebrovascular events or accidents).
  • the said cardiovascular complication is ACS or heart failure.
  • predicting the risk of mortality means that it the subject to be analyzed by the method of the present invention is allocated either into the group of subjects of a population having a normal, i.e. non-elevated, risk for mortality or into a group of subjects having a significantly elevated risk.
  • An elevated risk as referred to in accordance with the present invention means that the risk of mortality within a predetermined predictive window is elevated significantly for a subject with respect to the average risk for mortality in a population of subjects.
  • the average risk is within the range of 0.5 and 3.0 %, preferably, 1.5%.
  • An elevated risk as used herein preferably, relates to a risk of more than 3.0 %, preferably, more than 5.0 %, and, most preferably within 3.0 % and 8.0 % with respect to a predictive window of one year.
  • a device for identifying a subject to be admitted to the hospital adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the subject and means for comparing said amount to a reference amount, whereby a subject to be admitted to the hospital is identified.
  • the subject is a subject presenting to the emergency unit. In the hospital, the subject may be admitted to the intensive care unit.
  • the term "device” as used herein relates to a system of means comprising at least the aforementioned means operatively linked to each other as to allow the prediction.
  • Preferred means for determining the amount of GDF- 15 and means for carrying out the comparison are disclosed above in connection with the method of the invention. How to link the means in an operating manner will depend on the type of means included into the device. For example, where means for automatically determining the amount of the peptides are applied, the data obtained by said automatically operating means can be processed by, e.g., a computer program in order to obtain the desired results.
  • the means are comprised by a single device in such a case.
  • Said device may accordingly include an analyzing unit for the measurement of the amount of the peptides or polypeptides in an applied sample and a computer unit for processing the resulting data for the evaluation.
  • the means for comparison may comprise control stripes or tables allocating the determined amount to a reference amount.
  • the test stripes are, preferably, coupled to a ligand which specifically binds to the peptides or polypeptides referred to herein.
  • the strip or device preferably, comprises means for detection of the binding of said peptides or polypeptides to the said ligand. Preferred means for detection are disclosed in connection with embodiments relating to the method of the invention above.
  • the means are operatively linked in that the user of the system brings together the result of the determination of the amount and the diagnostic or prognostic value thereof due to the instructions and interpretations given in a manual.
  • the means may appear as separate devices in such an embodiment and are, preferably, packaged together as a kit.
  • Preferred devices are those which can be applied without the particular knowledge of a specialized clinician, e.g., test stripes or electronic devices which merely require loading with a sample.
  • the results may be given as output of raw data which need interpretation by the clinician.
  • the output of the device is, however, processed, i.e. evaluated, raw data the interpretation of which does not require a clinician.
  • Further preferred devices comprise the analyzing units/devices (e.g., biosensors, arrays, solid supports coupled to ligands specifically recognizing the natriuretic peptide, Plasmon surface resonace devices, NMR spectrometers, mass- spectrometers etc.) or evaluation units/devices referred to above in accordance with the method of the invention.
  • the present invention also relates to a device for predicting the risk of mortality or a further acute cardiovascular event for a subject adapted to carry out the method of the present invention comprising means for determining the amount of GDF- 15 and means for comparing said amounts to reference amounts, whereby it is predicted whether a subject is at risk of mortality or a further acute cardiovascular event.
  • a device for deciding about admitting a subject to the hospital adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the said subject and means for comparing said amount to a reference amount, whereby it is decided whether the subject is to be admitted to the hospital.
  • the present invention also relates to a device for predicting the risk of mortality in a subject, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF-15 in a sample of the subject and means for comparing said amounts to reference amounts, whereby it is predicted whether the subject is at risk of mortality.
  • the subject is a subject presenting to the emergency unit.
  • the subject may be admitted to the intensive care unit.
  • kits for carrying out the methods of the present invention for identifying a subject to be admitted to the hospital, deciding about admitting a subject to the hospital, or predicting the risk of mortality in a subject is envisaged by the present invention.
  • Said kit comprising means for determining the amount of GDF-15 in a sample of a subject and means for comparing said amounts to reference amounts, wherein a subject to be admitted to the hospital is identified, a decision about admitting the subject to the hospital or intensive care unit is made, or the risk of mortality in the subject is predicted.
  • kit refers to a collection of the aforementioned means, preferably, provided in separately or within a single container.
  • the container also preferably, comprises instructions for carrying out the method of the present invention.
  • GDF- 15 improves decision making in the emergency room and the overall management of patients presenting to emergency departments (EDs) by evaluating for incremental diagnostic and prognostic value of GDF- 15, and prospective Iy examining the clinical impact of GDF- 15 guided decision making regarding discharge, admission to hospital, and intensive care treatment.
  • GDF-15 was determined in 302 unselected consecutive patients.
  • Cut-off thresholds of GDF-15 suitable for risk stratification and medical decision making were calculated using ROC analysis.
  • Nonischemic heart disease including arrhythmias without acute ischemic trigger, vascular heart disease, and cardiomyopathies: 36 (11,8%)
  • Lung disorders including asthma, chronic obstructive disease, and pulmonary embolism: 41 (21,8%) 4.
  • the quartile ranges are displayed in Table 3 and Figure 2 showing that patients with increasing GDF- 15 values were admitted to the hospital.
  • Patients in the lrst quartile revealed an admission/discharge rate of 0,43 compared with an admission/discharge rate of 4,85 of patients in the 4 th quartile.
  • results of ROC analysis for prediction discharge or admission by using GDF- 15 values are shown in Table 4.
  • the area under the curve (AUC) of 0,722 demonstrates the high discrimination power of GDF- 15 between discharge and admission.
  • the GDF- 15 cut off value with optimal sensitivity/negative predictive value (66,7%) and optimal specifity/ positive predictive value (68,6%) was estimated as 1,56 ng/ml.
  • GDF- 15 "rule in" cut off value of 8,21 ng/ml could be obtained from the ROC analysis.
  • the corresponding sensitivity and negative predictive value is 99,3% and 95,2%, respectively.
  • Patients with GDF- 15 values > 8,21 ng/ml should be admitted to hospital (and not be discharged) independent of diagnosis and underlying disease.
  • GDF- 15 is a marker for decision making and a suitable tool for risk stratification of patients presenting at the emergency department. Moreover, by using GDF- 15 "rule in" cut off value patients could be identified who should be admitted to hospital and not be discharged independent of diagnosis and disease status of the patient. A 58 years old female was admitted to the emergency room with suspected gastrointestinal bleeding or acute gastritis. Case history:
  • GDF-15 value 1,56 ng/ml; ⁇ 8,21 ng/ml (rule in cut off value for admission to hospital);
  • the patient could be discharged to home!
  • GDF-15 value 20,62 ng/ml; > 8,21 ng/ml (rule in cut off value for admission to hospital).
  • the patient was admitted to the intensive care unit.
  • GDF-15 value 0,53 ng/ml; ⁇ 8,21 ng/ml (rule in cut off value for admission to hospital); ⁇ 1,66 ng/ml (optimized ROC cut off value)
  • GDF-15 value 0,91 ng/ml; ⁇ 8,21 ng/ml (rule in cut off value for admission to hospital); ⁇ 1,66 ng/ml (optimized ROC cut off value)
  • GDF-15 value 12,78 ng/ml; > 8,21 ng/ml (rule in cut off value for admission to hospital).
  • the patient was admitted to hospital.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention relates to a method of identifying if a subject is to be admitted to the hospital or intensive care unit, the method comprising a)determining the amount of GDF-15 in a sample of the subject; and b)comparing the amount of GDF-15 determined in step a) to a reference amount, whereby a subject to be admitted to the hospital or intensive care unit is to be identified. Moreover, the present invention pertains to a method for predicting the risk of mortality based on determining the amount of GDF-15 in a subject. Also encompassed by the present invention are devices and kits for carrying out the aforementioned methods.

Description

Means and methods for assessing the risk of patients presenting to emergency units based on GDF- 15
The present invention relates to a method of risk stratification in a subject. Preferably, the method according to the present invention permits identifying if a subject is to be admitted to the hospital or intensive care unit or can be discharged to home. In most cases, the subject presents to the emergency unit. The method is based on the determination of Growth-Differentiation Factor- 15 (GDF- 15) in a sample of the said subject. Also encompassed by the present invention are devices and kits for carrying out the aforementioned methods.
When subjects present to an emergency unit with any kind of discomfort, a rapid diagnosis of the pathological state of the subject is mandatory in order to identify the cause underlying his discomfort and avoid consequences to the subject's health. A highly relevant topic is the decision if the patient will be admitted to the hospital - for further, time consuming analysis and/or intensive care treatment, or if the patient can be discharged to home.
For example, in the case of acute cardiovascular events, a decision for a certain treatment regimen must be made, usually, within a short period of time. Cardiovascular complications, particularly heart diseases, are the leading cause of morbidity and mortality in the Western hemisphere. Cardiovascular complications can remain asymptomatic for long periods of time. However, they may have severe consequences once an acute cardiovascular event, such as myocardial infarction, as a cause of the cardiovascular complication occurs. Therefore, guidelines exist for the rapid diagnosis of patients presenting to a physician, generally in an emergency unit, and being suspected of suffering from an acute coronary syndrom ACS (i.e. unstable angina pectoris UAP or myocardial infarction MI), see J.Am.Coll.Cardiol. 2000; 36, pages 959-969. In a subject suspected to have MI, an electrocardiogram is recorded, and the level of Troponin T or I is determined. Further, it is analysed if the suspected subject shows evident syndroms like chest pain, palpitation, nausea, vomitting and further syndroms known to the person skilled in the art. If the subject is positive in 2 of the 3 criteriae, then he or she is admitted to the hospital for further examination. However, the composition of the subjects presenting to an emergency unit is heterogenous, comprising about 35% of subjects suffering from cardiovascular complications (including both acute, i.e. ischemic complications and non- ischemic complications), 10% of subjects suffering from pulmonary complications and 55 % or 56% subjects suffering from other complications, e.g. tumors, and which may additionally suffer from cardiovascular complications. The conventional diagnostic techniques, specifically for emergency situations, usually do not allow for a reliable diagnosis and/or risk assessment covering these various pathological states. A further drawback is the often occuring lack of personnel and the varying occupancy in emergency units.
At present, there does not exist a standardized diagnosis procedure covering the various diseases a physician may encounter in an emergency unit. Thus, a rapid and acurate diagnosis allowing a decision if the subject can be discharged to home or has to be admitted to the hospital for further examination or intensive care treatment (which can be live saving) cannot be carried out in the emergency unit with sufficient accuracy. As a consequence thereof, many patients will either be admitted or discharged in to home where the opposite would have been the appropriate measure.
In some cases, so-called molecular markers permit to establish rapid and sufficiently accurate diagnosis of the pathological state of a subject. A prominent example is troponin T and/or troponin I for the diagnosis of MI, as mentioned beforehand, or natriuretic peptides, in particular NT-proBNP for various non-ischemic heart diseases, e.g. heart failure.
Recently, GDF- 15 has been suggested to be an indicator for cardiovascular complications, too (US2003/0232385; Kempf 2006, Circ Res 98: 351-360). Growth-differentiation factor- 15 (GDF- 15) is a member of the transforming growth factor-β cytokine superfamily. GDF- 15 was first identified as macrophage-inhibitory cytokine- 1 (MIC-I), and later also named placental transforming growth factor-β (Bootcov 1997, Proc Natl Acad Sci 94:11514- 11519; Tan 2000, Proc Natl Acad Sci 97: 109-114). It has recently been shown that cultured cardiomyocytes express and secrete GDF-15 via nitric oxide and nitrosative stress- dependent signaling pathways when subjected to simulated ischemia and reperfusion. Moreover, it has been observed in a mouse model of myocardial ischemia and reperfusion injury that GDF- 15 expression levels rapidly increase in the ischemic area following coronary artery ligation, and remain elevated in the reperfused myocardium for several days (Kempf loc. cit.). The application PCT/EP2007/058007 filed August 2, 2007, relates to a method of identifying a subject being susceptible to a cardiac intervention based on the determination of GDF- 15 in a sample of a subject in need of a cardiac intervention. Moreover, the invention pertains to a method for predicting the risk of mortality or a further acute cardiovascular event for a subject suffering from a cardiovascular complication based on the determination of GDF- 15 and a natriuretic peptide and/or a cardiac Troponin in a sample the said subject.
Therefore, there is a need for diagnostic or prognostic measures which allow an assessment and/or an individual risk stratification for a subject presenting to the emergency unit. The measures should permit this assessment/risk strtification also in patients not presenting with cardiovascualr diseases, in particular not with acute cardiovascular diseases.
The technical problem underlying the present invention can be seen as the provision of means and methods for complying with the aforementioned needs.
The technical problem is solved by the embodiments characterized in the claims and herein below.
Accordingly, the present invention relates to a method of identifying if a subject is to be admitted to the hospital, the method comprising
a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount, whereby a subject to be admitted to the hospital is to be identified.
In general, the subject presents to a physician, often a general practitioner. Preferably, the subject is a subject presenting to an emergency unit, preferably in a hospital, even more preferably an internistic emergency unit.
In the context of the present invention, the term "emergency unit" refers to any location where individuals feeling uncomfortable present, in order to consult a person having a medical background, preferably a physician, to have an analysis of their pathological state and the cause underlying their discomfort. Typical examples are emergency departments or emergency rooms in hospitals, emergency ambulances, doctor's offices and other institutions suitable for treatement of critical ill patients. In the context of the present invention, the term "hospital" refers to any location where individuals which have a disease or are suspected of having a disease are taken up or kept, in order to be further diagnosed, watched, treated or taken care of. "Hospital" thus refers to hospitals as such, and includes any units therein, including normal care units. In a preferred embodimant of the present invention, the hospital unit is an intensive care unit. Therefore, in one embodiment, the method according to the present invention allows to identify subjects which are to be admitted to the intensive care unit, as a consequence of their physiological condition requiring more than a normal care unit
The method of the present invention, preferably, is an in vitro method. Moreover, it may comprise steps in addition to those explicitly mentioned above. For example, further steps may relate to sample pre-treatments or evaluation of the results obtained by the method. The method may be carried out manually or assisted by automation. Preferably, step (a) and/or (b) may in total or in part be assisted by automation, e.g., by a suitable robotic and sensory equipment for the determination in step (a) or a computer-implemented comparison in step (b).
The term "identifying" as used herein means assessing whether a subject is in a pathological state necessitating admission to the hospital for further examination, and/or intensive care treatment, or intervention. As will be understood by those skilled in the art, such an assessment is usually not intended to be correct for all (i.e. 100%) of the subjects to be identified. The term, however, requires that a statistically significant portion of subjects can be identified (e.g. a cohort in a cohort study). Whether a portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p- value determination, Student's t-test, Mann- Whitney test etc.. Details are found in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. Preferred confidence intervals are at least 90%, at least 95%, at least 97%, at least 98% or at least 99 %. The p-values are, preferably, 0.1, 0.05, 0.01, 0.005, or 0.0001. More preferably, at least
60%, at least 70%, at least 80% or at least 90% of the subjects of a population can be properly identified by the method of the present invention.
The term "subject" as used herein relates to animals, preferably mammals, and, more preferably, humans. In accordance with the present invention, the subject suffers from discomfort and/or a pathological state unknown to him and which he cannot diagnose on his own, and feels that a physician should be consulted. The subject is an average subject, i.e. belonging to the average subject population as disclosed beforehand (about 35 % suffering from cardiovascular complications, 10 % suffering from pulmonary complications and 55 % or 56 % suffering from other complications, e.g. tumors, and may additionally suffer from cardiovascular complications).
However, it is envisaged in accordance with the aforementioned method of the present invention that the subject shall not be suffering from a complication which can be diagnosed by following a standard diagnosis protocol, preferably an acute cardiovascular event as defined by the American College of Cardiology (see above), e.g. chest discomfort, dyspnoea, ECG changes and others as described above. More preferably, the subject shall not exhibit one or more episodes of angina lasting at least 5 min within the preceding 24 h, and not have either a positive cardiac troponin T or I test or at least 0-5 mm of transient or persistent ST-segment depression not known to be preexisting and not attributable to coexisting disorders. Alternatively, the subject shall not exhibit symptoms of ischaemia that were increasing or occurring at rest, or that warranted the suspicion of acute myocardial infarction, with the last episode within the preceding 48 h. Myocardial ischaemia has to be verified by electrocardiography (ST depression =0.1 mV or T-wave inversion =0-1 mV) or by raised biochemical markers (creatine kinase [CK]-MB >6 ug/L, troponin-T >0.01 ng/ml, qualitative troponin-T test positive, or catalytic activity of CK, CK-B, or CK MB higher than the local diagnostic limit for myocardial infarction).
Acute cardiovascular events are, preferably, acute coronary syndromes (ACS). ACS patients can show unstable angina pectoris (UAP) or myocardial infarction (MI). MI can be an ST-elevation MI (STEMI) or a non-ST-elevated MI (NSTEMI). The occurring of an ACS can be followed by a left ventricular dysfunction (LVD) and symptoms of heart failure.
In accordance with the foregoing, the method of the present invention can also be described as being a method of diagnosing or assessing if an individual having a disease or being suspected of having a disease has to be further diagnosed, watched, treated or taken care of, preferably in a hospital.
The term "sample" refers to a sample of a body fluid, to a sample of separated cells or to a sample from a tissue or an organ. Samples of body fluids can be obtained by well known techniques and include, preferably, samples of blood, plasma, serum, or urine, more preferably, samples of blood, plasma or serum. Tissue or organ samples may be obtained from any tissue or organ by, e.g., biopsy. Separated cells may be obtained from the body fluids or the tissues or organs by separating techniques such as centrifugation or cell sorting. Preferably, cell-, tissue- or organ samples are obtained from those cells, tissues or organs which express or produce the peptides referred to herein.
The term "Growth-Differentiation Factor- 15" or "GDF- 15" relates to a polypeptide being a member of the transforming growth factor (TGF)-β cytokine superfamily. The terms polypeptide, peptide and protein are used interchangeable throughout this specification. GDF- 15 was originally cloned as macrophage-inhibitory cytokine- 1 and later also identified as placental transforming growth factor-β, placental bone morphogenetic protein, non-steroidal anti- inflammatory drug-activated gene-1, and prostate-derived factor (Bootcov loc cit; Hromas, 1997 Biochim Biophys Acta 1354:40-44; Lawton 1997, Gene 203:17-26; Yokoyama-Kobayashi 1997, J Biochem (Tokyo), 122:622-626; Paralkar 1998, J Biol Chem 273 : 13760-13767). Similar to other TGF-β-related cytokines, GDF- 15 is synthesized as an inactive precursor protein, which undergoes disul fide-linked homodimerization. Upon proteolytic cleavage of the N-terminal pro-peptide, GDF- 15 is secreted as a ~28 kDa dimeric protein (Bauskin 2000, Embo J 19:2212-2220). Amino acid sequences for GDF-15 are disclosed in WO99/06445, WO00/70051, WO2005/113585, Bottner 1999, Gene 237: 105-111, Bootcov loc. cit, Tan loc. cit., Baek 2001, MoI Pharmacol 59: 901-908, Hromas loc cit, Paralkar loc cit, Morrish 1996, Placenta 17:431- 441 or Yokoyama-Kobayashi loc cit.. GDF-15 as used herein encompasses also variants of the aforementioned specific GDF-15 polypeptides. Such variants have at least the same essential biological and immunological properties as the specific GDF-15 polypeptides. In particular, they share the same essential biological and immunological properties if they are detectable by the same specific assays referred to in this specification, e.g., by ELISA assays using polyclonal or monoclonal antibodies specifically recognizing the said GDF-15 polypeptides. A preferred assay is described in the accompanying Examples. Moreover, it is to be understood that a variant as referred to in accordance with the present invention shall have an amino acid sequence which differs due to at least one amino acid substitution, deletion and/or addition wherein the amino acid sequence of the variant is still, preferably, at least 50%, 60%, 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% identical with the amino sequence of the specific GDF-15 polypeptides. The degree of identity between two amino acid sequences can be determined by algorithms well known in the art. Preferably, the degree of identity is to be determined by comparing two optimally aligned sequences over a comparison window, where the fragment of amino acid sequence in the comparison window may comprise additions or deletions (e.g., gaps or overhangs) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment. The percentage is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity. Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman Add. APL. Math. 2:482 (1981), by the homology alignment algorithm of Needleman and Wunsch J. MoI. Biol. 48:443 (1970), by the search for similarity method of Pearson and Lipman Proc. Natl. Acad Sci. (USA) 85: 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, PASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, WI), or by visual inspection. Given that two sequences have been identified for comparison, GAP and BESTFIT are preferably employed to determine their optimal alignment and, thus, the degree of identity. Preferably, the default values of 5.00 for gap weight and 0.30 for gap weight length are used. Variants referred to above may be allelic variants or any other species specific homo logs, paralogs, or orthologs. Moreover, the variants referred to herein include fragments of the specific GDF- 15 polypeptides or the aforementioned types of variants as long as these fragments have the essential immunological and biological properties as referred to above. Such fragments may be, e.g., degradation products of the GDF- 15 polypeptides. Further included are variants which differ due to posttranslational modifications such as phosphorylation or myristylation.
Determining the amount of GDF- 15 or any other peptide or polypeptide referred to in this specification relates to measuring the amount or concentration, preferably semi- quantitatively or quantitatively. Measuring can be done directly or indirectly. Direct measuring relates to measuring the amount or concentration of the peptide or polypeptide based on a signal which is obtained from the peptide or polypeptide itself and the intensity of which directly correlates with the number of molecules of the peptide present in the sample. Such a signal - sometimes referred to herein as intensity signal -may be obtained, e.g., by measuring an intensity value of a specific physical or chemical property of the peptide or polypeptide. Indirect measuring includes measuring of a signal obtained from a secondary component (i.e. a component not being the peptide or polypeptide itself) or a biological read out system, e.g., measurable cellular responses, ligands, labels, or enzymatic reaction products. In accordance with the present invention, determining the amount of a peptide or polypeptide can be achieved by all known means for determining the amount of a peptide in a sample. Said means comprise immunoassay devices and methods which may utilize labeled molecules in various sandwich, competition, or other assay formats. Said assays will develop a signal which is indicative for the presence or absence of the peptide or polypeptide. Moreover, the signal strength can, preferably, be correlated directly or indirectly (e.g. reverse- proportional) to the amount of polypeptide present in a sample. Further suitable methods comprise measuring a physical or chemical property specific for the peptide or polypeptide such as its precise molecular mass or NMR spectrum. Said methods comprise, preferably, biosensors, optical devices coupled to immunoassays, biochips, analytical devices such as mass- spectrometers, NMR- analyzers, or chromatography devices. Further, methods include micro-plate ELISA-based methods, fully-automated or robotic immunoassays (available for example on Elecsys™ analyzers), CBA (an enzymatic Cobalt Binding Assay, available for example on Roche-Hitachi™ analyzers), and latex agglutination assays (available for example on Roche-Hitachi™ analyzers).
Preferably, determining the amount of a peptide or polypeptide comprises the steps of (a) contacting a cell capable of eliciting a cellular response the intensity of which is indicative of the amount of the peptide or polypeptide with the said peptide or polypeptide for an adequate period of time, (b) measuring the cellular response. For measuring cellular responses, the sample or processed sample is, preferably, added to a cell culture and an internal or external cellular response is measured. The cellular response may include the measurable expression of a reporter gene or the secretion of a substance, e.g. a peptide, polypeptide, or a small molecule. The expression or substance shall generate an intensity signal which correlates to the amount of the peptide or polypeptide.
Also preferably, determining the amount of a peptide or polypeptide comprises the step of measuring a specific intensity signal obtainable from the peptide or polypeptide in the sample. As described above, such a signal may be the signal intensity observed at an m/z variable specific for the peptide or polypeptide observed in mass spectra or a NMR spectrum specific for the peptide or polypeptide.
Determining the amount of a peptide or polypeptide may, preferably, comprises the steps of (a) contacting the peptide with a specific ligand, (b) (optionally) removing non-bound ligand, (c) measuring the amount of bound ligand. The bound ligand will generate an intensity signal. Binding according to the present invention includes both covalent and non-covalent binding. A ligand according to the present invention can be any compound, e.g., a peptide, polypeptide, nucleic acid, or small molecule, binding to the peptide or polypeptide described herein. Preferred ligands include antibodies, nucleic acids, peptides or polypeptides such as receptors or binding partners for the peptide or polypeptide and fragments thereof comprising the binding domains for the peptides, and aptamers, e.g. nucleic acid or peptide aptamers. Methods to prepare such ligands are well-known in the art. For example, identification and production of suitable antibodies or aptamers is also offered by commercial suppliers. The person skilled in the art is familiar with methods to develop derivatives of such ligands with higher affinity or specificity. For example, random mutations can be introduced into the nucleic acids, peptides or polypeptides. These derivatives can then be tested for binding according to screening procedures known in the art, e.g. phage display. Antibodies as referred to herein include both polyclonal and monoclonal antibodies, as well as fragments thereof, such as Fv, Fab and F(ab)2 fragments that are capable of binding antigen or hapten. The present invention also includes single chain antibodies and humanized hybrid antibodies wherein amino acid sequences of a non- human donor antibody exhibiting a desired antigen-specificity are combined with sequences of a human acceptor antibody. The donor sequences will usually include at least the antigen-binding amino acid residues of the donor but may comprise other structurally and/or functionally relevant amino acid residues of the donor antibody as well. Such hybrids can be prepared by several methods well known in the art. Preferably, the ligand or agent binds specifically to the peptide or polypeptide. Specific binding according to the present invention means that the ligand or agent should not bind substantially to ("cross- react" with) another peptide, polypeptide or substance present in the sample to be analyzed. Preferably, the specifically bound peptide or polypeptide should be bound with at least 3 times higher, more preferably at least 10 times higher and even more preferably at least 50 times higher affinity than any other relevant peptide or polypeptide. Nonspecific binding may be tolerable, if it can still be distinguished and measured unequivocally, e.g. according to its size on a Western Blot, or by its relatively higher abundance in the sample. Binding of the ligand can be measured by any method known in the art. Preferably, said method is semi-quantitative or quantitative. Suitable methods are described in the following.
First, binding of a ligand may be measured directly, e.g. by NMR or surface plasmon resonance. Second, if the ligand also serves as a substrate of an enzymatic activity of the peptide or polypeptide of interest, an enzymatic reaction product may be measured (e.g. the amount of a protease can be measured by measuring the amount of cleaved substrate, e.g. on a Western Blot). Alternatively, the ligand may exhibit enzymatic properties itself and the "ligand/peptide or polypeptide" complex or the ligand which was bound by the peptide or polypeptide, respectively, may be contacted with a suitable substrate allowing detection by the generation of an intensity signal. For measurement of enzymatic reaction products, preferably the amount of substrate is saturating. The substrate may also be labeled with a detectable lable prior to the reaction. Preferably, the sample is contacted with the substrate for an adequate period of time. An adequate period of time refers to the time necessary for an detectable, preferably measurable, amount of product to be produced. Instead of measuring the amount of product, the time necessary for appearance of a given (e.g. detectable) amount of product can be measured.
Third, the ligand may be coupled covalently or non-covalently to a label allowing detection and measurement of the ligand. Labeling may be done by direct or indirect methods. Direct labeling involves coupling of the label directly (covalently or non-covalently) to the ligand. Indirect labeling involves binding (covalently or non-covalently) of a secondary ligand to the first ligand. The secondary ligand should specifically bind to the first ligand. Said secondary ligand may be coupled with a suitable label and/or be the target (receptor) of tertiary ligand binding to the secondary ligand. The use of secondary, tertiary or even higher order ligands is often used to increase the signal. Suitable secondary and higher order ligands may include antibodies, secondary antibodies, and the well-known streptavidin-biotin system (Vector Laboratories, Inc.). The ligand or substrate may also be "tagged" with one or more tags as known in the art. Such tags may then be targets for higher order ligands. Suitable tags include biotin, digoxygenin, His-Tag, Glutathion-S- Transferase, FLAG, GFP, myc-tag, influenza A virus haemagglutinin (HA), maltose binding protein, and the like. In the case of a peptide or polypeptide, the tag is preferably at the N-terminus and/or C-terminus. Suitable labels are any labels detectable by an appropriate detection method. Typical labels include gold particles, latex beads, acridan ester, luminol, ruthenium, enzymatically active labels, radioactive labels, magnetic labels ("e.g. magnetic beads", including paramagnetic and superparamagnetic labels), and fluorescent labels. Enzymatically active labels include e.g. horseradish peroxidase, alkaline phosphatase, beta-Galactosidase, Luciferase, and derivatives thereof. Suitable substrates for detection include di-amino-benzidine (DAB), 3,3'-5,5'-tetramethylbenzidine, NBT- BCIP (4-nitro blue tetrazolium chloride and 5-bromo-4-chloro-3-indolyl-phosphate, available as ready-made stock solution from Roche Diagnostics), CDP-Star™ (Amersham Biosciences), ECF™ (Amersham Biosciences). A suitable enzyme-substrate combination may result in a colored reaction product, fluorescence or chemoluminescence, which can be measured according to methods known in the art (e.g. using a light-sensitive film or a suitable camera system). As for measuring the enyzmatic reaction, the criteria given above apply analogously. Typical fluorescent labels include fluorescent proteins (such as GFP and its derivatives), Cy3, Cy5, Texas Red, Fluorescein, and the Alexa dyes (e.g. Alexa 568). Further fluorescent labels are available e.g. from Molecular Probes (Oregon). Also the use of quantum dots as fluorescent labels is contemplated. Typical radioactive labels include 35S, 125I, 32P, 33P and the like. A radioactive label can be detected by any method known and appropriate, e.g. a light-sensitive film or a phosphor imager. Suitable measurement methods according the present invention also include precipitation (particularly immunoprecipitation), electrochemiluminescence (electro-generated chemiluminescence), RIA (radioimmunoassay), ELISA (enzyme- linked immunosorbent assay), sandwich enzyme immune tests, electrochemiluminescence sandwich immunoassays (ECLIA), dissociation-enhanced lanthanide fluoro immuno assay (DELFIA), scintillation proximity assay (SPA), turbidimetry, nephelometry, latex- enhanced turbidimetry or nephelometry, or solid phase immune tests. Further methods known in the art (such as gel electrophoresis, 2D gel electrophoresis, SDS polyacrylamid gel electrophoresis (SDS-PAGE), Western Blotting, and mass spectrometry), can be used alone or in combination with labeling or other dectection methods as described above.
The amount of a peptide or polypeptide may be, also preferably, determined as follows: (a) contacting a solid support comprising a ligand for the peptide or polypeptide as specified above with a sample comprising the peptide or polypeptide and (b) measuring the amount peptide or polypeptide which is bound to the support. The ligand, preferably chosen from the group consisting of nucleic acids, peptides, polypeptides, antibodies and aptamers, is preferably present on a solid support in immobilized form. Materials for manufacturing solid supports are well known in the art and include, inter alia, commercially available column materials, polystyrene beads, latex beads, magnetic beads, colloid metal particles, glass and/or silicon chips and surfaces, nitrocellulose strips, membranes, sheets, duracytes, wells and walls of reaction trays, plastic tubes etc. The ligand or agent may be bound to many different carriers. Examples of well-known carriers include glass, polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amyloses, natural and modified celluloses, polyacrylamides, agaroses, and magnetite. The nature of the carrier can be either soluble or insoluble for the purposes of the invention. Suitable methods for fixing/immobilizing said ligand are well known and include, but are not limited to ionic, hydrophobic, covalent interactions and the like. It is also contemplated to use "suspension arrays" as arrays according to the present invention (Nolan 2002, Trends Biotechnol. 20(l):9-12). In such suspension arrays, the carrier, e.g. a microbead or microsphere, is present in suspension. The array consists of different microbeads or microspheres, possibly labeled, carrying different ligands. Methods of producing such arrays, for example based on solid-phase chemistry and photo-labile protective groups, are generally known (US 5,744,305).
The term "amount" as used herein encompasses the absolute amount of a polypeptide or peptide, the relative amount or concentration of the said polypeptide or peptide as well as any value or parameter which correlates thereto or can be derived therefrom. Such values or parameters comprise intensity signal values from all specific physical or chemical properties obtained from the said peptides by direct measurements, e.g., intensity values in mass spectra or NMR spectra. Moreover, encompassed are all values or parameters which are obtained by indirect measurements specified elsewhere in this description, e.g., response levels determined from biological read out systems in response to the peptides or intensity signals obtained from specifically bound ligands. It is to be understood that values correlating to the aforementioned amounts or parameters can also be obtained by all standard mathematical operations.
The term "comparing" as used herein encompasses comparing the amount of the peptide or polypeptide comprised by the sample to be analyzed with an amount of a suitable reference source specified elsewhere in this description. It is to be understood that comparing as used herein refers to a comparison of corresponding parameters or values, e.g., an absolute amount is compared to an absolute reference amount while a concentration is compared to a reference concentration or an intensity signal obtained from a test sample is compared to the same type of intensity signal of a reference sample. The comparison referred to in step (b) of the method of the present invention may be carried out manually or computer assisted. For a computer assisted comparison, the value of the determined amount may be compared to values corresponding to suitable references which are stored in a database by a computer program. The computer program may further evaluate the result of the comparison, i.e. automatically provide the desired assessment in a suitable output format. Based on the comparison of the amount determined in step a) and the reference amount, it is possible to assess whether a subject is susceptible for a cardiac intervention, i.e. belonging to the group of subjects which can be successfully treated by the cardiac intervention. Therefore, the reference amount is to be chosen so that either a difference or a similarity in the compared amounts allows identifying those the test subject which belong into the group of subjects susceptible for cardiac intervention or identifying those test subjects which are not susceptible for a cardiac intervention.
Accordingly, the term "reference amount" as used herein refers to an amount which allows assessing whether a subject is to be admitted to the hospital or can be discharged to home. Accordingly, the reference may e.g. be derived from (i) a subject known to have been successfully admitted to the hospital, i.e. who has been subject to further examination and subsequent or intensive care treatment based on the results of the further investigation without the occurrence of adverse effects such as mortality or side effects caused by unadapted treatment regimen, or (ii) a subject known to have not been admitted to the hospital and which died or developed side effects caused by unadapted treatment regimen. Moreover, the reference amount may define a threshold amount, whereby an amount larger than the threshold shall be indicative for a subject which should be admitted to the hospital for further examination and/or (intensive) treatment, while an amount lower than the threshold amount shall be an indicator for a subject which can be discharged to home. The reference amount applicable for an individual subject may vary depending on various physiological parameters such as age, gender, or subpopulation, as well as on the means used for the determination of the polypeptide or peptide referred to herein. A suitable reference amount may be determined from a reference sample to be analyzed together, i.e. simultaneously or subsequently, with the test sample. A preferred reference amount serving as a threshold may be derived from the upper limit of normal (ULN), i.e. the upper limit of the physiological amount to be found in a population of apparently healthy subjects. The ULN for a given population of subjects can be determined by various well known techniques. A suitable technique may be to determine the median of the population for the peptide or polypeptide amounts to be determined in the method of the present invention. A preferred threshold (i.e. reference amount) for GDF- 15 is at least one to two times the ULN. The ULN referred to in this context is, preferably, 1800 pg/ml
Thus, the reference amount defining a threshold amount for GDF- 15 as referred to in accordance with the present invention is 1200 pg/ml, preferably 1560 pg/ml and, most preferably, 8210 pg/ml (representing a negative predictive value of 99,3%) for admission to hospital.
An amount of GDF- 15 larger than the reference amount is, more preferably, indicative for a subject which should be admitted to hospital. In addition or alternatively, the above method of the present invention may be used to identify a subject susceptible to intensive care treatment in an intensive care unit. The intensive care treatment may comprise additional diagnostic procedures and therapeutic interventions. A therapeutic intervention may be a drug based therapy or comprises all kinds of surgical interventions.
Additional diagnostic procedures include: transthoracal echocardiography; transesophageal echocardiography; abdominal sonographie; CT (thorax); x-ray (thorax); spiral CT; invasive cardiac catherization diagnostic (left, right, combined); lung scintigraphic (inhalation and perfusion); compression sonographie of legs; stress echocardiography; bronchoscopy; phlebography; angiography.
Drugs which may be administered include: oral anticoagulants, unfractionated heparins and other antithrombins and fibrinolytic agents; ASS; clopidrogel; loop diuretics and other diuretics; beta blockers; ACE inhibitors; AT blockers; digitalis; calcium antagonists; nitrates; steroids (oral and inhalation); theophyllin; beta sympathomimetics and other bronchodilators; opiates; antibiotics.
It is to be understood that the definitions and explanations of the terms made above and below apply accordingly for all embodiments described in this specification and the accompanying claims.
The present invention further relates to a method of deciding about admitting a subject to the hospital, the method comprising
a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount; and c) deciding whether the subject is to be admitted to the hospital.
Preferably, the subject is a subject presenting to the emergency unit. In the hospital, the subject may be admitted to the intensive care unit.
Another preferred therapy to be selected for a subject in accordance with the present invention is an interventional therapy. An interventional therapy as referred to herein is a therapy which is based on physical interventions with the subject, e.g., by surgery. The subject in this embodiment of the invention is the same subject as defined in the earlier embodiments, in particular a subject not suffering from an acute cardiovascular event.
The present invention, furthermore, relates to a method for predicting the risk of mortality for a subject comprising
a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount; and c) predicting the risk of mortality based on the result of steps a) and b).
Preferably, the subject is a subject presenting to the emergency unit. In the hospital, the subject may be admitted to the intensive care unit.
The subject in this embodiment of the invention is the same subject as defined in the earlier embodiments, in particular a subject not suffering from an acute cardiovascular event.
The term "predicting" used herein refers to assessing the probability according to which a subject suffering from a disease will die within a defined time window (predictive window) in the future. The predictive window is an interval in which the subject will die according to the predicted probability. The predictive window may be the entire remaining lifespan of the subject upon analysis by the method of the present invention. Preferably, however, the predictive window is an interval of one month, six months or one, two, three, four, five or ten years after appearance of the cardiovascular complication (more preferably and precisely, after the sample to be analyzed by the method of the present invention has been obtained). As will be understood by those skilled in the art, such an assessment is usually not intended to be correct for 100% of the subjects to be analyzed. The term, however, requires that the assessment will be valid for a statistically significant portion of the subjects to be analyzed. Whether a portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p-value determination, Student's t-test, Mann- Whitney test, etc.. Details are found in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. Preferred confidence intervals are at least 90%, at least 95%, at least 97%, at least 98% or at least 99 %. The p- values are, preferably, 0.1, 0.05, 0.01, 0.005, or 0.0001. Preferably, the probability envisaged by the present invention allows that the prediction will be correct for at least 60%, at least 70%, at least 80%, or at least 90% of the subjects of a given cohort. The term "mortality" as used herein relates to mortality which is caused by.cardiovascular complications, lung diseases, lung embolism, thrombosis, thromboembolic complications, stroke, malignant diseases, sepsis, septic shock, bleeding disorders, organ failure, acute kidney disease, and others.
The term "cardiovascular complication" as used herein refers to any chronic disorder of the cardiovascular system or any acute cardiovascular event. Preferably, a chronic disorder of the cardiovascular system as used herein encompasses coronary heart diseases, stable angina pectoris (SAP) or heart failure, preferably chronic heart failure . Acute cardiovascular events are, preferably, acute coronary syndromes (ACS). ACS patients can show unstable angina pectoris (UAP) or myocardial infarction (MI). MI can be an ST- elevation MI (STEMI) or a non-ST-elevation MI (NSTEMI). NSTE-ACS as used herein encompasses UAP and NSTEMI. The occurring of an MI can be followed by a left ventricular dysfunction (LVD) or development of heart failure. Further preferred cardiovascular complications encompass cardiac brady- or tachyarrhythmias including sudden cardiac death and stroke (cerebrovascular events or accidents). Most preferably, the said cardiovascular complication is ACS or heart failure.
The expression "predicting the risk of mortality" as used herein means that it the subject to be analyzed by the method of the present invention is allocated either into the group of subjects of a population having a normal, i.e. non-elevated, risk for mortality or into a group of subjects having a significantly elevated risk. An elevated risk as referred to in accordance with the present invention means that the risk of mortality within a predetermined predictive window is elevated significantly for a subject with respect to the average risk for mortality in a population of subjects. Preferably, for a predictive window of one year, the average risk is within the range of 0.5 and 3.0 %, preferably, 1.5%. An elevated risk as used herein, preferably, relates to a risk of more than 3.0 %, preferably, more than 5.0 %, and, most preferably within 3.0 % and 8.0 % with respect to a predictive window of one year.
Encompassed by the present invention is, further, a device for identifying a subject to be admitted to the hospital, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the subject and means for comparing said amount to a reference amount, whereby a subject to be admitted to the hospital is identified. Preferably, the subject is a subject presenting to the emergency unit. In the hospital, the subject may be admitted to the intensive care unit.
The term "device" as used herein relates to a system of means comprising at least the aforementioned means operatively linked to each other as to allow the prediction. Preferred means for determining the amount of GDF- 15 and means for carrying out the comparison are disclosed above in connection with the method of the invention. How to link the means in an operating manner will depend on the type of means included into the device. For example, where means for automatically determining the amount of the peptides are applied, the data obtained by said automatically operating means can be processed by, e.g., a computer program in order to obtain the desired results. Preferably, the means are comprised by a single device in such a case. Said device may accordingly include an analyzing unit for the measurement of the amount of the peptides or polypeptides in an applied sample and a computer unit for processing the resulting data for the evaluation. Alternatively, where means such as test stripes are used for determining the amount of the peptides or polypeptides, the means for comparison may comprise control stripes or tables allocating the determined amount to a reference amount. The test stripes are, preferably, coupled to a ligand which specifically binds to the peptides or polypeptides referred to herein. The strip or device, preferably, comprises means for detection of the binding of said peptides or polypeptides to the said ligand. Preferred means for detection are disclosed in connection with embodiments relating to the method of the invention above. In such a case, the means are operatively linked in that the user of the system brings together the result of the determination of the amount and the diagnostic or prognostic value thereof due to the instructions and interpretations given in a manual. The means may appear as separate devices in such an embodiment and are, preferably, packaged together as a kit. The person skilled in the art will realize how to link the means without further ado. Preferred devices are those which can be applied without the particular knowledge of a specialized clinician, e.g., test stripes or electronic devices which merely require loading with a sample. The results may be given as output of raw data which need interpretation by the clinician. Preferably, the output of the device is, however, processed, i.e. evaluated, raw data the interpretation of which does not require a clinician. Further preferred devices comprise the analyzing units/devices (e.g., biosensors, arrays, solid supports coupled to ligands specifically recognizing the natriuretic peptide, Plasmon surface resonace devices, NMR spectrometers, mass- spectrometers etc.) or evaluation units/devices referred to above in accordance with the method of the invention. Moreover, the present invention also relates to a device for predicting the risk of mortality or a further acute cardiovascular event for a subject adapted to carry out the method of the present invention comprising means for determining the amount of GDF- 15 and means for comparing said amounts to reference amounts, whereby it is predicted whether a subject is at risk of mortality or a further acute cardiovascular event.
Further envisaged is a device for deciding about admitting a subject to the hospital, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the said subject and means for comparing said amount to a reference amount, whereby it is decided whether the subject is to be admitted to the hospital.
The present invention also relates to a device for predicting the risk of mortality in a subject, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF-15 in a sample of the subject and means for comparing said amounts to reference amounts, whereby it is predicted whether the subject is at risk of mortality.
Preferably, the subject is a subject presenting to the emergency unit. In the hospital, the subject may be admitted to the intensive care unit.
Furthermore, a kit for carrying out the methods of the present invention, for identifying a subject to be admitted to the hospital, deciding about admitting a subject to the hospital, or predicting the risk of mortality in a subject is envisaged by the present invention. Said kit comprising means for determining the amount of GDF-15 in a sample of a subject and means for comparing said amounts to reference amounts, wherein a subject to be admitted to the hospital is identified, a decision about admitting the subject to the hospital or intensive care unit is made, or the risk of mortality in the subject is predicted.
The term "kit" as used herein refers to a collection of the aforementioned means, preferably, provided in separately or within a single container. The container, also preferably, comprises instructions for carrying out the method of the present invention.
All references cited in this specification are herewith incorporated by reference with respect to their entire disclosure content and the disclosure content specifically mentioned in this specification. The figures show:
Examples
EXAMPLE 1
We tested the hypothesis that use of GDF- 15 improves decision making in the emergency room and the overall management of patients presenting to emergency departments (EDs) by evaluating for incremental diagnostic and prognostic value of GDF- 15, and prospective Iy examining the clinical impact of GDF- 15 guided decision making regarding discharge, admission to hospital, and intensive care treatment.
METHODS:
A total of 303 patients presenting to the emergency department (ED) of an university hospital were studied. Blood samples were obtained in the ED from all patients admitted. GDF-15 was determined in 302 unselected consecutive patients. Follow-up at discharge included the assessment of clinical course and treatment. The variables discharge, admission, and intensive care treatment were studied and associated to the baseline GDF- 15 values. Cut-off thresholds of GDF-15 suitable for risk stratification and medical decision making (discharge or admissions to ICU or general care units), were calculated using ROC analysis.
RESULTS:
Presenting diagnoses were grouped into 4 categories:
1. Confirmed or suspected ischemic heart disease, including acute coronary syndrome and ischemic heart failure: 33 (10,9%)
2. Nonischemic heart disease, including arrhythmias without acute ischemic trigger, vascular heart disease, and cardiomyopathies: 36 (11,8%)
3. Lung disorders, including asthma, chronic obstructive disease, and pulmonary embolism: 41 (21,8%) 4. AU other disorders: 193 (63,5%)
In summary, 63,5% of the patients presented at the ED with non-cardiovascular or nonrespiratory disorders whereas 22,7% presented with ischemic and non-ischemic heart diseases. Statistical ananlysis
1. Descriptive statistic
Table 1 shows the values of arithmetic mean, median, and percentile und Table 2 and Fig. 1 show admission and discharge rates depending on GDF- 15 values above or below the GDF- 15 median value. Patients with GDF- 15 values above the median of 1,6605 ng/ml were admitted more often (Admission/Discharge rate = 2,17) compared to those with GDF- 15 values below the median (Admission/Discharge rate = 0,53).
2. Quartiles
The quartile ranges are displayed in Table 3 and Figure 2 showing that patients with increasing GDF- 15 values were admitted to the hospital. Patients in the lrst quartile revealed an admission/discharge rate of 0,43 compared with an admission/discharge rate of 4,85 of patients in the 4th quartile.
3. ROC analysis
Results of ROC analysis for prediction discharge or admission by using GDF- 15 values are shown in Table 4. The area under the curve (AUC) of 0,722 demonstrates the high discrimination power of GDF- 15 between discharge and admission. The GDF- 15 cut off value with optimal sensitivity/negative predictive value (66,7%) and optimal specifity/ positive predictive value (68,6%) was estimated as 1,56 ng/ml.
4. "Rule in" GDF- 15 cut off value for admission
In order to identify patients presenting at the emergency department who have to be admitted to hospital a GDF- 15 "rule in" cut off value of 8,21 ng/ml could be obtained from the ROC analysis. The corresponding sensitivity and negative predictive value is 99,3% and 95,2%, respectively. Patients with GDF- 15 values > 8,21 ng/ml should be admitted to hospital (and not be discharged) independent of diagnosis and underlying disease.
CONCLUSION GDF- 15 is a marker for decision making and a suitable tool for risk stratification of patients presenting at the emergency department. Moreover, by using GDF- 15 "rule in" cut off value patients could be identified who should be admitted to hospital and not be discharged independent of diagnosis and disease status of the patient.
Figure imgf000022_0001
Figure imgf000023_0001
A 58 years old female was admitted to the emergency room with suspected gastrointestinal bleeding or acute gastritis. Case history:
Chronic hepatitis B, liver cirrhosis
Physical examination: Weight: 62 Kg
Size: 162 cm
Heart rate: 60/min Blood pressure: 70/138 mmHg
Clinical signs and symptoms:
• No clinical signs for cardiovascular impairment.
• No clear clinical signs related to gastrointestinal bleeding or gastritis.
Laboratory:
GDF-15 value: 1,56 ng/ml; < 8,21 ng/ml (rule in cut off value for admission to hospital);
< 1,66 ng/ml (optimized ROC cut off value)
Result:
The patient could be discharged to home!
EXAMPLE 3
A 81 years old male was admitted to the emergency room with suspected myocardial infarction in an unclear clinical situation. Case history: Stable coronary heart disease, kidney disease
Physical examination:
Weight: 100 Kg
Size: 178 cm
Temperature: 39 0C
Heart rate : 103/min Blood pressure: 70/138 mmHg
Clinical signs and symptoms: • Dyspnoea
• Cardiac murmur but no clinical signs of decompensated acute heart failure.
• No signs of myocardial infarction.
Laboratory:
GDF-15 value: 20,62 ng/ml; > 8,21 ng/ml (rule in cut off value for admission to hospital).
Result:
The patient was admitted to the intensive care unit.
EXAMPLE 4
A 27 years old male was admitted to the emergency department with suspected acute gastritis. Case history:
Patient has never been hospitalized, and takes no medication. She exhibited no signs of health problems.
Physical examination: Weight: 85 Kg
Size: 180 cm
Temperature: 36 0C
Heart rate: 65/min
Blood pressure: 85/126 mmHg
Clinical signs and symptoms:
• Tickle/itch paresthesia
• Hperventilation
Laboratory:
GDF-15 value: 0,53 ng/ml; < 8,21 ng/ml (rule in cut off value for admission to hospital); < 1,66 ng/ml (optimized ROC cut off value)
Result: The patient was discharged to home.
EXAMPLE 5 A 37 years old female was admitted to the emergency room with vasovagale syncope.
Physical examination: Weight: 98 Kg
Size: 167 cm
Temperature: 37,2 0C Heart rate: 84/min
Blood pressure: 90/140 mmHg
Clinical signs and symptoms:
• Chest pain, atypical for myocardial infarction
• No other signs of cardiovascular disease
Laboratory:
GDF-15 value: 0,91 ng/ml; < 8,21 ng/ml (rule in cut off value for admission to hospital); < 1,66 ng/ml (optimized ROC cut off value)
Result: The patient was discharged to home.
EXAMPLE 6
A 46 female was admitted to the emergency room with intoxitation and suspected acute kindney dysfunction.
Case history: Anorexia nervosa
Physical examination:
Weight: 47 Kg
Size: 165 cm
Temperature: 36,5 0C Heart rate: 80/min Blood pressure: 76/134 mmHg
Clinical signs and symptoms: • Dyspnoea
Labortory:
GDF-15 value: 12,78 ng/ml; > 8,21 ng/ml (rule in cut off value for admission to hospital).
Result:
The patient was admitted to hospital.

Claims

Claims
1. A method of identifying if a subject is to be admitted to the hospital, the method comprising
a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount, whereby a subject to be admitted to the hospital is to be identified.
2. The method according to claim 1, wherein the subject presents to an emergency cunit.
3. The method according to claim 1 or 2, wherein the subject is admitted to a normal care unit in the hospital.
4. The method according to any of claims 1 to 3, wherein the subject is admitted to an intensive care unit in the hospital.
5. The method according to any of claims 1 to 4, wherein the subject is not suffering from an acute cardiovascular event
6. The method according to any of claims 1 to 5, wherein the reference amount identifying a subject to be admitted to the hospital is 1200 pg/ml or higher.
7. The method according to any of claims 1 to 6, wherein the reference amount identifying a subject to be admitted to the hospital is 1560 pg/ml or higher.
8. The method according to any of claims 1 to 7, wherein the reference amount identifying a subject to be admitted to the hospital is 8210 pg/ml or higher.
9. A method of deciding about admitting a subject to the hospital, the method comprising a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount; and c) deciding whether the subject is to be admitted to the hospital.
10. A method of predicting the risk of mortality for a subject comprising a) determining the amount of GDF- 15 in a sample of the subject ; and b) comparing the amount of GDF- 15 determined in step a) to a reference amount; and c) predicting the risk of mortality based on the result of steps a) and b).
11. A device for identifying a subject to be admitted to the hospital, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the subject and means for comparing said amount to a reference amount, whereby a subject to be admitted to the hospital is identified.
12. A device for deciding about admitting a subject to the hospital, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the said subject and means for comparing said amount to a reference amount, whereby it is decided whether the subject is to be admitted to the hospital.
13. A device for predicting the risk of mortality in a subject, adapted to carry out the method of the present invention, comprising means for determining the amount of GDF- 15 in a sample of the subject and means for comparing said amounts to reference amounts, whereby it is predicted whether the subject is at risk of mortality.
14. A kit for carrying out the methods of the present invention.as described in any of claims 1 to 8, comprising means for determining the amount of GDF- 15 in a sample of a subject and means for comparing said amounts to reference amounts.
PCT/EP2009/050170 2008-01-08 2009-01-08 Means and methods for assessing the risk of patients presenting to emergency units based on gdf-15 WO2009087190A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP09700201A EP2227696A1 (en) 2008-01-08 2009-01-08 Means and methods for assessing the risk of patients presenting to emergency units based on gdf-15
JP2010541072A JP2011509403A (en) 2008-01-08 2009-01-08 Means and method for assessing the risk of a patient visiting an emergency room based on GDF-15
US12/822,690 US20100261284A1 (en) 2008-01-08 2010-06-24 Using gdf 15 to assess patients presenting to emergency units

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP08150098.5 2008-01-08
EP08150098 2008-01-08

Publications (1)

Publication Number Publication Date
WO2009087190A1 true WO2009087190A1 (en) 2009-07-16

Family

ID=39689087

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/050170 WO2009087190A1 (en) 2008-01-08 2009-01-08 Means and methods for assessing the risk of patients presenting to emergency units based on gdf-15

Country Status (4)

Country Link
US (1) US20100261284A1 (en)
EP (1) EP2227696A1 (en)
JP (1) JP2011509403A (en)
WO (1) WO2009087190A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011058335A1 (en) * 2009-11-13 2011-05-19 Universitetet I Oslo Sgii as a prognostic marker in conditions which require critical care
EP2388594A1 (en) * 2010-05-17 2011-11-23 Roche Diagnostics GmbH GDF-15 based means and methods for survival and recovery prediction in acute inflammation
EP3943946A1 (en) * 2020-07-20 2022-01-26 F. Hoffmann-La Roche AG Gdf-15 for predicting the disease severity of a patient with covid-19

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013148117A1 (en) 2012-03-27 2013-10-03 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
BR112015018104A2 (en) 2013-01-30 2017-11-21 Ngm Biopharmaceuticals Inc modified gdf15 polypeptides, their use, compositions comprising them and sterile container comprising said compositions
US9161966B2 (en) 2013-01-30 2015-10-20 Ngm Biopharmaceuticals, Inc. GDF15 mutein polypeptides
EA035581B1 (en) 2014-07-30 2020-07-10 НДжМ БИОФАРМАСЬЮТИКАЛЗ, ИНК. Compositions and methods of use for treating metabolic disorders
TN2017000113A1 (en) 2014-10-31 2018-07-04 Ngm Biopharmaceuticals Inc Compositions and methods of use for treating metabolic disorders
US11143659B2 (en) 2015-01-27 2021-10-12 Arterez, Inc. Biomarkers of vascular disease
US11604194B2 (en) 2016-02-29 2023-03-14 Public University Corporation Yokohama City University Method for detecting castration-resistant prostate cancer and detection reagent
PE20190126A1 (en) 2016-03-31 2019-01-17 Ngm Biopharmaceuticals Inc BINDING PROTEINS AND METHODS OF USING THEM

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1884777A1 (en) * 2006-08-04 2008-02-06 Medizinische Hochschule Hannover Means and methods for assessing the risk of cardiac interventions based on GDF-15
EP1995596A1 (en) * 2007-05-24 2008-11-26 F. Hoffmann-La Roche AG Means and methods for assessing heart failure in patients with atrial fibrillation using GDF-15 and natriuretic petpides

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2390820A1 (en) * 2002-06-17 2003-12-17 St. Vincent's Hospital Sydney Limited Methods of diagnosis, prognosis and treatment of cardiovascular disease

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1884777A1 (en) * 2006-08-04 2008-02-06 Medizinische Hochschule Hannover Means and methods for assessing the risk of cardiac interventions based on GDF-15
EP1995596A1 (en) * 2007-05-24 2008-11-26 F. Hoffmann-La Roche AG Means and methods for assessing heart failure in patients with atrial fibrillation using GDF-15 and natriuretic petpides

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
EGGERS KAI M ET AL: "Growth-differentiation factor-15 for early risk stratification in patients with acute chest pain", EUROPEAN HEART JOURNAL, vol. 29, no. 19, October 2008 (2008-10-01), pages 2327 - 2335, XP002526245, ISSN: 0195-668X *
KEMPF TIBOR ET AL: "Circulating concentrations of growth-differentiation factor 15 in apparently healthy elderly individuals and patients with chronic heart failure as assessed by a new immunoradiometric sandwich assay", CLINICAL CHEMISTRY, vol. 53, no. 2, 1 February 2007 (2007-02-01), pages 284 - 291, XP002438496 *
KEMPF TIBOR ET AL: "Growth-Differentiation Factor-15 is an Independent Predictor of Cardiovascular Events and Mortality in Patients with Stable Coronary Artery Disease", CIRCULATION, vol. 118, no. 18, Suppl. 2, October 2008 (2008-10-01), & 81ST ANNUAL SCIENTIFIC SESSION OF THE AMERICAN-HEART-ASSOCIATION; NEW ORLEANS, LA, USA; NOVEMBER 08 -12, 2008, pages S1132, XP002526244, ISSN: 0009-7322 *
WOLLERT KAI C ET AL: "Growth differentiation factor 15 for risk stratification and selection of an invasive treatment strategy in non ST-elevation acute coronary syndrome.", CIRCULATION 2 OCT 2007, vol. 116, no. 14, 2 October 2007 (2007-10-02), pages 1540 - 1548, XP002493258, ISSN: 1524-4539 *
WOLLERT KAI C ET AL: "Prognostic value of growth-differentiation factor-15 in patients with non-ST-elevation acute coronary syndrome", CIRCULATION, LIPPINCOT WILLIAMS AND WILKINS, BALTIMORE, US, vol. 115, no. 8, 27 February 2007 (2007-02-27), pages 962 - 971, XP002438129, ISSN: 1524-4539 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011058335A1 (en) * 2009-11-13 2011-05-19 Universitetet I Oslo Sgii as a prognostic marker in conditions which require critical care
EP2388594A1 (en) * 2010-05-17 2011-11-23 Roche Diagnostics GmbH GDF-15 based means and methods for survival and recovery prediction in acute inflammation
WO2011144571A3 (en) * 2010-05-17 2012-03-15 F. Hoffmann-La Roche Ag Gdf-15 based means and methods for survival and recovery prediction in acute inflammation
EP3943946A1 (en) * 2020-07-20 2022-01-26 F. Hoffmann-La Roche AG Gdf-15 for predicting the disease severity of a patient with covid-19
WO2022017980A1 (en) * 2020-07-20 2022-01-27 F. Hoffmann-La Roche Ag Gdf-15 for predicting the disease severity of a patient with covid-19

Also Published As

Publication number Publication date
JP2011509403A (en) 2011-03-24
EP2227696A1 (en) 2010-09-15
US20100261284A1 (en) 2010-10-14

Similar Documents

Publication Publication Date Title
US20100261284A1 (en) Using gdf 15 to assess patients presenting to emergency units
US7927812B2 (en) Cardiac troponin as an indicator of coronary artery disease
US20110033886A1 (en) Gdf-15 as biomarker in type 1 diabetes
US20130035603A1 (en) Troponin based rule-in and rule-out algorithm of myocardial infarction
EP2209003A1 (en) Means and methods for differentiating between fibrosis and cirrhosis
WO2009100907A1 (en) Means and methods for assessing the risk of patients presenting to emergency departments based on very low concentrations of troponin i or t or using a combination of markers
EP1890154B1 (en) Cardiac troponin as an indicator of advanced coronary artery disease
US8497095B2 (en) Biochemical markers for acute pulmonary embolism
US20170261517A1 (en) NT-pro ANP and SFlt-1 For The Differentiation Between Circulatory And Ischemic Events
US20190257841A1 (en) USE OF sCD14 OR ITS FRAGMENTS OR DERIVATIVES FOR RISK STRATIFICATION, DIAGNOSIS AND PROGNOSIS
JP2013527453A (en) Means and methods based on GDF-15 for estimating survival and recovery in acute inflammation
JP5715641B2 (en) Method for diagnosing and monitoring cardiac ischemia in a patient with acute chest pain and no myocardial infarction
EP2291660A1 (en) Means and methods for determining the arteriosclerotic stenosis using inflammatory biomarkers
WO2009034121A1 (en) Vascular markers in the remodelling of cardiac injury
EP2367006A1 (en) PLGF-based means and methods for diagnosing cardiac causes in acute inflammation
EP2444813A1 (en) S100B-based means and methods for diagnosing cerebral damages in acute inflammation

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: 09700201

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010541072

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2009700201

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE