WO2005071408A1 - Method of diagnosing infectious disease by measuring the level of soluble trem-1 in a sample - Google Patents

Method of diagnosing infectious disease by measuring the level of soluble trem-1 in a sample Download PDF

Info

Publication number
WO2005071408A1
WO2005071408A1 PCT/GB2005/000273 GB2005000273W WO2005071408A1 WO 2005071408 A1 WO2005071408 A1 WO 2005071408A1 GB 2005000273 W GB2005000273 W GB 2005000273W WO 2005071408 A1 WO2005071408 A1 WO 2005071408A1
Authority
WO
WIPO (PCT)
Prior art keywords
strem
disease
bacterial
sample
level
Prior art date
Application number
PCT/GB2005/000273
Other languages
French (fr)
Inventor
Marie Nathalie Kolopp-Sarda
Marie-Christine Bene
Paola Panina
Pietro Di Lucia
Bruno Levy
Pierre-Edouard Bollaert
Gilbert Faure
Sebastien Gibot
Original Assignee
Bioxell S.P.A.
Universite Henri Poincare, Nancy I
Thomas, Neil, Ciaron
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 Bioxell S.P.A., Universite Henri Poincare, Nancy I, Thomas, Neil, Ciaron filed Critical Bioxell S.P.A.
Priority to JP2006550296A priority Critical patent/JP4911600B2/en
Priority to EP05702028A priority patent/EP1709444B1/en
Priority to US10/587,356 priority patent/US8021836B2/en
Priority to AU2005207111A priority patent/AU2005207111A1/en
Priority to ES05702028T priority patent/ES2386966T3/en
Priority to CA002554769A priority patent/CA2554769A1/en
Publication of WO2005071408A1 publication Critical patent/WO2005071408A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/12Pulmonary diseases
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/26Infectious diseases, e.g. generalised sepsis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/56Staging of a disease; Further complications associated with the disease

Definitions

  • This invention relates generally to the field of immunology. More particularly, the present invention relates to inflammation and the use of markers that allow the prompt diagnosis of infectious disease (for example of bacterial or fungal) origin and the follow up of infected patients during pharmacological treatment. These markers have particular applications in the diagnosis of pneumonia and sepsis.
  • infectious disease for example of bacterial or fungal
  • the Inventors describe a rapid detection test of the soluble form of the human TREM-1 receptor (sTREM-1) in bronchoalveolar fluid of mechanically ventilated patients to accurately diagnose bacterial or fungal pneumonia. Many non-infectious processes lead to fever and new pulmonary infiltrates in the mechanically ventilated patient, rendering the diagnosis of pneumonia (and especially ventilator-associated pneumonia) very challenging.
  • the systemic signs of infection are non-specific findings and can be caused by any condition that releases cytokines.
  • Pugin etal. Am Rev Respir Dis 1991;143:1121-9 combined body temperature, white biood cells count, volume and appearance of tracheal secretions, ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (PaO2/FiO2), chest-X-ray, and tracheal aspirate cultures into a clinical pulmonary infection score (CPIS) and reported that a score >6 was associated with a high likelihood of pneumonia.
  • CPIS clinical pulmonary infection score
  • TNF ⁇ tumor necrosis factor- ⁇
  • IL-1 ⁇ interleukin-1 ⁇
  • other cytokines have been demonstrated to be increased in various types of pulmonary infectious challenges with potential prognostic implications.
  • the Inventors were unable to determine accurate discriminating cut-off level of such mediators for the diagnosis of pneumonia.
  • the Inventors using an easy-to-perform immunoblot technique, demonstrate herein that a soluble form of TREM-1 (sTREM-1) is released locally in the bronchoalveolar lavage fluid from patients suffering from pneumonia with a sensitivity of at least 98 percent.
  • sTREM-1 was detected in only 6 out of 64 patients without pneumonia. Bronchoalveolar lavage fluid levels of sTREM-1 were not correlated to any of the clinical or biological parameters tested and stood as an independent parameter of high specificity. In a multiple logistic regression analysis, presence of sTREM-1 in bronchoalveolar lavage fluid was shown to be the best predictor of pneumonia with an odds ratio as high as 41.52. Presence of sTREM-1 by itself was more accurate that any clinical findings or laboratory values in identifying the existence of bacterial or fungal pneumonia. Thus rapid detection of sTREM-1 in bronchoalveolar lavage fluid is useful in establishing or excluding the diagnosis of bacterial or fungal pneumonia.
  • PCT procalcitonin
  • CRP C-reactive protein
  • SIRS systemic inflammatory response syndrome
  • Sepsis is further stratified into severe sepsis when there is evidence of organ hypoperfusion, made evident by ' signs of organ dysfunction such as hypoxaemia, oliguria, lactic acidosis or altered cerebral function.
  • Septic shock is severe sepsis complicated by hypotension defined as systolic blood pressure less than 90 mmHg despite adequate fluid resuscitation.
  • Sepsis and SIRS may be complicated by the failure of two or more organs, termed multiple organ failure (MOF), due to disordered organ perfusion and oxygenation.
  • MOF multiple organ failure
  • a systemic inflammatory response may occur in severe inflammatory conditions such as pancreatitis and burns. The appearance of signs of an inflammatory response is less well defined following traumatic insults.
  • gram-negative bacteria are implicated in 50 to 60% of sepsis with gram-positive bacteria accounting for a further 35 to 40% of cases. The remainder of cases are due to the less common causes of fungi, viruses and protozoa.
  • SIRS Systemic Inflammatory Response Syndrome
  • Early recognition of sepsis and Systemic Inflammatory Response Syndrome (SIRS) in the critically ill patient may avoid the increased morbidity, mortality and length of stay associated with multiple organ failure.
  • SIRS Systemic Inflammatory Response Syndrome
  • the present invention is directed towards circumventing the existing problems associated with diagnosing sepsis to provide an accurate and consistent method of detection.
  • the Inventors describe the value of assaying the soluble form of TREM-1 (sTREM-1) in plasma samples of newly admitted critically ill patients with suspected sepsis as a new approach to accurately diagnose infectious processes.
  • sTREM-1 soluble form of TREM-1
  • Early identification of infection has a major impact on the clinical course, management and outcome of critical patients.
  • Critical care physicians have at their disposal a variety of indicators to serve as a guide in discriminating infectious from non-infectious conditions in newly admitted patients.
  • the diagnosis of sepsis becomes clear after completing the medical history and physical examination of a newly admitted patient (Bates DW, et al. Ann Intern Med. 1990;113:495-500).
  • plasmatic sTREM-1 level assay offers a higher degree of certainty than other currently available candidates. This accuracy can usefully guide physicians in their clinical decision-making and stepwise approach to the complex management of critically ill patients.
  • the immunoblot technique used here can be performed within 3 to 4 hours and may provide valuable information long before blood culture results are back. Moreover, it is of low cost and can be applied to small series or even individual samples.
  • the results reported here demonstrate that rapid measurement of the plasmatic sTREM-1 levels may improve the ability of clinicians to differentiate patients with sepsis from those with systemic inflammation of non-infectious origin. This should be especially useful among patients in whom the diagnosis is not clinically straightforward.
  • the immunoblot technique described is rapid, accurate, of low cost and can be applied to small series or even individual samples. Use of this test to assess plasmatic sTREM-1 levels should lead to a more accurate diagnosis of sepsis in patients admitted in ICUs with a clinical suspicion of infection.
  • the triggering receptor expressed on myeloid cells-1 is a member of the Ig-superfamily, the expression of which is up-regulated on phagocytic cells in the presence of bacteria or fungi (Bouchon A et al. Nature 2001;230:1103-7).
  • the inventors have determined that TREM-1 is shed or secreted from the membrane of activated phagocytes and can be found in a soluble form in body fluids and is therefore a useful diagnostic marker.
  • the presence of a soluble form of TREM-1 (sTREM-1) in bronchoalveolar lavage (BAL) fluid from mechanically ventilated patients is shown herein to be a good indicator of infectious pneumonia.
  • BAL bronchoalveolar lavage
  • the present invention provides methods and compositions for the clinical screening and diagnosis of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • the present invention provides methods and compositions for monitoring the effectiveness of the treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis, for selecting participants in clinical trials relating disease of bacterial or fungal origin, for identifying subjects most likely to respond to a particular therapeutic treatment for disease of bacterial or fungal origin and for screening and development of drugs for treatment of disease of bacterial or fungal origin.
  • the invention provides a method of diagnosis of disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject.
  • the disease is an inflammatory state
  • said method is capable of identifying a microbial origin for said inflammatory state.
  • inflammatory states pneumonia and sepsis of bacterial or fungal origin.
  • the invention provides a method of diagnosis of pneumonia in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject.
  • the invention provides a method of diagnosing sepsis of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject.
  • the invention provides methods of diagnosing or monitoring disease of bacterial or fungal origin, for example, pneumonia or sepsis in a patient, comprising: measuring the level of sTREM-1 in a sample from the patient, wherein the level is an indicator of presence or extent of disease of bacterial or fungal origin in the patient.
  • sTREM-1 is a soluble form of the TREM-1 Receptor which can be detected in certain body fluid samples by an antibody raised against the TREM-1 Receptor.
  • sTREM-1 means, an inflammation of the lung caused by infection by extracellular pathogens such as bacterial infection, and non-bacterial infections (for example, infection by Blastomyces dermatitidis, Histoplasma capsulatum, Coccidioides, Sporothrix schenckii, Pneumocystis carinii, Cryptococcus, Aspergillus, or Mucorsp.), protozoal infections or parasitic infections (for example, those caused by Toxoplasma gondii, Strongyloides stercoralfs, Ascaris, hookworm, Dirofilaria, Paragonimus, or Entamoeba histolytica) where increased expression of sTREM-1 can be detected.
  • non-bacterial infections for example, infection by Blastomyces dermatitidis, Histoplasma capsulatum, Coccidioides, Sporothrix schenckii, Pneumocystis carinii, Cryptococcus, Aspergill
  • Pneumonia includes "Lobar Pneumonia” (which occurs in one lobe of the lung) and Bronchopneumonia (tends to be irregularly located in the lung). Furthermore, pneumonia is often classified into two categories that may help predict the organisms that are the most likely culprits. "Community- acquired (pneumonia contracted outside the hospital). Pneumonia” in this setting often follows a viral respiratory infection. It affects nearly 4 million adults each year. It is likely to be caused by Streptococcus pneumoniae, the most common pneumonia-causing bacteria. Other organisms, such as atypical bacteria called Chlarnydia or Mycoplasma pneumonia are also common causes of community-acquired pneumonia.
  • SIRS Systemic Inflammatory Response Syndrome
  • extracellular pathogens such as bacterial infection, for example bacteremia (the presence of bacteria in the blood) with or without organ failure, and non- bacterial infections, such as fungemia (for example, yeast infection by Candida albicans), protozoal infections or parasitemia (such as in filariasis, malaria, and trypanosomiasis) where increased expression of sTREM-1 can be detected.
  • bacterial infection for example bacteremia (the presence of bacteria in the blood) with or without organ failure
  • non- bacterial infections such as fungemia (for example, yeast infection by Candida albicans), protozoal infections or parasitemia (such as in filariasis, malaria, and trypanosomiasis) where increased expression of sTREM-1 can be detected.
  • fungemia for example, yeast infection by Candida albicans
  • protozoal infections or parasitemia such as in filariasis, malaria, and trypanosomiasis
  • the measurement of the level of sTREM-1 comprises the steps of (a) contacting said biological sample with a compound capable of binding sTREM-1 ; and (b) detecting the level of sTREM-1 present in the sample by observing the level of binding between said compound and sTREM- 1.
  • the assay or measurement of the sample for the levels of sTREM-1 present in the sample may be carried out using standard protocols known in the art. For example, where the observation of binding between sTREM-1 and the compound capable of binding sTREM-1 takes place, this observation may be carried out using known methodologies.
  • the binding may be detected through use of a competitive immunoassay, a non-competitive assay system using techniques such as western blots, a radioimmunoassay, an ELISA (enzyme linked immunosorbent assay), a "sandwich” immunoassay, an immunoprecipitation assay, a precipitin reaction, a gel diffusion precipitin reaction, an immunodiffusion assay, an agglutination assay, a complementfixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, an immunoprecipitation assay, an immunohistochemical assay, a competition or sandwich ELISA, a radioimmunoassay, a Western blot assay, an immunohistological assay, an immunocytochemical assay, a dot blot assay, a fluorescence polarization assay, a scintillation proximity assay, a homogeneous time resolved fluorescence
  • the determination of the incidence of of bacterial or fungal origin may comprise deriving the likelihood ratio using a multivariate analysis based on distribution parameters from a set of reference data derived from analysis of the levels of sTREM-1 in patients in which disease of bacterial or fungal origin is absent, present or in remission.
  • the invention therefore also provides diagnostic means capable of measuring levels of sTREM-1 and/or comparing said levels to known levels that are indicative of the disease state of the patient.
  • diagnostic means can take the form of a stick test, for example carrying the necessary reagents to perform the method of the invention and to produce, for example, a colorimetric result which can be compared against a colour chart.
  • the method according to the first aspect of the invention can comprise the further step of c) correlating the detected level of sTREM-1 with the presence or absence of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • a correlation can be made by comparing the measured level of sTREM-1 in the sample with a mean level in samples obtained from a control population of individuals not having disease of bacterial or fungal origin, to indicate the presence or extent of disease of bacterial or fungal origin in the patient.
  • the method according to the first aspect of the invention can be used in monitoring the progression or remission of disease of bacterial or fungal origin, in other words, to indicate the progression or remission of the disease.
  • Such methods can be used to monitor the effectiveness and/or progress of therapy in a subject.
  • the method further comprises the steps of measuring the level of sTREM-1 in a second or further sample from the patient, the first and second or further samples being obtained at different times; and comparing the levels in the samples to indicate the progression or remission of the disease of bacterial or fungal origin.
  • the diagnostic methods according to the present invention are carried out ex vivo.
  • Biological samples for analysis by the methods of the invention can be obtained using methods known in the art from various sources, in particular from body fluids such as whole blood, blood serum, blood plasma, urine and bronchoalveolar lavage fluid.
  • the sample should be a sample treated such that any sTREM-1 present is not removed prior to the assay or is rendered undetectable.
  • a preferred biological sample is a sample of bronchoalveolar lavage fluid.
  • a preferred biological sample is a sample of blood serum.
  • the methods of the invention are applicable to mammals, for example humans, non-human primates, sheep, pigs, cows, horses, goats, dogs, cats and rodents, such as mouse and rat.
  • the biological sample tested by the methods of the invention is a human sample.
  • the biological sample should generally contain protein molecules from the test subject and is handled such that proteins in the sample are not rendered undetectable by the compound chosen to detect them.
  • the term "compound capable of binding sTREM-1” means polypeptides, ligands, antibodies or otherwise discriminating entities which predominantly, preferably specifically, bind to sTREM-1.
  • Such binding compounds, or "sTREM-1 binding partners” can be a naturally occurring sTREM-1 binding molecule, for example a ligand for the TREM-1 Receptor or sTREM-1 and natural and synthetic variants thereof.
  • binding compounds include, a chemically modified or genetically modified derivative of a sTREM-1 binding molecule, an artificially (for example chemically produced) sTREM-1 binding molecule or a recombinant or engineered soluble sTREM-1 binding molecule.
  • antibodies which bind predominately, preferably specifically or exclusively to, sTREM-1 including, but not limited to, those antibodies which are: mono-or polyclonal antibodies (for example, raised against sTREM-1), bi-specific, multi-specific, human, humanized, chimeric antibodies, single chain antibodies, antibodies derived from phage display techniques, Fab fragments, F(ab')2 fragments, disulfide- linked Fvs, and fragments containing either a VL or VH domain or even a complementary determining region (CDR) that specifically binds to sTREM-1.
  • mono-or polyclonal antibodies for example, raised against sTREM-1
  • bi-specific, multi-specific, human, humanized, chimeric antibodies single chain antibodies
  • antibodies derived from phage display techniques Fab fragments, F(ab')2 fragments, disulfide- linked Fvs, and fragments containing either a VL or VH domain or even a complementary determining region (CDR) that specifically
  • modified immunoglobulins are also included within the scope of the invention, for example a fusion of the TREM-1 -Receptor to one or more immunoglobulin-derived protein domains, for example to confer solubility and/or stability, for example human IgG or IgM Fc fragments.
  • substances or products mimicking the tertiary structure of a ligand for the TREM-1 -Receptor can be used as binding partners specific for sTREM-1. It is possible to design such on the basis of computer modelling. The product can be produced synthetically using chemical means. Use of recombinant DNA technology to engineer the required structure is also possible as is chemical modification.
  • a compound capable of binding sTREM-1 is an antibody raised against the TREM-1 receptor, a fragment thereof or a variant thereof, provided that it is capable of binding sTREM-1.
  • a compound capable of binding sTREM-1 is an antibody raised against the TREM-1 receptor, a fragment thereof or a variant thereof, provided that it is capable of binding sTREM-1.
  • such an antibody is one raised against TREM-1 human Fc (TREM-1 -Fc) fusion protein (see Example 1 herein).
  • the invention provides a compound capable of binding sTREM-1 for use in the diagnosis, prognosis, treatment or monitoring of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • the invention provides use of a compound capable of binding sTREM-1 in a method of treatment or diagnosis of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • the invention provides use of a compound capable of binding sTREM-1 in the manufacture of a medicament for the diagnosis, prognosis, treatment or monitoring of the treatment disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • the methods described herein can furthermore be used as screening assays to identify a subject with, or at risk of developing, disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • Such assays can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat disease of bacterial or fungal origin.
  • an agent e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate
  • agents e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate
  • agents e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate
  • such methods can be used to determine whether a subject
  • the present invention provides methods for determining whether a subject can be effectively treated with an agent for disease of bacterial or fungal origin, for example, pneumonia or sepsis in which a test sample is obtained and TREM-1 is detected.
  • a further embodiment of the invention provides a pharmaceutical composition comprising a compound capable of binding sTREM-1 together with a pharmaceutically acceptable diluent, carrier orexcipient for use in the diagnosis or treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • a pharmaceutically acceptable diluent, carrier orexcipient for use in the diagnosis or treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • the invention also provides a compound capable of binding sTREM-1 for use in, or used in, a method of diagnosis or treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • pharmaceutically acceptable diluent, carrier or excipient is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • a third aspect of the invention provides a method of identifying agonists or antagonists of sTREM-1 said method comprising comparing the level of binding in a sample containing said sTREM-1 and a compound capable of binding sTREM-1, in the presence and absence of a compound to be tested. Also provided by are agonists or antagonists of sTREM-1 identified according to the method of this aspect of the invention.
  • kits, associated reagents and contacting means comprising at least one compound capable of binding sTREM-1 and reagents for detecting binding of said compound to sTREM-1.
  • kits comprising at least one compound capable of binding sTREM-1 and means for contacting said compound with a sample containing sTREM-1.
  • the kit can comprise, for example: (1) a binding compound (e.g., attached to a solid support) that binds to sTREM-1; and, optionally, (2) a second, different binding compound e.g. an antibody, which binds to either the sTREM-1 or the first binding compound and is conjugated to a detectable agent.
  • kits can also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent.
  • the kit can also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate).
  • the kit can also contain a control sample or a series of control samples which can be assayed and compared to the test sample contained.
  • Each component of the kit is usually enclosed within an individual container, and all of the various containers are within a single package, along with instructions for determining whether the subject from which. the sample is derived is suffering from or is at risk of developing disease of bacterial or fungal origin, for example, pneumonia or sepsis.
  • an antibody raised against the TREM-1 -Receptor, a fragment thereof or a variant thereof can function as a compound c pable of binding sTREM-1.
  • Antibodies are preferably raised against the human TREM-1 -Receptor (triggering receptor expressed on myeloid cells) for which the cDNA sequence is given in [SEQ ID NO:1].
  • the TREM-1 -Receptor is expressed on human myeloid cells, is a transmembrane protein of the immunoglobulin superfamily (Ig-SF).
  • the TREM-1 -Receptor is a transmembrane glycoprotein having the amino acid sequence of [SEQ I D NO:2] that is selectively expressed on blood neutrophils and a subset of monocytes but not on lymphocytes and other cell types.
  • the invention encompasses antibodies raised against isolated or recombinantly prepared TREM proteins or polypeptides or fragments, homologues, derivatives, or variants thereof, as defined herein, as "TREM-1 -Receptor-derived polypeptides"
  • TREM-1 -Receptor-derived polypeptides In accordance with the definition of "compound capable of binding sTREM-1", such antibodies raised against “TREM-1 -Receptor-derived polypeptides" predominantly, preferably specifically, bind sTREM-1.
  • Such antibodies may be tested for binding with cells expressing the TREM-1 receptor and preferably also a sample from a patient known to have been suffering from pneumonia or sepsis of bacterial or fungal origin.
  • the term "homologue,” especially "TREM-1 -Receptor homologue” as used herein refers to any member of a series of peptides to which antibodies capable of binding sTREM-1 can be raised-. TREM-1 -Receptor homologues can be from either the same or different species of animals.
  • variant refers either to a naturally occurring allelic variation of a given peptide or a recombinantly prepared variation of a given peptide or protein in which one or more amino acid residues have been modified by amino acid substitution, addition, or deletion.
  • the term "derivative" as used herein refers to a variation of given peptide or protein that are otherwise modified, i.e., by covalent attachment of any type of molecule, preferably having bioactivity, to the peptide or protein, including non-naturally occurring amino acids.
  • the human TREM-1 -Receptor cDNA is 884-nucleotide long (Fig. 1; [SEQ ID NO:1]) and the open reading frame of TREM-1 -Receptor is nucleotides 48 to 752 of [SEQ ID NO:1], which encodes a transmembrane protein comprising the 234 amino acid sequence shown in Fig. 2 [SEQ 1 D NO:2].
  • the human TREM-1 -Receptor cDNA can be found in the GenBank database under accession number AF 196329.
  • the putative transmembrane domain starts from amino acid residues 201 to 229 of [SEQ ID NO:2J and contains a charged lysine residue at position 217. Its cytoplasmic tail consists of 5 amino acid residues and appears to contain no signaling motifs.
  • antibodies for binding sTREM-1 are raised against a TREM-1 -Receptor-derived polypeptide comprising at least an extracellular domain comprising amino acid residues 17 to 200 of [SEQ ID NO:2].
  • antibodies suitable for use in the invention are those antibodies having the ability to bind sTREM- 1 which are raised against homologues of TREM-1-Receptor from either the same or different species of animal, preferably from mammals, more preferably from rodents, such as mouse and rat, and most preferably from human.
  • Homologues of the TREM-1 -Receptor nucleic acid molecule i.e., [SEQ ID NO:1]
  • aspects of fiie invention can be also applied in the framework of multiple diagnosis of a subject.
  • a method of screening a patient for presence or susceptibility to disease comprising performing a plurality of diagnostic tests on a tissue sample from the patient for a plurality of diseases
  • the invention provides the improvement wherein one of the diagnostic tests comprises measuring the level of sTREM-1.
  • a diagnostic means for detecting disease of bacterial or fungal origin for example, pneumonia or sepsis
  • a diagnostic kit comprising such a diagnostic means
  • a method of treatment of infection which includes the step of screening an individual for disease of bacterial or fungal origin, for example, pneumonia or sepsis, wherein disease of bacterial or fungal origin is correlated with the levels of sTREM-1 in a sample from said individual, and if disease of bacterial or fungal origin is identified, treating that individual to prevent or reduce the infection
  • a bronchoalveolar lavage sample from a patient with lung-related symptoms would be used to diagnose pneumonia based upon elevated levels of sTREM-1.
  • a blood serum sample from a patient exhibiting symptoms of SIRS would be used to diagnose sepsis of bacterial or fungal origin based upon elevated levels of sTREM-1.
  • the invention also provides a method of diagnosing disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 and the step of measuring the level of TREM- 1 -Ligand in one or more biological samples obtained from said subject.
  • the Inventors have developed an immuno-enzymatic (in this case ELISA) based method for the detection of soluble TREM-1.
  • the invention provides a method of diagnosing disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject and wherein the level of sTREM-1 is measured by an immunochemical technique.
  • immunochemical techniques are indirect immunofluorescence (I IF), immunoperoxydase (POD), western immunoblotting (WB), radioimmunoprecipitation (RIPA), enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and agglutination assays.
  • I IF indirect immunofluorescence
  • POD immunoperoxydase
  • WB western immunoblotting
  • RIPA radioimmunoprecipitation
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • agglutination assays agglutination assays.
  • an ELISA method using an anti-human Trem-1 antibody is used to measure the level of sTREM-1.
  • WO2004081233 describes a method of diagnosing bacterial or fungal sepsis in a subject by measuring the level of TREM-1 -Ligand in a biological sample obtained from the subject and compounds capable of binding TREM- 1 -Ligan
  • the level of TREM-1 -Ligand present in the sample is measured by observing the level of binding between these compounds and TREM-1 -Ligand.
  • the Inventors have determined that measurement of both soluble TREM-1 (as described herein) and membrane associated TREM-1 Ligand (as described in WO2004081233) in newly admitted critically ill patients allows the rapid identification those with infection.
  • Figure 1 shows Human TREM-1 -Receptor cDNA [SEQ ID NO:1].
  • Figure 2 shows Human TREM-1-Receptor amino acid sequence [SEQ ID NO:2].
  • Figure 3 shows the levels of sTREM-1 in bronchoalveolar lavage fluid from patients according to diagnosis. Individual values are plotted and the bars represent the means of the values. P ⁇ 0.001 between CAP and NP and between VAP and NP.
  • Figure 4. shows receiver-operating-characteristic curves for various cut-off levels of bronchoalveolar lavage fluid sTREM-1, Tumor necrosis factor- ⁇ and Interleukin-1 ⁇ in differentiating between presence and absence of pneumonia.
  • sTREM-1 0.93 (95% confidence interval, 0.92 to 0.95)
  • Tumor necrosis factor- ⁇ 0.64 (95% confidence interval, 0.62 to 0.69)
  • lnterleukin-1 ⁇ 0.69 (95% confidence interval, 0.67 to 0.72)
  • Figure 5 shows bronchoalveolar lavage fluid (BAL) supernatants examined by Western blot analysis using 21 C7, an anti TREM-1 monoclonal antibody: Lane 1 : positive control (sTREM-1 , 50 pg/mL) Lane 2: BAL supernatant from a patient with pneumonia Lane 3: BAL supernatant from a patient without pneumonia
  • Figure 6. shows a flow-chart of the patients admitted to the ICU during the study period.
  • Figure 8 shows Receiver-operating-characteristic curves for various cut-off levels of plasmatic C-Reactive Protein, Procalcitonin and sTREM-1 in differentiating between presence and absence of infection. Areas under the ROC. curves for: C-Reactive Protein: 0.77 (95% confidence interval, 0.69 to 0.85) Procalcitonin: 0.85 (95% confidence interval, 0.81 to 0.89) sTREM-1: 0.97 (95% confidence interval, 0.94 to 1.0)
  • FIG. 9 shows admission plasmatic levels of C-Reactive Protein
  • Figure 10 shows a standard curve for an immuno-enzymatic assay to detect soluble TREM-1 in the sera of patients with suspected sepsis
  • Figure 11 shows the kinetics of immuno-enzymatic assay to detect soluble TREM-1 (panel A) and the cytofluorimetric analysis of TREM-1 Ligand (panel B) in a patient having SIRS without infection (HSR34) and in a sepsis patient (HSR37).
  • FIG. 16 shows TREM-1 expression pattern on monocytes during septic shock according to outcome. Results are expressed as Mean Fluorescence Intensity. Respective p values are depicted above time points. 'Baseline' corresponds to the first determination and 'Last value' to the last determination of TREM-1 before intensive care unit discharge or death.
  • EXAMPLE 1 Production of antibodies against TREM-1 Receptor which are capable of binding sTREM-1
  • Antibodies were raised against a fusion protein of the TREM-1 receptor with the human lgG Fc region.
  • the cDNA fragment encoding the TREM-1 extracellular region was amplified by PCR and cloned into an expression vector containing the exons for hinge, CH2, and CH3 region of human lgG1 (see Bouchon et al. The Journal of Immunology, 2000, 164: 4991-4995). Briefly, the 760-bp TREM-1 was amplified by RT-PCR, cloned into pCR2.1 (Invitrogen, Carlsbad, CA), and sequenced.
  • PCR primers used were: 5'-GCTGGTGCACAGGAAGGATG [SEQ ID NO: 3] 3'-GGCTGGAAGTCAGAGGACATT [SEQ ID NO: 4]
  • This chimeric gene was transfected into mouse myeloma cell line J558L, screening of culture supernatants, and purification of TREM-1-Fc can then be performed, as previously described (Traunecker, et al., 1991, Trends Biotechnol. 9:109)).
  • Anti-TREM-1 monoclonal antibodies were produced by immunising BALB/c mice with TREM-1-Fc .
  • mice received an initial injection of 100 ⁇ g of TREM-1 -Fc fusion protein (TREM-1 -Fc), mixed 1:1 (vol/vol) with Alu-Gel-S (Serva Biochemicals, Paramus, NJ), behind the neck.
  • TREM-1 -Fc TREM-1 -Fc fusion protein
  • Alu-Gel-S Serva Biochemicals, Paramus, NJ
  • mice were sacrificed and draining lymph node cells were isolated and fused with the myeloma fusion partner, Ag8.653, using polyethylene glycol 4000.
  • Hybridoma supernatants were screened in two steps. First, an ELISA was performed using TREM-1 -Fc in the coating step and human-adsorbed alkaline phosphatase-labeled goat anti-mouse lgG as secondary antibody. Supernatants from clones that were positive in ELISA were then tested by FACS ® analysis for staining cells by flow cytometry.
  • EXAMPLE 2 Rapid detection of the soluble form of TREM-1 (sTREM) in the diagnosis of pneumonia
  • SAPS II score SAPS II score
  • SOFA score body temperature
  • leukocyte count ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (Pa0 2 /FiO z ); serum levels of C reactive protein and procalcitonin
  • presence of shock defined as systolic arterial pressure lower than 90 mm Hg with signs of peripheral hypoperfusion or need for continuous infusion of vasopressor or inotropic agents
  • duration of previous mechanical ventilation and use of previous antimicrobial therapy.
  • a clinical pulmonary infection score (CPIS) was calculated as previously described in Pugin J, et al. Am Rev Respir Dis 1991;143:1121-9. The duration of mechanical ventilation, length of ICU stay and ICU mortality were also recorded.
  • Mini-bronchoalveolar lavages (BAL) and microbiological specimen processing were performed as described in detail in Papazian L et al. Am J Respir Crit Care Med 1995;152:1982-91 and Duflo F et al. Anesthesiology 2002;1:74-9. Briefly, mini-bronchoalveolar lavage was performed using the Combicath, a single-sheathed, 50-cm, sterile, plugged, telescopic catheter (Plastimed, St Leu La Foret, France).
  • the recovered BAL fluid (13+3mL out of 20mL of instilled saline serum) was divided into two samples: one was used for direct microscopic examination and quantitative culture; the other was centrifuged at 10000 revolutions per minute for 30 min and the supernatant was frozen at -80°C until used for sTREM-1 and cytokine measurements.
  • concentration of micro-organisms considered significant for the potential diagnosis of pneumonia was >10 3 CFU/mL of BAL fluid.
  • Post hoc diagnosis of pneumonia was made from a combination of already mentioned clinical criteria with microbiological evidence of microbial infection. These criteria were similar to those used for ventilator-associated pneumonia described in Pugin J et al. Am Rev Respir Dis 1991;143:1121-9.
  • Pneumonia was considered to be absent when an alternative cause for pulmonary infiltrate was established and there was non-significant bacterial growth in culture of BAL in association with full recovery from fever, infiltrate, and leukocytosis without antimicrobial therapy.
  • Two intensivists reviewed all medical records pertaining to the patient and independently classified the diagnosis as community-acquired pneumonia, ventilator-associated pneumonia or no pneumonia. A consensus concerning the diagnosis was achieved in all cases. Both intensivists were blinded to the results of sTREM-1 and cytokines levels.
  • sTREM-1 and cytokines assays Assessment of sTREM-1 levels in BAL fluid samples was performed using an immunoblot technique with 21C7, a monoclonal murine lgG1 directed against human TREM-1 prepared as described in Example 1 . Briefly, 100 ⁇ L of each BAL fluid supernatant was dotted on a nitrocellulose membrane, dried, and overcoated in phosphate buffer-saline (PBS) supplemented with 3% bovine serum albumin. The nitrocellulose sheet was then incubated for 60 min in the presence of diluted 1:2000 diluted 21 C7.
  • PBS phosphate buffer-saline
  • the sheet was further incubated for 60 min with diluted 1:1000 diluted goat anti- mouse immunoglobuiins (Dako, Glostrup, Denmark), washed in PBS supplemented with 20% dimethylsulfoxyde and incubated for 30 min with diluted 1: 000 diluted horseradish peroxydase-conjugated streptavidin (Bio- Rad, Cergy, France).
  • the enzyme substrate chromogen Opti-4CN Bio-Rad was then added, and colour developed in proportion to the amount of sTREM- 1 bound to the membrane.
  • Each sheet also contained calibration samples of a known concentration of sTREM-1 (0 to 200 pg/mL).
  • sTREM-1 concentration from each sample was determined by comparing the optical densities of the samples to the standard curve. All measurements were performed in duplicate and results are expressed as the mean concentration in picograms per millilitre of bronchoalveolar lavage fluid.
  • the sensitivity of this technique allows the detection of sTREM-1 level as low as 5 pg/mL and the entire procedure takes less than 3 hours. The coefficient of variation of the assay was lower than 5 percent.
  • Tumor necrosis factor- ⁇ and interleukin-1 ⁇ were determined in BAL fluid by solid-phase ELISA method according to the recommendations of the manufacturer (BD Biosciences, Le Pont de Claix, France). The sensitivity of the technique allows the detection of levels as low as 2 pg/mL for tumor necrosis factor- ⁇ and 3.9 pg/mL for interleukin-1 ⁇ .
  • ROC Receiver-operating- characteristic
  • SAPS II scoref mean 52 ⁇ 17 53 ⁇ 20 50 ⁇ 15 53 ⁇ 1 0.76 ( ⁇ SD)
  • Acute respiratory failure Neurologic 41 (27.7) 7 (18) 15 (33) 19 (30) 0.45 Shock 37 (25) 6 (16) 16 (35) 15 (23) 0.18 Miscellaneous 28 (19) 2 (5) 11 (24) 15 (23) 0.08
  • ARDS Acute respiratory distress syndrome
  • ARDS of extra-pulmonary origin
  • Clinical characteristics of the three groups did not differ significantly at inclusion (table 1).
  • P values are comparisons between CAP, VAP and NP groups
  • Monomicrobial pneumonia n(%) 36 (95) 37 (80) Polymicrobial pneumonia, n(%) 2 (5) 9 (20) Total number of pathogens*, n 40 58 Bacilli, n(%) Pse ⁇ domonas aeruginosa 12 (20.7) Haemophilus influenzae 10 (25) 10 (17.2) Acinetobacter baumanii 4 (6.9) Serratia marcescens 6 (10.3) Kiebsiella species 11 ((22..55)) 6 (10.3) Legionnella pneumophilia 3 (7.5) Miscellaneous 2 (5) 2 (3.4)
  • Organisms shown are those that were isolated at significant concentrations from quantitative cultures of bronchoalveolar lavage fluid (>10 3 colony-forming units/mL). Legionnella pneumophilia infection was diagnosed by the detection the soluble urinary antigen.
  • sTREM-1 Tumor Necrosis Factor- ⁇ and lnterleukin-1 ⁇ levels
  • the levels of sTREM-1 were higher in BAL fluid from community- acquired and ventilator-associated pneumonia patients than from no- pneumonia patients (P ⁇ 0.001) but did not differ significantly between community-acquired and ventilator-associated pneumonia patients (figure 3).
  • Tumor necrosis factor- ⁇ and interleukin-1 ⁇ levels showed the same trend (P ⁇ 0.001) but with a large overlap of values.
  • sTREM-1 was detected in BAL fluid among 36 out of 38 community-acquired pneumonia patients (sensitivity: 95 percent, 2 false negatives), 46 out of 46 ventilator-associated pneumonia patients (sensitivity: 100 percent), and in 6 out of 64 no-pneumonia patients (6 false positives).
  • sensitivity 95 percent, 2 false negatives
  • 46 out of 46 ventilator-associated pneumonia patients sensitivity: 100 percent
  • 6 out of 64 no-pneumonia patients 6 false positives.
  • the capacity of sTREM-1 to differentiate pneumonia from no pneumonia was assessed with a ROC curve analysis (figure 4).
  • the area under the ROC curve when sTREM-1 was used to differentiate pneumonia from no pneumonia was 0.93 (95 percent Cl 0.92 to 0.95, PO.001).
  • a sTREM-1 cut-off value of 5 pg/mL (which represented the technique's threshold of detection) had a sensitivity of 98 percent (95 percent Cl, 95 to 100) a specificity of 90 percent (95 percent Cl, 84 to 96).
  • the Inventors determined that the presence of sTREM-1 in BAL fluid was the strongest independent predictor of pneumonia with an odds ratio of 41.52 (table 5).
  • the best clinical predictor of pneumonia was a clinical pulmonary infection score >6 (odds ratio: 2.98).
  • Table 5 Multiple logistic-regression analysis of factors used for differentiating between patients with and those without pneumonia PREDICTOR P Value ODDS RATIO (95% Confidence Interval) CPIS * >6 0.002 2.98 (1.51 to 5.86) BALTNF ⁇ >150 pg/mL 0.004 2.44 (1.82 to 5.75) BAL lL-1 ⁇ >75 pg/mL 0.003 2.70 (1.97 to 13.18) BAL sTREM-1>5 pg/mL ⁇ 0.001 41.52 (20.90 to 77.62) CPIS: clinical pulmonary infection 1 score « These results demonstrate that rapid detection of the sTREM-1 in bronchoalveolar lavage fluid improves the ability of clinicians to differentiate patients with bacterial or fungal pneumonia from those without pneumonia.
  • the immunoblot technique is rapid, accurate, of very low cost and can be applied to small series or even individual samples. Use of this test to detect the presence of sTREM-1 in bronchoalveolar lavage fluid will lead to more accurate diagnoses of pneumonia in mechanically ventilated patients. Microbiological documentation was obtained in all cases of community-acquired and ventilator-associated pneumonia. When pneumonia was considered to be absent, either a non-infectious alternative cause for pulmonary infiltrate was established or patients fully recovered from fever, infiltrate, and leukocytosis without antimicrobial therapy.
  • EXAMPLE 3 Diagnostic value of plasmatic levels of the soluble form of triggering receptor expressed on myeloid cells (TREM)-1 in critically ill patients with suspected sepsis
  • Clinically suspected infection was defined as an explicit statement by the attending physician indicating the suspicion of an ongoing infection, combined with the initiation of a diagnostic work-up to identify or rule out infection and the prescription of antimicrobial therapy.
  • Patients were not enrolled if they were older than 80 years of age or were immunocompromised (treatment with corticosteroids, bone marrow or organ transplant recipients, leukopenia [white blood cells count ⁇ 1 G/L] or neutropenia [polymorphonuclear granulocyte count ⁇ 0.5 G/L], hematologic malignancy or acquired immune deficiency syndrome). Patients who presented with early death or discharge (within 12 hours after admission) or complete absence of antimicrobial treatment were also excluded. Patients originated either from the emergency room, the general wards, or from the operating room. Approval of the institutional review board and informed consent from patients or their relatives were obtained before inclusion.
  • Plasmatic PCT concentrations were measured using an immunoassay with a sandwich technique and a chemiluminescent detection system, according to the manufacturer's protocol (LumiTest; Brahms Diagnostica, Berlin, Germany). Assessment of plasmatic sTREM-1 levels was performed as described in Example 2.
  • the sheet was further incubated for 60 min with 1:1000 diluted goat anti-mouse immunoglobulins (Dako, Glostrup, Denmark), washed in PBS supplemented with 20% dimethylsulfoxyde and incubated for 30 min with 1 :1000 diluted horseradish peroxydase-conjugated streptavidin (Bio-Rad, Cergy, France).
  • the enzyme substrate chromogen Opti-4CN Bio- Rad
  • Each sheet also contained calibration samples of a known concentration of sTREM-1 (0 to 5000 ng/mL).
  • sTREM-1 concentration from each sample was determined by plotting the optical densities of the samples to the standard curve. All measurements were performed in duplicate and results expressed as mean concentration in nanograms per mL of plasma. The sensitivity of this technique allows the detection of sTREM-1 levels as low as 5 ng/mL and the entire procedure takes less than 3 hours. The coefficient of variation of the assay was lower than 5 percent.
  • the predictors included clinical and laboratory findings along with information on plasmatic sTREM-1 level.
  • logistic regression analysis which requires binary outcome events, subjects classified as confirmed sepsis, severe sepsis, or septic shock (sepsis syndrome) were compared to patients with SIRS and initial suspicion of infection.
  • Receiver-operating-characteristic (ROC) curves were constructed to illustrate various cut-off values of sTREM-1, PCT and CRP. Sensitivity, specificity, and positive and negative predictive values of each parameter were calculated according to standard methods. These values were calculated for the cut-off that represented the best discrimination as derived from the areas under ROC curves. Analysis was completed with Statview software (Abacus Concepts, Berkeley CA) and a two- tailed P ⁇ 0.05 was considered significant.
  • Diagnosis was established as SIRS in 29 patients (38 %), sepsis or severe sepsis (grouped as 'Sepsis') in 22 patients (29 %) and septic shock in 25 patients (33 %).
  • Clinical characteristics did not differ significantly at inclusion between septic and non- septic patients (Table 6).
  • Infections were microbiologically proven in 40 of 49 infected patients (82 %) with 55 % Gram-negative, 42 % Gram-positive bacteria, and 3 % fungal infections. The major sources of infection were the respiratory tract (55 %) and abdomen (22 %). Twenty-four percent of infected patients had a documented bloodstream infection. Neither site of infection nor microbial strains differed between surviving and non-surviving patients (Table 7).
  • Baseline Plasmatic Levels of CRP, PCT and sTREM-1 Baseline plasmatic levels of CRP, PCT and sTREM-1 were higher among septic patients than among subjects with SIRS only (Table 6, Figure 7). Plasmatic sTREM-1 levels appeared to be most helpful in differentiating patients with sepsis from those with SIRS. Mean plasmatic sTREM-1 levels on admission were 229 ng/mL for SIRS; 1836 ng/mL for sepsis and 1413 ng/mL for septic shock (PO.001). The accuracy of the candidate parameters to distinguish patients with SIRS from those with septic conditions was highly variable (Table 8).
  • plasmatic sTREM-1 levels yielded the highest discriminative value with an area under the ROC curve (AUC) of 0.97 (95 % confidence interval [Cl], 0.94 to 1.0) followed by PCT (AUC, 0.85; Cl, 0.81 to 0.89) and CRP (AUC, 0.77; Cl, 0.69 to 0.85; p ⁇ 0.001).
  • AUC area under the ROC curve
  • PCT AUC, 0.85; Cl, 0.81 to 0.89
  • CRP AUC, 0.77; Cl, 0.69 to 0.85; p ⁇ 0.001
  • sTREM-1 yielded a sensitivity of 96 % (95 % Cl, 0.92 to 100 %) and a specificity of 89 % (Cl, 82 to 95 %) to differentiate patients with SIRS from those with sepsis or septic shock.
  • CRP area under the ROC curve
  • the study population was large and comprised a diverse group of critically ill adult patients admitted to a medical ICU in various phases of infectious and ⁇ on-infectious conditions, which allowed a generalization of the study findings.
  • the diagnosis was determined by blinded investigators without knowledge of the plasmatic sTREM-1 levels and the patients were classified as having SIRS of non-infectious origin after incorporation of all other available clinical and laboratory data (Bone RC, et al. Chest. 1992;101:1644-55.).
  • the inventors' study was designed as a real-life study, not including control patients without suspected infection but only patients with a high pre-test probability of sepsis, covering the spectrum of patients that is likely to be encountered in the future use of this test.
  • Example 4 Use of an immuno-enzymatic assay to detect soluble TREM-1 in the sera of patients with suspected sepsis
  • the method is as follows:
  • Coating buffer carbonate pH 9,6: 0.015 M Na 2 C0 3 (0.794g in 500 ml H 2 0), 0.035 M NaHC0 3 (1 ,47g in 500 ml H 2 0)
  • Wash buffer 0.1% Tween 20 in PBS, pH 7,4
  • Substrate solution 30 mM potassium citrate ,pH 4.1, immediately before use, add 1 tablet of 3,3',5,5'-Tetramethylbenzidine (Sigma # T-3405) for 10 ml of buffer and add 2,5 ⁇ l of H 2 0 2 30%.
  • Time 0 represents the day of admission into the Intensive Care Unit. Samples were obtained every 48 hours until day 15.
  • FIG. 12 shows the time course of median (with interquartile range) plasma levels of sTREM-1 in surviving (squares) and non-surviving (triangles) patients.
  • monocyte see Figure 14
  • polymorphonuclear see Figure 15
  • TREM-1 cell surface expression of TREM-1 was analysed in flow cytometry after labeling with a mouse monoclonal antibody anti-human TREM- 1, PE-labelled (clone 193015, R&D, Abingdon, UK). Results were expressed as Mean Fluorescence Intensity (MFI).
  • MFI Mean Fluorescence Intensity
  • MFI on monocytes from patients with septic shock are significantly higher than MFI from non-septic patients or healthy controls.
  • FIG. 16 shows TREM-1 expression pattern on monocytes during septic shock according to outcome. Results are expressed as Mean Fluorescence Intensity. Respective p values are depicted above time points. 'Baseline' corresponds to the first determination and 'Last value' to the last determination of TREM-1 before intensive care unit discharge or death. These results demonstate that among patients with sepsis, those with lower levels of TREM- 1 expression on monocytes, but not on neutrophils, can be predicted to have a positive outcome.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention concerns a method of diagnosing disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of Strem-1 in a biological sample obtained from said subject.

Description

METHOD OF DIAGNOSING INFECTIOUS DISEASE BY MEASURING THE LEVEL OF SOLUBLE TREM-1 IN A SAMPLE
This invention relates generally to the field of immunology. More particularly, the present invention relates to inflammation and the use of markers that allow the prompt diagnosis of infectious disease (for example of bacterial or fungal) origin and the follow up of infected patients during pharmacological treatment. These markers have particular applications in the diagnosis of pneumonia and sepsis.
The diagnosis and treatment of infectious pneumonia in ventilated patients remain a challenge for clinicians. A presumptive clinical diagnosis of pneumonia is often made when a patient develops a new radiographic infiltrate associated with fever, leukocytosis and purulent tracheal secretions and when micro-organisms are isolated from the airways. Unfortunately, many non- infectious processes may be responsible for fever and new pulmonary infiltrates in mechanically ventilated patients and then, clinical approaches lead to an overestimation of the incidence of pneumonia. Moreover, whatever the microbiological diagnostic procedure chosen, it requires further laboratory processing with unavoidable delays of 24 to 48 hours before obtaining definitive quantitative microbial culture results. Meanwhile, clinicians often feel uncomfortable about the diagnosis and, in many cases, unneeded antibiotics are administered while waiting for laboratory results. Therefore, many biological markers have previously been studied to improve the rapidity and performance of the diagnosis procedure but with disappointing results. In the Examples herein the Inventors describe a rapid detection test of the soluble form of the human TREM-1 receptor (sTREM-1) in bronchoalveolar fluid of mechanically ventilated patients to accurately diagnose bacterial or fungal pneumonia. Many non-infectious processes lead to fever and new pulmonary infiltrates in the mechanically ventilated patient, rendering the diagnosis of pneumonia (and especially ventilator-associated pneumonia) very challenging. The systemic signs of infection, such as fever, tachycardia, and leukocytosis, are non-specific findings and can be caused by any condition that releases cytokines. Pugin etal. (Am Rev Respir Dis 1991;143:1121-9) combined body temperature, white biood cells count, volume and appearance of tracheal secretions, ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (PaO2/FiO2), chest-X-ray, and tracheal aspirate cultures into a clinical pulmonary infection score (CPIS) and reported that a score >6 was associated with a high likelihood of pneumonia. This was confirmed in the Inventors' study since a clinical pulmonary infection score >6 was the best clinical predictor of pneumonia with an odds ratio of 2.98. However, the diagnostic accuracy of this score remains to be confirmed. In terms of clinical decision-making in patients in whom pneumonia is suspected, the major problem with the microbiological diagnostic procedure chosen, which is still matter of debate, is that it requires samples cultures, which implies waiting for at least 24 to 48 hrs after sampling. During this delay, the uncertainty of the clinician towards the patient's diagnosis often leads to the prescription of unneeded antibiotics. However, the use of empirical broad- spectrum antibiotics in patients without infection is potentially harmful, facilitating colonization and superinfection with multiresistant bacteria and has been shown to be correlated with an increased length of hospital stay and therefore increased hospital costs. In addition, antibiotic overuse in such critically ill patients delays the proper diagnosis and treatment of the true cause of fever and pulmonary infiltrate. Many biological markers have been studied in the hope to improve the rapidity and performance of the diagnosis procedure. Among them, serum C reactive protein and procalcitonin have been disappointing in critically ill patients. Similar results have been obtained in the Inventors' studies with no significant differences between pulmonary infected patients and non-infected patients. When anatomical and mechanical defence mechanisms preventing micro-organisms from reaching alveoli are overwhelmed, a complex host response develops. This response comprises the activation, by microbial products, of alveolar macrophages which locally release multiple endogenous mediators. Among these mediators, tumor necrosis factor-α (TNFα), interleukin-1β (IL-1β) and other cytokines have been demonstrated to be increased in various types of pulmonary infectious challenges with potential prognostic implications. However, in agreement with other studies, (for example see Monton C et al. Crit Care Med 1999;9:1745-53).the Inventors were unable to determine accurate discriminating cut-off level of such mediators for the diagnosis of pneumonia. The Inventors, using an easy-to-perform immunoblot technique, demonstrate herein that a soluble form of TREM-1 (sTREM-1) is released locally in the bronchoalveolar lavage fluid from patients suffering from pneumonia with a sensitivity of at least 98 percent. In striking contrast, sTREM-1 was detected in only 6 out of 64 patients without pneumonia. Bronchoalveolar lavage fluid levels of sTREM-1 were not correlated to any of the clinical or biological parameters tested and stood as an independent parameter of high specificity. In a multiple logistic regression analysis, presence of sTREM-1 in bronchoalveolar lavage fluid was shown to be the best predictor of pneumonia with an odds ratio as high as 41.52. Presence of sTREM-1 by itself was more accurate that any clinical findings or laboratory values in identifying the existence of bacterial or fungal pneumonia. Thus rapid detection of sTREM-1 in bronchoalveolar lavage fluid is useful in establishing or excluding the diagnosis of bacterial or fungal pneumonia. Sepsis is a common cause of morbidity and mortality in intensive care units (ICUs). Clinical and laboratory signs of systemic inflammation including changes in body temperature, tachycardia or leukocytosis are neither sensitive nor specific enough for the diagnosis of sepsis and can be misleading because critically ill patients often present a systemic inflammatory response syndrome (SIRS) without infection . This issue is of paramount importance owing to the fact that therapy, and outcome differ greatly between patients with and those without sepsis. Moreover, the widespread use of antibiotics for all such patients is likely to increase antibiotic resistance, toxicity and costs. Thus, there is a so far unsatisfied need for clinical or laboratory tools allowing to distinguish between SIRS and sepsis. Among the potentially useful markers of sepsis, procalcitonin (PCT) has been suggested to be the most promising one. Procalcitonin levels have been postulated to be superior to clinical variables or commonly used laboratory tests, such as C-reactive protein (CRP) levels or leukocyte count, and even to correlate with the severity of microbial invasion. However, several investigators have questioned the diagnostic and prognostic accuracy of routine PCT measurements, reporting inconsistent and variable results depending on the severity of illness and infection in the patient population studied. Sepsis constitutes a significant consumption of intensive care resources and remains an ever-present problem in the intensive care unit. It has been estimated that between 400 000 and 500 000 patients are so affected each year in both the USA and Europe. Morbidity and mortality have remained high despite improvements in both supportive and anti-microbial therapies. Mortality rates vary from 40% for uncomplicated sepsis to 80% in those suffering from septic shock and multi-organ dysfunction. The pathogenesis of the conditions is now becoming better understood. Greater understanding of the complex network of immune, inflammatory and haematologica! mediators may allow the development of rational and novel therapies. The condition of sepsis has previously been associated with many terms and nomenclature, reflecting both the complexity of the condition and the similarity of the inflammatory response secondary to other aetiologies. To illustrate the complex nature of sepsis, sepsis has been defined by Edward O. Uthman, MD, as "a constellation of clinical and laboratory findings from which an experienced physician concludes that the patient may have a serious infection". His definition was purposely made as a nebulous, subjective, and tautological definition, because attempts to define "sepsis" in the literature have stirred a great deal of disagreement and qualification. In 1991 , the American College of Chest Physicians and the American Society of Critical Care Medicine published definitions for systemic inflammatory response syndrome (SIRS) and sepsis, with the aim of clarifying the diagnosis and treatment of these conditions and to aid interpretation of research in this field (see Table 1). Table 1: Definitions for the systemic inflammatory response syndrome (S RS) and sepsis
Figure imgf000007_0001
A pattern of physiological variables have been shown in critically ill patients in response to a range of insults including; trauma, burns, pancreatitis and infection. These include inflammatory responses, leucocytosis or severe leucopaenia, hyperthermia or hypothermia, tachycardia and tachypnoea and have been collectively termed the systemic inflammatory response syndrome (SIRS). This definition emphasises the importance of the inflammatory process in these conditions regardless of the presence of infection. The term sepsis is reserved for SIRS when infection is suspected or proven. Sepsis is further stratified into severe sepsis when there is evidence of organ hypoperfusion, made evident by'signs of organ dysfunction such as hypoxaemia, oliguria, lactic acidosis or altered cerebral function. Septic shock is severe sepsis complicated by hypotension defined as systolic blood pressure less than 90 mmHg despite adequate fluid resuscitation. Sepsis and SIRS may be complicated by the failure of two or more organs,, termed multiple organ failure (MOF), due to disordered organ perfusion and oxygenation. In addition to systemic effects of infection, a systemic inflammatory response may occur in severe inflammatory conditions such as pancreatitis and burns. The appearance of signs of an inflammatory response is less well defined following traumatic insults. In the intensive care unit, gram-negative bacteria are implicated in 50 to 60% of sepsis with gram-positive bacteria accounting for a further 35 to 40% of cases. The remainder of cases are due to the less common causes of fungi, viruses and protozoa. Early recognition of sepsis and Systemic Inflammatory Response Syndrome (SIRS) in the critically ill patient may avoid the increased morbidity, mortality and length of stay associated with multiple organ failure. However, there are major problems associated with diagnosis of sepsis and a clear need exists for rapid, reliable and sensitive methods to detect, monitor and treat SIRS due to infectious agents (sepsis). The present invention is directed towards circumventing the existing problems associated with diagnosing sepsis to provide an accurate and consistent method of detection. In the Examples herein the Inventors describe the value of assaying the soluble form of TREM-1 (sTREM-1) in plasma samples of newly admitted critically ill patients with suspected sepsis as a new approach to accurately diagnose infectious processes. Early identification of infection has a major impact on the clinical course, management and outcome of critical patients. Critical care physicians have at their disposal a variety of indicators to serve as a guide in discriminating infectious from non-infectious conditions in newly admitted patients. In some cases, the diagnosis of sepsis becomes clear after completing the medical history and physical examination of a newly admitted patient (Bates DW, et al. Ann Intern Med. 1990;113:495-500). In other circumstances of non-infectious insults causing SIRS (e.g., trauma, haemorrhage, burn, pancreatitis, etc.), the diagnosis of sepsis remains challenging. Efforts have thus been made to identify a reliable marker of infection. However, to date, no single clinical or biological indicator of sepsis has gained widespread acceptance. Among the potentially useful sepsis markers, procalcitonin has been proposed to be the most promising one, but this has been challenged by several authors. In the study described in Example 3 herein, plasmatic sTREM-1 level appears to be the best independent predictor of sepsis. At a cut-off level of 600 ng/mL, the positive and negative predictive values are 94 and 92 % respectively. This study has an important implication for clinicians. As a putative new test to diagnose sepsis upon ICU admission, plasmatic sTREM-1 level assay offers a higher degree of certainty than other currently available candidates. This accuracy can usefully guide physicians in their clinical decision-making and stepwise approach to the complex management of critically ill patients. The immunoblot technique used here can be performed within 3 to 4 hours and may provide valuable information long before blood culture results are back. Moreover, it is of low cost and can be applied to small series or even individual samples. The results reported here demonstrate that rapid measurement of the plasmatic sTREM-1 levels may improve the ability of clinicians to differentiate patients with sepsis from those with systemic inflammation of non-infectious origin. This should be especially useful among patients in whom the diagnosis is not clinically straightforward. The immunoblot technique described is rapid, accurate, of low cost and can be applied to small series or even individual samples. Use of this test to assess plasmatic sTREM-1 levels should lead to a more accurate diagnosis of sepsis in patients admitted in ICUs with a clinical suspicion of infection.
The triggering receptor expressed on myeloid cells-1 (TREM-1) is a member of the Ig-superfamily, the expression of which is up-regulated on phagocytic cells in the presence of bacteria or fungi (Bouchon A et al. Nature 2001;230:1103-7). The inventors have determined that TREM-1 is shed or secreted from the membrane of activated phagocytes and can be found in a soluble form in body fluids and is therefore a useful diagnostic marker. The presence of a soluble form of TREM-1 (sTREM-1) in bronchoalveolar lavage (BAL) fluid from mechanically ventilated patients is shown herein to be a good indicator of infectious pneumonia. Furthermore, as described herein, the use of a plasmatic sTREM-1 assay in a group of severely ill patients admitted with signs of acute, severe inflammation can distinguish sepsis from severe systemic non-infectious inflammation
Accordingly, the present invention provides methods and compositions for the clinical screening and diagnosis of disease of bacterial or fungal origin, for example, pneumonia or sepsis. In addition, the present invention provides methods and compositions for monitoring the effectiveness of the treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis, for selecting participants in clinical trials relating disease of bacterial or fungal origin, for identifying subjects most likely to respond to a particular therapeutic treatment for disease of bacterial or fungal origin and for screening and development of drugs for treatment of disease of bacterial or fungal origin. Thus, in a first aspect the invention provides a method of diagnosis of disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject. Generally, the disease is an inflammatory state, and said method is capable of identifying a microbial origin for said inflammatory state. Examples of such inflammatory states pneumonia and sepsis of bacterial or fungal origin. Thus, in a first embodiment of this aspect, the invention provides a method of diagnosis of pneumonia in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject. In a second embodiment of this aspect, the invention provides a method of diagnosing sepsis of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject. In other words, the invention provides methods of diagnosing or monitoring disease of bacterial or fungal origin, for example, pneumonia or sepsis in a patient, comprising: measuring the level of sTREM-1 in a sample from the patient, wherein the level is an indicator of presence or extent of disease of bacterial or fungal origin in the patient. As stated above sTREM-1 is a soluble form of the TREM-1 Receptor which can be detected in certain body fluid samples by an antibody raised against the TREM-1 Receptor. The term "pneumonia" as defined herein, means, an inflammation of the lung caused by infection by extracellular pathogens such as bacterial infection, and non-bacterial infections (for example, infection by Blastomyces dermatitidis, Histoplasma capsulatum, Coccidioides, Sporothrix schenckii, Pneumocystis carinii, Cryptococcus, Aspergillus, or Mucorsp.), protozoal infections or parasitic infections (for example, those caused by Toxoplasma gondii, Strongyloides stercoralfs, Ascaris, hookworm, Dirofilaria, Paragonimus, or Entamoeba histolytica) where increased expression of sTREM-1 can be detected. Pneumonia includes "Lobar Pneumonia" (which occurs in one lobe of the lung) and Bronchopneumonia (tends to be irregularly located in the lung). Furthermore, pneumonia is often classified into two categories that may help predict the organisms that are the most likely culprits. "Community- acquired (pneumonia contracted outside the hospital). Pneumonia" in this setting often follows a viral respiratory infection. It affects nearly 4 million adults each year. It is likely to be caused by Streptococcus pneumoniae, the most common pneumonia-causing bacteria. Other organisms, such as atypical bacteria called Chlarnydia or Mycoplasma pneumonia are also common causes of community-acquired pneumonia. "Hospital-acquired pneumonia" contracted within the hospital is often called nosocomial pneumonia. Hospital patients are particularly vulnerable to gram-negative bacteria and staphylococci. The term "sepsis of bacterial or fungal origin" as defined herein, means,
SIRS (Systemic Inflammatory Response Syndrome) associated with infection by extracellular pathogens such as bacterial infection, for example bacteremia (the presence of bacteria in the blood) with or without organ failure, and non- bacterial infections, such as fungemia (for example, yeast infection by Candida albicans), protozoal infections or parasitemia (such as in filariasis, malaria, and trypanosomiasis) where increased expression of sTREM-1 can be detected. Without wishing to be bound by theory, the Inventors suspect that sTREM-1 expression is not usually increased in incidences of infection and sepsis caused by intracellular pathogens such as viruses. In this aspect, the measurement of the level of sTREM-1 comprises the steps of (a) contacting said biological sample with a compound capable of binding sTREM-1 ; and (b) detecting the level of sTREM-1 present in the sample by observing the level of binding between said compound and sTREM- 1. The assay or measurement of the sample for the levels of sTREM-1 present in the sample may be carried out using standard protocols known in the art. For example, where the observation of binding between sTREM-1 and the compound capable of binding sTREM-1 takes place, this observation may be carried out using known methodologies. For example the binding may be detected through use of a competitive immunoassay, a non-competitive assay system using techniques such as western blots, a radioimmunoassay, an ELISA (enzyme linked immunosorbent assay), a "sandwich" immunoassay, an immunoprecipitation assay, a precipitin reaction, a gel diffusion precipitin reaction, an immunodiffusion assay, an agglutination assay, a complementfixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, an immunoprecipitation assay, an immunohistochemical assay, a competition or sandwich ELISA, a radioimmunoassay, a Western blot assay, an immunohistological assay, an immunocytochemical assay, a dot blot assay, a fluorescence polarization assay, a scintillation proximity assay, a homogeneous time resolved fluorescence assay, a lAsys analysis, and a BlAcore analysis The determination of the incidence of disease of bacterial or fungal origin, for example, pneumonia or sepsis (depending on the state of the patient and the type of sample) can be undertaken by comparing the levels of sTREM-1 present in the sample with those in a control sample, the median level in a group of control samples (for example, samples from healthy individuals) or with data derived from previous analyses (for example provided as a standard curve or illustration with a diagnostic kit of the invention or data within a computer program, for example associated with a diagnostic means of the invention). The determination of the incidence of of bacterial or fungal origin may comprise deriving the likelihood ratio using a multivariate analysis based on distribution parameters from a set of reference data derived from analysis of the levels of sTREM-1 in patients in which disease of bacterial or fungal origin is absent, present or in remission. The invention therefore also provides diagnostic means capable of measuring levels of sTREM-1 and/or comparing said levels to known levels that are indicative of the disease state of the patient. Such diagnostic means can take the form of a stick test, for example carrying the necessary reagents to perform the method of the invention and to produce, for example, a colorimetric result which can be compared against a colour chart. Other diagnostic means which include a sample measuring means and/or a data processing means containing standard data, as mentioned above, with associated programs for comparing such data with data from a sample are also envisaged. Thus, in the above embodiments, the method according to the first aspect of the invention can comprise the further step of c) correlating the detected level of sTREM-1 with the presence or absence of disease of bacterial or fungal origin, for example, pneumonia or sepsis. For example, a correlation can be made by comparing the measured level of sTREM-1 in the sample with a mean level in samples obtained from a control population of individuals not having disease of bacterial or fungal origin, to indicate the presence or extent of disease of bacterial or fungal origin in the patient. In a further embodiment, the method according to the first aspect of the invention can be used in monitoring the progression or remission of disease of bacterial or fungal origin, in other words, to indicate the progression or remission of the disease. Such methods can be used to monitor the effectiveness and/or progress of therapy in a subject. In this embodiment, the method further comprises the steps of measuring the level of sTREM-1 in a second or further sample from the patient, the first and second or further samples being obtained at different times; and comparing the levels in the samples to indicate the progression or remission of the disease of bacterial or fungal origin. The diagnostic methods according to the present invention are carried out ex vivo. Biological samples for analysis by the methods of the invention can be obtained using methods known in the art from various sources, in particular from body fluids such as whole blood, blood serum, blood plasma, urine and bronchoalveolar lavage fluid. The sample should be a sample treated such that any sTREM-1 present is not removed prior to the assay or is rendered undetectable. Where a patient has symptoms of suspected pneumonia, a preferred biological sample is a sample of bronchoalveolar lavage fluid. Where a patient has symptoms of SIRS, a preferred biological sample is a sample of blood serum. The methods of the invention are applicable to mammals, for example humans, non-human primates, sheep, pigs, cows, horses, goats, dogs, cats and rodents, such as mouse and rat. Generally, the biological sample tested by the methods of the invention is a human sample. The biological sample should generally contain protein molecules from the test subject and is handled such that proteins in the sample are not rendered undetectable by the compound chosen to detect them. In the present application, the term "compound capable of binding sTREM-1" means polypeptides, ligands, antibodies or otherwise discriminating entities which predominantly, preferably specifically, bind to sTREM-1. Such binding compounds, or "sTREM-1 binding partners" can be a naturally occurring sTREM-1 binding molecule, for example a ligand for the TREM-1 Receptor or sTREM-1 and natural and synthetic variants thereof. Further examples of binding compounds include, a chemically modified or genetically modified derivative of a sTREM-1 binding molecule, an artificially (for example chemically produced) sTREM-1 binding molecule or a recombinant or engineered soluble sTREM-1 binding molecule. Included within the scope of the invention are antibodies which bind predominately, preferably specifically or exclusively to, sTREM-1 including, but not limited to, those antibodies which are: mono-or polyclonal antibodies (for example, raised against sTREM-1), bi-specific, multi-specific, human, humanized, chimeric antibodies, single chain antibodies, antibodies derived from phage display techniques, Fab fragments, F(ab')2 fragments, disulfide- linked Fvs, and fragments containing either a VL or VH domain or even a complementary determining region (CDR) that specifically binds to sTREM-1. Otherwise modified immunoglobulins are also included within the scope of the invention, for example a fusion of the TREM-1 -Receptor to one or more immunoglobulin-derived protein domains, for example to confer solubility and/or stability, for example human IgG or IgM Fc fragments. In addition, substances or products mimicking the tertiary structure of a ligand for the TREM-1 -Receptor can be used as binding partners specific for sTREM-1. It is possible to design such on the basis of computer modelling. The product can be produced synthetically using chemical means. Use of recombinant DNA technology to engineer the required structure is also possible as is chemical modification. Furthermore, it is envisaged that isolated TREM-1 -Receptor or sTREM-
1, or computer modelling using the structure of TREM-1 -Receptor or sTREM- 1, may be used to produce binding partners specific for sTREM-1 using methods known in the art. In a preferred embodiment, a compound capable of binding sTREM-1 is an antibody raised against the TREM-1 receptor, a fragment thereof or a variant thereof, provided that it is capable of binding sTREM-1. For example, such an antibody is one raised against TREM-1 human Fc (TREM-1 -Fc) fusion protein (see Example 1 herein). According to a second aspect of the invention there is provided, compounds and pharmaceutical compositions for use in the diagnosis, prognosis, treatment or monitoring of the treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis. In one embodiment of this second aspect, the invention provides a compound capable of binding sTREM-1 for use in the diagnosis, prognosis, treatment or monitoring of disease of bacterial or fungal origin, for example, pneumonia or sepsis. In another embodiment, the invention provides use of a compound capable of binding sTREM-1 in a method of treatment or diagnosis of disease of bacterial or fungal origin, for example, pneumonia or sepsis. In a further embodiment, the invention provides use of a compound capable of binding sTREM-1 in the manufacture of a medicament for the diagnosis, prognosis, treatment or monitoring of the treatment disease of bacterial or fungal origin, for example, pneumonia or sepsis. The methods described herein can furthermore be used as screening assays to identify a subject with, or at risk of developing, disease of bacterial or fungal origin, for example, pneumonia or sepsis. Such assays can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat disease of bacterial or fungal origin. For example, such methods can be used to determine whether a subject can be effectively treated with a specific agent or class of agents (e.g., antibacterial or antifungal agents). Thus, the present invention provides methods for determining whether a subject can be effectively treated with an agent for disease of bacterial or fungal origin, for example, pneumonia or sepsis in which a test sample is obtained and TREM-1 is detected. A further embodiment of the invention provides a pharmaceutical composition comprising a compound capable of binding sTREM-1 together with a pharmaceutically acceptable diluent, carrier orexcipient for use in the diagnosis or treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis. Accordingly, also provided is the use of a compound capable of binding sTREM-1 in a method of treatment or diagnosis of disease of bacterial or fungal origin, for example, pneumonia or sepsis. In other words, the use in diagnosis and treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis, of a compound capable of binding sTREM-1. The invention also provides a compound capable of binding sTREM-1 for use in, or used in, a method of diagnosis or treatment of disease of bacterial or fungal origin, for example, pneumonia or sepsis. As used herein the language "pharmaceutically acceptable diluent, carrier or excipient " is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. Pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration A third aspect of the invention provides a method of identifying agonists or antagonists of sTREM-1 said method comprising comparing the level of binding in a sample containing said sTREM-1 and a compound capable of binding sTREM-1, in the presence and absence of a compound to be tested. Also provided by are agonists or antagonists of sTREM-1 identified according to the method of this aspect of the invention. Also provided is a method of screening compounds for use in the therapy of disease of bacterial or fungal origin, for example, pneumonia or sepsis, comprising determining the effect of those compounds on levels of sTREM-1 present in samples brought into contact with said compounds. Accordingly, the invention also provides a method of treating disease of bacterial or fungal origin, for example, pneumonia or sepsis, in a subject, which method comprises administering to an individual in need thereof an effective amount of an inhibitor of expression or activity of sTREM-1. In a fourth aspect, the invention provides kits, associated reagents and contacting means. In one embodiment the invention provides a kit comprising at least one compound capable of binding sTREM-1 and reagents for detecting binding of said compound to sTREM-1. One embodiment provides a kit comprising at least one compound capable of binding sTREM-1 and means for contacting said compound with a sample containing sTREM-1. For sTREM-1 binding compound-based kits, the kit can comprise, for example: (1) a binding compound (e.g., attached to a solid support) that binds to sTREM-1; and, optionally, (2) a second, different binding compound e.g. an antibody, which binds to either the sTREM-1 or the first binding compound and is conjugated to a detectable agent. Such kits can also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent. The kit can also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate). The kit can also contain a control sample or a series of control samples which can be assayed and compared to the test sample contained. Each component of the kit is usually enclosed within an individual container, and all of the various containers are within a single package, along with instructions for determining whether the subject from which. the sample is derived is suffering from or is at risk of developing disease of bacterial or fungal origin, for example, pneumonia or sepsis. As discussed above "an antibody raised against the TREM-1 -Receptor, a fragment thereof or a variant thereof can function as a compound c pable of binding sTREM-1. Antibodies are preferably raised against the human TREM-1 -Receptor (triggering receptor expressed on myeloid cells) for which the cDNA sequence is given in [SEQ ID NO:1]. The TREM-1 -Receptor is expressed on human myeloid cells, is a transmembrane protein of the immunoglobulin superfamily (Ig-SF). The TREM-1 -Receptor is a transmembrane glycoprotein having the amino acid sequence of [SEQ I D NO:2] that is selectively expressed on blood neutrophils and a subset of monocytes but not on lymphocytes and other cell types. Accordingly, the invention encompasses antibodies raised against isolated or recombinantly prepared TREM proteins or polypeptides or fragments, homologues, derivatives, or variants thereof, as defined herein, as "TREM-1 -Receptor-derived polypeptides" In accordance with the definition of "compound capable of binding sTREM-1", such antibodies raised against "TREM-1 -Receptor-derived polypeptides" predominantly, preferably specifically, bind sTREM-1. Such antibodies may be tested for binding with cells expressing the TREM-1 receptor and preferably also a sample from a patient known to have been suffering from pneumonia or sepsis of bacterial or fungal origin. The term "homologue," especially "TREM-1 -Receptor homologue" as used herein refers to any member of a series of peptides to which antibodies capable of binding sTREM-1 can be raised-. TREM-1 -Receptor homologues can be from either the same or different species of animals. The term "variant" as used herein refers either to a naturally occurring allelic variation of a given peptide or a recombinantly prepared variation of a given peptide or protein in which one or more amino acid residues have been modified by amino acid substitution, addition, or deletion. The term "derivative" as used herein refers to a variation of given peptide or protein that are otherwise modified, i.e., by covalent attachment of any type of molecule, preferably having bioactivity, to the peptide or protein, including non-naturally occurring amino acids. The human TREM-1 -Receptor cDNA is 884-nucleotide long (Fig. 1; [SEQ ID NO:1]) and the open reading frame of TREM-1 -Receptor is nucleotides 48 to 752 of [SEQ ID NO:1], which encodes a transmembrane protein comprising the 234 amino acid sequence shown in Fig. 2 [SEQ 1 D NO:2]. The human TREM-1 -Receptor cDNA can be found in the GenBank database under accession number AF 196329. The putative transmembrane domain starts from amino acid residues 201 to 229 of [SEQ ID NO:2J and contains a charged lysine residue at position 217. Its cytoplasmic tail consists of 5 amino acid residues and appears to contain no signaling motifs. In a particular and preferred embodiment, antibodies for binding sTREM-1 are raised against a TREM-1 -Receptor-derived polypeptide comprising at least an extracellular domain comprising amino acid residues 17 to 200 of [SEQ ID NO:2]. In addition to the antibodies described above, other antibodies suitable for use in the invention are those antibodies having the ability to bind sTREM- 1 which are raised against homologues of TREM-1-Receptor from either the same or different species of animal, preferably from mammals, more preferably from rodents, such as mouse and rat, and most preferably from human. Homologues of the TREM-1 -Receptor nucleic acid molecule (i.e., [SEQ ID NO:1]) can be isolated based on their close nucleotide sequence identity to the human nucleic acid molecules disclosed herein, by standard hybridization techniques under stringent or moderately stringent conditions, as defined herein below, using the human cDNA of the invention or a portion thereof as a hybridization probe. Aspects of fiie invention can be also applied in the framework of multiple diagnosis of a subject. For example, in a method of screening a patient for presence or susceptibility to disease, comprising performing a plurality of diagnostic tests on a tissue sample from the patient for a plurality of diseases, the invention provides the improvement wherein one of the diagnostic tests comprises measuring the level of sTREM-1. The various aspects and embodiments of the invention described above also apply to the following: a diagnostic means for detecting disease of bacterial or fungal origin, for example, pneumonia or sepsis; a diagnostic kit comprising such a diagnostic means; a method of treatment of infection, which includes the step of screening an individual for disease of bacterial or fungal origin, for example, pneumonia or sepsis, wherein disease of bacterial or fungal origin is correlated with the levels of sTREM-1 in a sample from said individual, and if disease of bacterial or fungal origin is identified, treating that individual to prevent or reduce the infection; and the use, in the manufacture of means for detecting disease of bacterial or fungal origin, for example, pneumonia or sepsis, of a compound capable of binding sTREM-1. For clarity it should be noted that in the aspects and embodiments of the invention described above, the diagnosis of pneumonia alone or sepsis alone will be inferred by both the detected level of sTREM-1 and the symptoms of the patient. Generally a bronchoalveolar lavage sample from a patient with lung-related symptoms would be used to diagnose pneumonia based upon elevated levels of sTREM-1. A blood serum sample from a patient exhibiting symptoms of SIRS would be used to diagnose sepsis of bacterial or fungal origin based upon elevated levels of sTREM-1. Thus the invention also provides a method of diagnosing disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 and the step of measuring the level of TREM- 1 -Ligand in one or more biological samples obtained from said subject. As described in Example 4 herein, the Inventors have developed an immuno-enzymatic (in this case ELISA) based method for the detection of soluble TREM-1. Thus the invention provides a method of diagnosing disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1 in a biological sample obtained from said subject and wherein the level of sTREM-1 is measured by an immunochemical technique. Examples of such immunochemical techniques are indirect immunofluorescence (I IF), immunoperoxydase (POD), western immunoblotting (WB), radioimmunoprecipitation (RIPA), enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and agglutination assays. In a preferred embodiment an ELISA method, using an anti-human Trem-1 antibody is used to measure the level of sTREM-1. WO2004081233 describes a method of diagnosing bacterial or fungal sepsis in a subject by measuring the level of TREM-1 -Ligand in a biological sample obtained from the subject and compounds capable of binding TREM- 1 -Ligand. The level of TREM-1 -Ligand present in the sample is measured by observing the level of binding between these compounds and TREM-1 -Ligand. As described in Example 4 herein, the Inventors have determined that measurement of both soluble TREM-1 (as described herein) and membrane associated TREM-1 Ligand (as described in WO2004081233) in newly admitted critically ill patients allows the rapid identification those with infection.
Preferred features of each aspect of the invention are applicable to each other aspect, mutatis mutandis.
The present invention will now be described with reference to the following non-limiting examples, with reference to the figures, in which:
Figure 1. shows Human TREM-1 -Receptor cDNA [SEQ ID NO:1].
Figure 2. shows Human TREM-1-Receptor amino acid sequence [SEQ ID NO:2].
Figure 3. shows the levels of sTREM-1 in bronchoalveolar lavage fluid from patients according to diagnosis. Individual values are plotted and the bars represent the means of the values. P<0.001 between CAP and NP and between VAP and NP. NP: patients without pneumonia (n=64); CAP: Community-acquired pneumonia (n=38); VAP: Ventilator-associated pneumonia (n=46) Figure 4. shows receiver-operating-characteristic curves for various cut-off levels of bronchoalveolar lavage fluid sTREM-1, Tumor necrosis factor-α and Interleukin-1β in differentiating between presence and absence of pneumonia. Areas under the ROC curves for: sTREM-1: 0.93 (95% confidence interval, 0.92 to 0.95) Tumor necrosis factor-α: 0.64 (95% confidence interval, 0.62 to 0.69) lnterleukin-1β: 0.69 (95% confidence interval, 0.67 to 0.72)
Figure 5. shows bronchoalveolar lavage fluid (BAL) supernatants examined by Western blot analysis using 21 C7, an anti TREM-1 monoclonal antibody: Lane 1 : positive control (sTREM-1 , 50 pg/mL) Lane 2: BAL supernatant from a patient with pneumonia Lane 3: BAL supernatant from a patient without pneumonia
Figure 6. shows a flow-chart of the patients admitted to the ICU during the study period.
Figure 7. shows admission plasmatic levels of C-Reactive Protein, Procalcitonin and sTREM-1 according to diagnosis. Individual values are plotted and the bars represent the means of the values. P<0.001 between SIRS and Sepsis and between SIRS and Septic Shock: SIRS: patients with systemic inflammatory response syndrome of non- infectious origin (n=29) Sepsis: patients with sepsis or severe sepsis (n=22) Septic Shock: patients with septic shock (n=25)
Figure 8. shows Receiver-operating-characteristic curves for various cut-off levels of plasmatic C-Reactive Protein, Procalcitonin and sTREM-1 in differentiating between presence and absence of infection. Areas under the ROC. curves for: C-Reactive Protein: 0.77 (95% confidence interval, 0.69 to 0.85) Procalcitonin: 0.85 (95% confidence interval, 0.81 to 0.89) sTREM-1: 0.97 (95% confidence interval, 0.94 to 1.0)
Figure 9. shows admission plasmatic levels of C-Reactive Protein,
Procalcitonin and sTREM-1 in patients with sepsis, severe sepsis and septic shock according to outcome. Individual values are plotted and the bars represent the means of the values. P Values are 0.26, 0.64 and 0.05 between Survivors and Non-Survivors for C-Reactive Protein, Procalcitonin and sTREM-1 respectively.
Figure 10 shows a standard curve for an immuno-enzymatic assay to detect soluble TREM-1 in the sera of patients with suspected sepsis
Figure 11 shows the kinetics of immuno-enzymatic assay to detect soluble TREM-1 (panel A) and the cytofluorimetric analysis of TREM-1 Ligand (panel B) in a patient having SIRS without infection (HSR34) and in a sepsis patient (HSR37).
Figure 12 shows the time course of median (with interquartile range) plasma levels of sTREM-1 in surviving (squares) and non-surviving (triangles) patients in a series of 63 patients, some with sepsis (n=30) others with septic shock (n=33).
Figure 13 shows Kapian-Meier analysis of patients with sTREM-1>180 pg/mL (n=32) and <180 pg/mL (n=31). There was a significant difference between the two curves (Log-Rank test, p<0.01).
Figure 14 shows analysis of cell surface expression of TREM-1 in monocytes from septic patients (n=25) and non-septic patients (n=15) or healthy controls (n=7). Results were expressed as Mean Fluorescence Intensity (MFI). Respective p values (Student's t test) are depicted above each scatter plot.
Figure 15 shows analysis of cell surface expression of TREM-1 in polymorphonuclear cells from septic patients (n=25) and non-septic patients (n=15) or healthy controls (n=7). Results were expressed as Mean Fluorescence Intensity (MFI). Respective p values (Student's t test) are depicted above each scatter plot.
Figure 16 shows TREM-1 expression pattern on monocytes during septic shock according to outcome. Results are expressed as Mean Fluorescence Intensity. Respective p values are depicted above time points. 'Baseline' corresponds to the first determination and 'Last value' to the last determination of TREM-1 before intensive care unit discharge or death.
EXAMPLES
EXAMPLE 1: Production of antibodies against TREM-1 Receptor which are capable of binding sTREM-1
Antibodies were raised against a fusion protein of the TREM-1 receptor with the human lgG Fc region. To produce soluble TREM-1 -Fc, the cDNA fragment encoding the TREM-1 extracellular region was amplified by PCR and cloned into an expression vector containing the exons for hinge, CH2, and CH3 region of human lgG1 (see Bouchon et al. The Journal of Immunology, 2000, 164: 4991-4995). Briefly, the 760-bp TREM-1 was amplified by RT-PCR, cloned into pCR2.1 (Invitrogen, Carlsbad, CA), and sequenced. The PCR primers used were: 5'-GCTGGTGCACAGGAAGGATG [SEQ ID NO: 3] 3'-GGCTGGAAGTCAGAGGACATT [SEQ ID NO: 4] This chimeric gene was transfected into mouse myeloma cell line J558L, screening of culture supernatants, and purification of TREM-1-Fc can then be performed, as previously described (Traunecker, et al., 1991, Trends Biotechnol. 9:109)). Anti-TREM-1 monoclonal antibodies (mAbs) were produced by immunising BALB/c mice with TREM-1-Fc . Briefly, 10-wk-old, female BALB/c mice (Iffa-Credo, L'Arbresle, France) received an initial injection of 100 μg of TREM-1 -Fc fusion protein (TREM-1 -Fc), mixed 1:1 (vol/vol) with Alu-Gel-S (Serva Biochemicals, Paramus, NJ), behind the neck. Four weeks later, they were given a booster immunization with the same immunogen, followed after 2 weeks by a final injection of 100 μg of purified TREM-1 -Fc. Three days later, mice were sacrificed and draining lymph node cells were isolated and fused with the myeloma fusion partner, Ag8.653, using polyethylene glycol 4000. Hybridoma supernatants were screened in two steps. First, an ELISA was performed using TREM-1 -Fc in the coating step and human-adsorbed alkaline phosphatase-labeled goat anti-mouse lgG as secondary antibody. Supernatants from clones that were positive in ELISA were then tested by FACS® analysis for staining cells by flow cytometry.
EXAMPLE 2: Rapid detection of the soluble form of TREM-1 (sTREM) in the diagnosis of pneumonia
Materials and Methods Study population Approval of the institutional review board and informed consent from patients or their relatives were obtained before inclusion. All patients 18 years or older hospitalized in the Inventors' medical ICU were prospectively enrolled in the study if they met the following criteria: 1) need for mechanical ventilation; 2) clinical suspicion of infectious pneumonia defined by a newly developed and persistent infiltrate on chest radiography associated with at least one of the following: purulent tracheal secretions, body temperature of at least 38.3 °C, and leukocytosis (>10000/mm3) or ieukopenia (<4000/mm3). Ventilator-associated pneumonia was defined by acquisition of the disease after 48h of mechanicai ventilation. On admission into the ICU, the following items were recorded for each patient: age; sex; severity of underlying medical condition stratified according to the criteria of McCabe and Jackson (McCabe WR, Jackson GG. Arch Intern Med 1982;110:847-64); SAPS II score; Sepsis- related Organ Failure Assessment (SOFA) score (range, 0 to 24, with scores for each organ system [respiration, coagulation, liver, cardiovascular, central nervous system, and kidney] ranging from 0 [normal] to 4 [most abnormal]); and reason for admission into the ICU. The following baseline variables were also recorded at inclusion: SAPS II score; SOFA score; body temperature; leukocyte count; ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (Pa02/FiOz); serum levels of C reactive protein and procalcitonin; presence of shock, defined as systolic arterial pressure lower than 90 mm Hg with signs of peripheral hypoperfusion or need for continuous infusion of vasopressor or inotropic agents; duration of previous mechanical ventilation; and use of previous antimicrobial therapy. A clinical pulmonary infection score (CPIS) was calculated as previously described in Pugin J, et al. Am Rev Respir Dis 1991;143:1121-9. The duration of mechanical ventilation, length of ICU stay and ICU mortality were also recorded.
Confirmation of the diagnosis Mini-bronchoalveolar lavages (BAL) and microbiological specimen processing were performed as described in detail in Papazian L et al. Am J Respir Crit Care Med 1995;152:1982-91 and Duflo F et al. Anesthesiology 2002;1:74-9. Briefly, mini-bronchoalveolar lavage was performed using the Combicath, a single-sheathed, 50-cm, sterile, plugged, telescopic catheter (Plastimed, St Leu La Foret, France). The recovered BAL fluid (13+3mL out of 20mL of instilled saline serum) was divided into two samples: one was used for direct microscopic examination and quantitative culture; the other was centrifuged at 10000 revolutions per minute for 30 min and the supernatant was frozen at -80°C until used for sTREM-1 and cytokine measurements. The concentration of micro-organisms considered significant for the potential diagnosis of pneumonia was >103 CFU/mL of BAL fluid. Post hoc diagnosis of pneumonia was made from a combination of already mentioned clinical criteria with microbiological evidence of microbial infection. These criteria were similar to those used for ventilator-associated pneumonia described in Pugin J et al. Am Rev Respir Dis 1991;143:1121-9. Pneumonia was considered to be absent when an alternative cause for pulmonary infiltrate was established and there was non-significant bacterial growth in culture of BAL in association with full recovery from fever, infiltrate, and leukocytosis without antimicrobial therapy. Two intensivists reviewed all medical records pertaining to the patient and independently classified the diagnosis as community-acquired pneumonia, ventilator-associated pneumonia or no pneumonia. A consensus concerning the diagnosis was achieved in all cases. Both intensivists were blinded to the results of sTREM-1 and cytokines levels.
sTREM-1 and cytokines assays Assessment of sTREM-1 levels in BAL fluid samples was performed using an immunoblot technique with 21C7, a monoclonal murine lgG1 directed against human TREM-1 prepared as described in Example 1 . Briefly, 100μL of each BAL fluid supernatant was dotted on a nitrocellulose membrane, dried, and overcoated in phosphate buffer-saline (PBS) supplemented with 3% bovine serum albumin. The nitrocellulose sheet was then incubated for 60 min in the presence of diluted 1:2000 diluted 21 C7. After thorough rinsing, the sheet was further incubated for 60 min with diluted 1:1000 diluted goat anti- mouse immunoglobuiins (Dako, Glostrup, Denmark), washed in PBS supplemented with 20% dimethylsulfoxyde and incubated for 30 min with diluted 1: 000 diluted horseradish peroxydase-conjugated streptavidin (Bio- Rad, Cergy, France). The enzyme substrate chromogen Opti-4CN (Bio-Rad) was then added, and colour developed in proportion to the amount of sTREM- 1 bound to the membrane. Each sheet also contained calibration samples of a known concentration of sTREM-1 (0 to 200 pg/mL). Colorimetric determination was achieved by means of a reflectance scanner and the Quantity One Quantitation Software (Bio-Rad). sTREM-1 concentration from each sample was determined by comparing the optical densities of the samples to the standard curve. All measurements were performed in duplicate and results are expressed as the mean concentration in picograms per millilitre of bronchoalveolar lavage fluid. The sensitivity of this technique allows the detection of sTREM-1 level as low as 5 pg/mL and the entire procedure takes less than 3 hours. The coefficient of variation of the assay was lower than 5 percent. Tumor necrosis factor-α and interleukin-1β were determined in BAL fluid by solid-phase ELISA method according to the recommendations of the manufacturer (BD Biosciences, Le Pont de Claix, France). The sensitivity of the technique allows the detection of levels as low as 2 pg/mL for tumor necrosis factor-α and 3.9 pg/mL for interleukin-1β.
Statistical analysis Descriptive results of continuous variables were expressed as mean (±SD). The results of BAL sTREM-1 and cytokines levels were expressed as mean (±SD).Variables were tested for their association with diagnosis using Pearson χ2 test for categorial data and Mann-Whitney U test for numerical data. Comparison between the different groups was conducted by using Mann-Whitney U test (or non-parametric Kruskall-Wallis test when appropriate) for numerical data and using Pearson χ 2 test for categorial data. The relations between sTREM-1 and clinical or biological features were assessed using Spearman's correlation test. To evaluate the value of the presence of sTREM-1 in BAL fluid, the Inventors used a multiple stepwise logistic regression model with the use of P value 0.05 or less for entry into the model. The predictors included clinical and laboratory findings along with information on the presence of sTREM-1 in BAL fluid. Receiver-operating- characteristic (ROC) curves were constructed to illustrate various cut-off values of sTREM-1, tumor necrosis factor-α and interleukin-1β. Analysis was completed with Statview software (Abacus Concepts, Berkeley CA) and a two- tailed P<0.05 was considered significant.
Results
Characteristics of the patients 1097 patients were admitted into the ICU. All the 148 patients fulfilling the inclusion criteria were enrolled. The baseline characteristics of the overall study group are shown in table 2.
Table 2: Characteristics of the studied population.
Characteristic All patients Community- Ventilator- No P value (n=148) acquired associated pneumonia pneumonia pneumonia (n=64) (n=38) (n=46)
Age, years (±SD) 60±15 58±17 59±14 62±14 0.53
Sex, n (%) 95 (64) 24 (63) 29 (63) 42 (66)
Male 53 (36) 14 (37) 17 (37) 22 (34) 0.97
Female
McCabe score, mean 1.85±0.95 1.77±0.92 1.81±0.92 1.88±0.91 0.79 (±SD)
History of COPD*, n 39 (26) 9 (23) 2 (26) 18 (28) 0.93
(%)
SAPS II scoref, mean 52±17 53±20 50±15 53±1 0.76 (±SD)
SOFA score§*, mean 7.8±3.9 8.5±4.4 7.0±3.5 8.1 ±4.0 0.43 (±SD)
Reason for admission 42 (28.3) 23 (61) 4 (9) 15 (24) 0.002 n (%)
Acute respiratory failure Neurologic 41 (27.7) 7 (18) 15 (33) 19 (30) 0.45 Shock 37 (25) 6 (16) 16 (35) 15 (23) 0.18 Miscellaneous 28 (19) 2 (5) 11 (24) 15 (23) 0.08
Length of mechanical 14±12 8±7 21 ±19 11±9 O.001 ventilation, days (±SD)
Length of ICU stay, 18±15 11±8 26±21 15±9 <0.001 days (±SD)
Mortality, n (%) 50 (34) 11 (29) 19 (41) 20 (31) 0.58
*COPD: chronic obstructive pulmonary disease fSAPS II: Simplified Acute Physiologic Score II
§ SOFA: Sepsis-related Organ failure Assessment
P values are comparisons between CAP, VAP and NP groups Most of the patients had an associated co-morbidity and 38 (26 percent) had a history of chronic obstructive pulmonary disease (COPD). Mean (±SD) SAPSII and SOFA scores were 52 (±17) and 7.8 (±3.9) respectively. The ICU mortality rate of 34 percent was in agreement with the predictive risk of death based on the SAPSII score (Le Gall JR et al. JAMA 1993;270:2957-63). Diagnosis was established as community-acquired pneumonia (CAP) in 38 patients (26 percent), ventilator-associated pneumonia (VAP) in 46 patients (31 percent) and no pneumonia (NP) in 64 patients (43 percent). Among the NP group, diagnoses were established as follows: Acute exacerbation of COPD (n=11 ); Acute respiratory distress syndrome (ARDS) of extra-pulmonary origin (abdominal or uro-genital sepsis: n=19; pancreatitis: n=6; others: n=4); ARDS of pulmonary origin (near-drowning: n=1; fire smoke inhalation: n=1); Cardiogenic shock (n=12) and Unknown (n=10). Clinical characteristics of the three groups did not differ significantly at inclusion (table 1). Community-acquired pneumonia patients were more often referred to the ICU with acute respiratory failure than others (P=0.002). As expected, the duration of mechanical ventilation and length of ICU stay were higher among ventilator-associated pneumonia patients (P<0.001 ). Mortality did not differ between the three groups. A clinical pulmonary infection score (CPIS) >6 was more frequent in community-acquired and ventilator-associated pneumonia patients than in no-pneumonia patients (P=0.02). Body temperature, leukocyte count, ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (Pao2/Fio2), serum C reactive protein (CRP) and procalcitonin levels did not differ between the three groups (Table 3). Microbial species grew at a significant concentration from BAL (>103
CFU/mL) of all except 2 community-acquired pneumonia patients infected with Legionella pneumophila and of all ventilator-associated pneumonia patients as shown in Table 4. Table 3: Characteristics of the 3 groups of patients at inclusion.
Characteristic Community- Ventilator- No pneumonia P value acquired associated (n=64) pneumonia (n=38) pneumonia (n=46)
Duration of mechanical ventilation before study 0.4±0.2 6.4±8.5 2.1±4.8 <0.001 entry, days (±SD)
Previous antimicrobial 33 (87) 19(41) 30 (47) <0.001 therapy, n(%)
Shock, n(%) 18 (47) 19(41) 30 (47) 0.49
Body temperature, °C (±SD) 37.9±2.0 38.1±0.9 37.7±1.1 0.82
Leukocyte count, cells/mm3 12800±7900 13400±8500 12500±5800 0.99
(±SD)
Pa02/Fi02 *, mmHg (±SD) 181±80 203±67 206±91 0.51
CPIS^β, n(%) 23 (60) 28(61) 22 (34) 0.02
Procalcitonin, ng/mL (±SD) 3.7±1.9 2.6±0.8 2.5±1.2 0.58
C reactive protein, mg/L 197±128 184±108 141±110 0.34
(±SD)
BAL§ fluid TNFα, pg/mL 298.2±47.7 290.5±39.7 147.2±25.1 O.001
(±SD)
BAL§ fluid IL-1β, pg/mL 92.5±22.5 95.1±29.4 41.5±12.5 <0.001
(±SD)
BAL§ fluid sTREM-1, pg/mL 23.2±2.8 33.6±5.1 1.8±0.9 <0.001
(±SD)
*Pa02/Fi02: ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen
+CPIS: clinical pulmonary infection score
§BAL: bronchoalveolar lavage
P values are comparisons between CAP, VAP and NP groups
Table 4: Features and organisms associated with pneumonia Feature or Organism Community-acquired Ventilator-associated pneumonia (n: =38) pneumonia (n=46)
Monomicrobial pneumonia, n(%) 36 (95) 37 (80) Polymicrobial pneumonia, n(%) 2 (5) 9 (20) Total number of pathogens*, n 40 58 Bacilli, n(%) Pseυdomonas aeruginosa 12 (20.7) Haemophilus influenzae 10 (25) 10 (17.2) Acinetobacter baumanii 4 (6.9) Serratia marcescens 6 (10.3) Kiebsiella species 11 ((22..55)) 6 (10.3) Legionnella pneumophilia 3 (7.5) Miscellaneous 2 (5) 2 (3.4)
Cocci, n(%) Staphylococcus aureus 4 (10) 14 (24.1) Streptococcus species 1 (2.5) Streptococcus pneumonia 17 (42.5) 1 (1.7)
Fungi 2 (5) 3 (5.2)
Organisms shown are those that were isolated at significant concentrations from quantitative cultures of bronchoalveolar lavage fluid (>103 colony-forming units/mL). Legionnella pneumophilia infection was diagnosed by the detection the soluble urinary antigen.
sTREM-1, Tumor Necrosis Factor-α and lnterleukin-1β levels The levels of sTREM-1 were higher in BAL fluid from community- acquired and ventilator-associated pneumonia patients than from no- pneumonia patients (P<0.001) but did not differ significantly between community-acquired and ventilator-associated pneumonia patients (figure 3). Tumor necrosis factor-α and interleukin-1 β levels showed the same trend (P<0.001) but with a large overlap of values. Among patients with pneumonia, there was a trend (P=0.07) towards higher sTREM-1 levels in non-survivors than in survivors with 31.2±5.7 pg/mL and 24.9±3.0 pg/mL respectively. There was no correlation between sTREM-1 levels and previous history of chronic obstructive pulmonary disease, amount of inflammatory cells in BAL fluid, microbial species or any other clinical and biological features. Diagnostic value ofsTREM-1 assay The Inventors next determined whether the presence of sTREM-1 in bronchoalveolar lavage fluid could discriminate between presence and absence of pneumonia. Since there was no difference between community- acquired and ventilator-associated pneumonia patients for the following analyses, pooled data are presented. Whatever the level at or above 5pg/mL, sTREM-1 was detected in BAL fluid among 36 out of 38 community-acquired pneumonia patients (sensitivity: 95 percent, 2 false negatives), 46 out of 46 ventilator-associated pneumonia patients (sensitivity: 100 percent), and in 6 out of 64 no-pneumonia patients (6 false positives). Thus, among the whole population of patients, the presence of sTREM-1 in BAL fluid is associated with a likelihood ratio of 10.38. The capacity of sTREM-1 to differentiate pneumonia from no pneumonia was assessed with a ROC curve analysis (figure 4). The area under the ROC curve when sTREM-1 was used to differentiate pneumonia from no pneumonia was 0.93 (95 percent Cl 0.92 to 0.95, PO.001). A sTREM-1 cut-off value of 5 pg/mL (which represented the technique's threshold of detection) had a sensitivity of 98 percent (95 percent Cl, 95 to 100) a specificity of 90 percent (95 percent Cl, 84 to 96). In a multiple logistic regression analysis, the Inventors determined that the presence of sTREM-1 in BAL fluid was the strongest independent predictor of pneumonia with an odds ratio of 41.52 (table 5). The best clinical predictor of pneumonia was a clinical pulmonary infection score >6 (odds ratio: 2.98).
Table 5: Multiple logistic-regression analysis of factors used for differentiating between patients with and those without pneumonia PREDICTOR P Value ODDS RATIO (95% Confidence Interval) CPIS*>6 0.002 2.98 (1.51 to 5.86) BALTNFα>150 pg/mL 0.004 2.44 (1.82 to 5.75) BAL lL-1β>75 pg/mL 0.003 2.70 (1.97 to 13.18) BAL sTREM-1>5 pg/mL <0.001 41.52 (20.90 to 77.62) CPIS: clinical pulmonary infection1 score « These results demonstrate that rapid detection of the sTREM-1 in bronchoalveolar lavage fluid improves the ability of clinicians to differentiate patients with bacterial or fungal pneumonia from those without pneumonia. This should be especially useful among patients in whom the diagnosis is not clinically straightforward. The immunoblot technique is rapid, accurate, of very low cost and can be applied to small series or even individual samples. Use of this test to detect the presence of sTREM-1 in bronchoalveolar lavage fluid will lead to more accurate diagnoses of pneumonia in mechanically ventilated patients. Microbiological documentation was obtained in all cases of community-acquired and ventilator-associated pneumonia. When pneumonia was considered to be absent, either a non-infectious alternative cause for pulmonary infiltrate was established or patients fully recovered from fever, infiltrate, and leukocytosis without antimicrobial therapy. However, the Inventors could not exclude that some patients with a true ventilator- associated pneumonia could have been misclassified in the no-pneumonia group and spontaneously recovered. This could have artificially lowered the specificity of the test and may have been responsible for some of the 6 false- positives in the no-pneumonia group. Finally, and without wishing to be bound by theory, none of the patients tested presented with a viral pneumonia and thus, results are not generalisable to viral infections. EXAMPLE 3: Diagnostic value of plasmatic levels of the soluble form of triggering receptor expressed on myeloid cells (TREM)-1 in critically ill patients with suspected sepsis
Materials and Methods
Study population All consecutive patients newly hospitalized in a teaching hospital medical ICU in France were prospectively enrolled in the study if they had a clinically suspected infection and fulfilled at least two criteria of SIRS (Bone RC, er a/. Chest. 1992;101 :1644-55.). Clinically suspected infection was defined as an explicit statement by the attending physician indicating the suspicion of an ongoing infection, combined with the initiation of a diagnostic work-up to identify or rule out infection and the prescription of antimicrobial therapy. Patients were not enrolled if they were older than 80 years of age or were immunocompromised (treatment with corticosteroids, bone marrow or organ transplant recipients, leukopenia [white blood cells count < 1 G/L] or neutropenia [polymorphonuclear granulocyte count < 0.5 G/L], hematologic malignancy or acquired immune deficiency syndrome). Patients who presented with early death or discharge (within 12 hours after admission) or complete absence of antimicrobial treatment were also excluded. Patients originated either from the emergency room, the general wards, or from the operating room. Approval of the institutional review board and informed consent from patients or their relatives were obtained before inclusion.
Data Collection Upon admission into the ICU, the following items were recorded for each patient: age; sex; severity of underlying medical condition stratified according to the criteria of McCabe and Jackson (Arch Intern Med. 1982;110:847-64); Simplified Acute Physiology Score II (SAPSII) (Le Gall JR et al. JAMA. 1993;270:2957-63); Sepsis-related Organ Failure Assessment (SOFA) score (range 0 to 24, with scores for each organ system [respiration, coagulation, liver, cardiovascular, central nervous system, and kidney] ranging from 0 [normal] to 4 [most abnormal]) (Vincent JL et al. Intensive Care Med. 1996;22:707-10); reason for admission into the ICU; principal diagnosis; vital signs; respiratory parameters; routine blood tests and microbiologic culture results. Survival or death in the ICU was assessed during a follow-up period as long as 28 days. Microbiologic tests and antimicrobial therapy were prescribed by the attending physician according to the usual practice of the ICU without interference by the research team. Two intensivists retrospectively reviewed all medical records pertaining to each patient and independently classified the diagnosis as SIRS, sepsis, severe sepsis, or septic shock at the time of admission, according to established consensus definitions (Bone RC, et al. Chest. 1992;101:1644-55.). Agreement concerning the diagnosis was achieved in all cases. Both intensivists were blinded to the results of plasmatic sTREM-1 values.
Measurements of Procalcitonin and sTREM-1 Plasma Levels Within 12 hours after admission and enrolment in the study, 5 mL of whole heparinized blood was drawn via an arterial line for PCT and sTREM-1 determinations. Plasma was collected by centrifugation at 4°C, aliquoted, and stored at -80°C until the day of assay. Plasmatic PCT concentrations were measured using an immunoassay with a sandwich technique and a chemiluminescent detection system, according to the manufacturer's protocol (LumiTest; Brahms Diagnostica, Berlin, Germany). Assessment of plasmatic sTREM-1 levels was performed as described in Example 2. Briefly, 100μL of each plasma sample was dotted on a nitrocellulose membrane, dried, and overcoated in phosphate buffer-saline (PBS) supplemented with 3% bovine serum albumin. The nitrocellulose sheet was then incubated for 60 min in the presence of monoclonal anti-TREM-1 antibody 21C7, a murine IgG1 directed against human TREM-1, prepared as described in Example 1. After thorough rinsing, the sheet was further incubated for 60 min with 1:1000 diluted goat anti-mouse immunoglobulins (Dako, Glostrup, Denmark), washed in PBS supplemented with 20% dimethylsulfoxyde and incubated for 30 min with 1 :1000 diluted horseradish peroxydase-conjugated streptavidin (Bio-Rad, Cergy, France). The enzyme substrate chromogen Opti-4CN (Bio- Rad) was then added, and colour developed in proportion to the amount of sTREM-1 bound to the membrane. Each sheet also contained calibration samples of a known concentration of sTREM-1 (0 to 5000 ng/mL). Colorimetric determination was achieved by means of a reflectance scanner and the Quantity One Quantitation Software (Bio-Rad). sTREM-1 concentration from each sample was determined by plotting the optical densities of the samples to the standard curve. All measurements were performed in duplicate and results expressed as mean concentration in nanograms per mL of plasma. The sensitivity of this technique allows the detection of sTREM-1 levels as low as 5 ng/mL and the entire procedure takes less than 3 hours. The coefficient of variation of the assay was lower than 5 percent.
Statistical Analysis Descriptive results of continuous variables were expressed as mean (±SD). The results of plasmatic sTREM-1 and PCT levels were expressed as mean (±SD) .Variables were tested for their association with the diagnosis using Pearson χz test for categorial data and Mann-Whitney U test for numerical data. Comparison between the different groups.was conducted by using Mann-Whitney U test (or non-parametric Kruskal-Wailis test when appropriate) for numerical data and using Pearson x 2 test for categorial data. The relations between sTREM-1 and clinical or biological features were assessed using Spearman's correlation test. To evaluate the value of the sTREM-1 plasmatic levels assay, the Inventors used a multiple stepwise logistic regression model. The predictors included clinical and laboratory findings along with information on plasmatic sTREM-1 level. For the purpose of logistic regression analysis, which requires binary outcome events, subjects classified as confirmed sepsis, severe sepsis, or septic shock (sepsis syndrome) were compared to patients with SIRS and initial suspicion of infection. Receiver-operating-characteristic (ROC) curves were constructed to illustrate various cut-off values of sTREM-1, PCT and CRP. Sensitivity, specificity, and positive and negative predictive values of each parameter were calculated according to standard methods. These values were calculated for the cut-off that represented the best discrimination as derived from the areas under ROC curves. Analysis was completed with Statview software (Abacus Concepts, Berkeley CA) and a two- tailed P<0.05 was considered significant.
RESULTS
Characteristics of the Study Population 98 patients were admitted into an ICU with clinical suspicion of infection, of whom 22 were not included in the study because of early death, immunocompromised state, age over 80 years old, absence of consent or protocol violation (Figure 6). The baseline characteristics of the overall study group are shown in table 6. Mean (±SD) SAPSII and SOFA scores were 50.5 (±22.6) and 8.3 (±4.5) respectively. The ICU mortality rate of 26.3 % was in agreement with the predictive risk of death based on the SAPSII score.
Diagnosis was established as SIRS in 29 patients (38 %), sepsis or severe sepsis (grouped as 'Sepsis') in 22 patients (29 %) and septic shock in 25 patients (33 %). Causative conditions of SIRS were as follow: cardiac surgery (n=6); cardiogenic shock (n=5); acute exacerbation of chronic obstructive pulmonary disease (n=5); acute pancreatitis (n=3); heat stroke (n=3); gastrointestinal haemorrhage (n=2); trauma (n=1) and unknown (n=4). Clinical characteristics did not differ significantly at inclusion between septic and non- septic patients (Table 6). Infections were microbiologically proven in 40 of 49 infected patients (82 %) with 55 % Gram-negative, 42 % Gram-positive bacteria, and 3 % fungal infections. The major sources of infection were the respiratory tract (55 %) and abdomen (22 %). Twenty-four percent of infected patients had a documented bloodstream infection. Neither site of infection nor microbial strains differed between surviving and non-surviving patients (Table 7).
Table 6. Clinical and biological data at admission and outcome of the patients.
Characteristic" Total (π=76) Septic patients Non-septic P value (n=47) patients (n=29)
Age, years 60 (15) 61 (14) 59 (15) 0.55
Sex1,
Male 54 (71) 37 (79) 17 (59) 0.06
Female 22 (29) 10 (21) 12 (41)
McCabe 1.3 (0.8) 1.3 (0.8) 1.3 (0.9) 0.57
Simplified Acute Physiology 50.5 (22.6) 52.6 (23.8) 46.5 (20.5) 0.65
Score II
SOFA score 8.3 (4.5) 9.7 (4.8) 5.8 (2.6) 0.38
Temperature, °C 37.9 (1.0) 37.9 (1.1) 37.9 (1.0) 0.38
Leukocytes, G/L 14.4 (7.6) 14.4 (8.2) 13.9 (3.8) 0.61
C-Reactive Protein, mg/L 154.1 (142.8) 203.9 (147.7) 62.7 (65.3) 0.002
Procalcitonin, ng/mL 20.9 (44.3) 31.4 (52.4) 1.1 (2.2) <0.001 sTREM-1 , ng/mL 1121 (953) 1611 (826) 229 (341) <0.001
Length of ICU stay, days 6.4 (7.9) 6.4 (5.3) 6.3 (11.5) 0.37
Mortality rate 20 (26.3) 15 (31.9) 5 (17.2) 0.16
Values are expressed as mean (so) unless otherwise indicated. P values are for the comparison of Septic vs Non-septic patients.
Values are expressed as number (percentage)
Table 7. Septic patients: Sites of infection and strains diagnosed at the onset of sepsis according to outcome.
Non-Survivors Total (n=49) Survivors (n=34) P value* (n=15)
Patients who had positive microbial documentation of 40(82) 28 (82) 12 (80) 0.96 infection
Patients who had positive blood culture result 12 (24) 7 (21) 5 (33) 0.54
Site of infection
Lung 27 (55) 18 (53) 9 (60) 0.67
Abdominal 11 (22) 6 (18) 5 (33) 0.53
Genito-urinary 5 (11) 5 (15) 0 (0) 0.26
Cellulitis 3 (6) 2 (6) 1 (7) 0.97
Others 3 (6) 3 (8) 0 (0) 0.22
Micro-organisms N=40 N=28 N=12
Gram-positive 17 (42) 12 (43) 5 (42) 0.61
Gram-negative 22 (55) 16 (57) 6 (50) 0.64
Fungi 1 (3) 0 (0) 1 (8) 0.21
*P values are for comparison between Survivors vs Non-Survivors
Baseline Plasmatic Levels of CRP, PCT and sTREM-1 Baseline plasmatic levels of CRP, PCT and sTREM-1 were higher among septic patients than among subjects with SIRS only (Table 6, Figure 7). Plasmatic sTREM-1 levels appeared to be most helpful in differentiating patients with sepsis from those with SIRS. Mean plasmatic sTREM-1 levels on admission were 229 ng/mL for SIRS; 1836 ng/mL for sepsis and 1413 ng/mL for septic shock (PO.001). The accuracy of the candidate parameters to distinguish patients with SIRS from those with septic conditions was highly variable (Table 8). As shown in Figure 8, plasmatic sTREM-1 levels yielded the highest discriminative value with an area under the ROC curve (AUC) of 0.97 (95 % confidence interval [Cl], 0.94 to 1.0) followed by PCT (AUC, 0.85; Cl, 0.81 to 0.89) and CRP (AUC, 0.77; Cl, 0.69 to 0.85; p<0.001). At a cut-off of 600 ng/mL, sTREM-1 yielded a sensitivity of 96 % (95 % Cl, 0.92 to 100 %) and a specificity of 89 % (Cl, 82 to 95 %) to differentiate patients with SIRS from those with sepsis or septic shock. There was no correlation between sTREM-1 levels and CRP or PCT levels, microbial species or any other clinical and biological features.
Table 8. Diagnostic performance of different sepsis predictors. sTREM-1 Procalcitonin C-Reactive Pr
Cut-off value* 600 ng/mL 0.6 ng/mL 70 mg/L
Sensitivity, % 96 84 76
Specificity, % 89 70 67
Positive predictive value, % 94 84 80
Negative predictive value, % 92 70 60
Likelihood ratio 8.6 2.8 2.2
Area under the receiver operating curve 0.97 0.85 0.77
(95% confidence interval) (0.94-1.00) (0.81-0.89) (0.69-0.85)
*Sensitivity, Specificity and Predictive values were calculated for the cut-off, which represented the best discrimination as derived from the receiver operating characteristic curves.
Clinical Significance of plasmatic sTREM-1 level In order to investigate the diagnostic performance of plasmatic sTREM- 1 levels from a clinical perspective, the Inventors conducted a multiple stepwise analysis including CRP, PCT and sTREM-1 levels. Plasmatic sTREM-1 level was found to be the strongest independent predictor of infection with an adjusted odds ratio (AOR) of 9.58 (95 % Cl, 2.31 to 38.90, P=0.002) (Table 9).
Table 9. Multivariate logistic regression analyses*
Regression Odds Ratio Variable SE P Value Coefficient (95% Confidence Interval)
Intercept -6.25 2.13 NA 0.003
C-Reactive Protein, mg/L 0.17 0.09 1.46 (0.79-2.69) 0.23
Procalcitonin, ng/mL 0.24 0.19 3.83 (1.00-14.66) 0.05 sTREM-1, ng/mL 0.52 0.16 9.58 (2.31-38.90) 0.002
*Results of stepwise selection procedures. Other variables entered in the model were Simplified Acute Physiology Score ll, Sepsis-related Organ Failure Assessment score, White Blood Cells count and Body temperature. NA indicates not applicable.
Severity of Sepsis and Outcome The Inventors further evaluated plasmatic sTREM-1 levels in relation to the patient's prognosis. Values of plasmatic CRP, PCT and sTREM-1 levels in infected patients at the time of admission, in relation to outcome, are shown in Figure 9. The most discriminative parameter to predict death among infected patients at the time of admission was a plasmatic sTREM-1 level below 1500 ng/mL (odds ratio, 6.6; 95 percent Cl 4.5 to 20.0, P=0.03). The Inventors' study has several strengths. The study population was large and comprised a diverse group of critically ill adult patients admitted to a medical ICU in various phases of infectious and ήon-infectious conditions, which allowed a generalization of the study findings. The diagnosis was determined by blinded investigators without knowledge of the plasmatic sTREM-1 levels and the patients were classified as having SIRS of non-infectious origin after incorporation of all other available clinical and laboratory data (Bone RC, et al. Chest. 1992;101:1644-55.). Finally, the inventors' study was designed as a real-life study, not including control patients without suspected infection but only patients with a high pre-test probability of sepsis, covering the spectrum of patients that is likely to be encountered in the future use of this test.
Example 4: Use of an immuno-enzymatic assay to detect soluble TREM-1 in the sera of patients with suspected sepsis
An ELISA based method for the detection of soluble human TREM-1 with applications in the diagnosis of bacterial or fungal infection, in particular sepsis, has been developed by the Inventors.
In one example, the method is as follows:
Materials:
Plate: Nunc Maxisorp 96 well
Coating buffer: carbonate pH 9,6: 0.015 M Na2C03 (0.794g in 500 ml H20), 0.035 M NaHC03 (1 ,47g in 500 ml H20)
Wash buffer: 0.1% Tween 20 in PBS, pH 7,4
Assay buffer: PBS + 0.2% BSA
Blocking solution: PBS + 3% BSA
Substrate solution: 30 mM potassium citrate ,pH 4.1, immediately before use, add 1 tablet of 3,3',5,5'-Tetramethylbenzidine (Sigma # T-3405) for 10 ml of buffer and add 2,5 μl of H20230%. Method:
a) Coating: 100μl/well of anti-human Trem-1 Antibody (Polyclonal R&D Systems Inc., Minneapolis, MN, USA#AF1278) (1:1000), [Cmother]:100 μg/ml- [Cfjnai]: 100ng/ml) diluted in coating buffer pH 9,6.
b)Seal plate and incubate overnight at +4°C
c) Wash 3 times with wash buffer
d) Block plates by adding 200μl of blocking solution
e) Incubate at 37°C for 1 hour or 2 hours at RT
f) Discard supernatant and plate 100μl standards and samples dilute in assay buffer
g) Seal plate and incubate overnight at +4°C or 2 hours at 37°C
h) Wash 6 times with wash buffer
i) Add 100μl of anti-human Trem-1 antibody (clone 21 C7) diluted in assay buffer, 1:1000 [Cm0fher]:1mg/ml - [C ^] 1 μg/ml
j) incubate 2 hours at RT
k) Wash 6 times with wash buffer I) Add 100μl of goat anti-mouse lgG HRP (Pierce #31430) diluted 1:5000 in assay buffer,
m) Seal plate and incubate 2 hours at RT
n) Wash 6 times with wash buffer
o) Add 10Qμl of substrate solution to each well, p) Incubate at room temperature
q) Stop the reaction with 1 M H2S0 50μl/well
r) Determine the optical density of each well using a microtiter reader at 450 nm
Results
The results for samples from two patients within an on-going study assayed using the method described above are shown in Table 10:
SERUM HSR34 (ng/ml)
Figure imgf000049_0001
SERUM HSR37 (ng/ml)
Figure imgf000049_0002
Table 10:
Patients with suspected sepsis were analyzed at different time points (/0, 11). Time 0 represents the day of admission into the Intensive Care Unit. Samples were obtained every 48 hours until day 15.
This example demonstrates that in the patients with sepsis (HSR37) soluble TREM-1 was detected at low levels (3.96 ng/ml) at the time of admission into the ICU and reached its maximal level between T2 (day 4) and T3 (day 6) (26 ng/ml). Soluble TREM-1 was not detected in the patients with SIRS with no associated sepsis. Levels of membrane associated TREM-1 Ligand (the detection of which is described in WO2004081233) were not detectable in the patient with sepsis (HSR37) at the time of admission into the ICU and reached maximal expression at T4 (day 8). These results indicate that both soluble TREM-1 and membrane associated TREM-1 Ligand are associated with the sepsis status and their expression correlates with the clinical course of the disease. Measurement of both soluble TREM-1 and membrane associated TREM-1 Ligand in newly admitted critically ill patients could help to rapidly identify those with infection.
Example 5: sTREM-1 Assay in Plasma from Septic Patients
In another series of 63 patients, some with sepsis (n=30) others with septic shock (n=33), plasma levels of sTREM-1 were assayed. Figure 12 shows the time course of median (with interquartile range) plasma levels of sTREM-1 in surviving (squares) and non-surviving (triangles) patients. Figure 13 shows Kaplan-Meier analysis of patients with sTREM-1>180 pg/mL (n=32) and <180 pg/mL (n=31) at the time of admission into the ICU. There was a significant difference between the two curves (Log-Rank test, p<0.01), thus underscoring the value of assaying the soluble form of TREM-1 in plasma samples of critically ill septic patients as a useful method for assessing the evolution of the disease.
Example 6: TREM-1 expression on PMNs and monocvtes
In further series of patients, monocyte (see Figure 14) and polymorphonuclear (see Figure 15) cell surface expression of TREM-1 was analysed in flow cytometry after labeling with a mouse monoclonal antibody anti-human TREM- 1, PE-labelled (clone 193015, R&D, Abingdon, UK). Results were expressed as Mean Fluorescence Intensity (MFI). Three groups of patients were studied, septic patients (n=25) and non-septic patients (n=15) or healthy controls (n=7). Respective p values (Student's t test) are depicted above each scatter plot. No significant difference for the expression of membrane TREM-1 expression on neutrophils from patients of the three groups was observed. MFI on monocytes from patients with septic shock are significantly higher than MFI from non-septic patients or healthy controls.
Figure 16 shows TREM-1 expression pattern on monocytes during septic shock according to outcome. Results are expressed as Mean Fluorescence Intensity. Respective p values are depicted above time points. 'Baseline' corresponds to the first determination and 'Last value' to the last determination of TREM-1 before intensive care unit discharge or death. These results demonstate that among patients with sepsis, those with lower levels of TREM- 1 expression on monocytes, but not on neutrophils, can be predicted to have a positive outcome.

Claims

1. A method of diagnosing disease of bacterial or fungal origin in a subject, which method comprises the step of measuring the level of sTREM-1
5 in a biological sample obtained from said subject.
2. The method of claim 1 wherein said step of measuring the level of sTREM-1 comprises the steps of: (a) contacting said biological sample with a compound capable of binding0 sTREM-1; (b) detecting the level of sTREM-1 present in the sample by observing the level of binding between said compound and sTREM-1.
3. The method of claim 1 or claim 2, comprising the further step of:5 c) correlating the detected level of sTREM-1 with the presence or absence of disease of bacterial or fungal origin.
4. The method of claim 3 where said correlation is made by comparing the measured level of sTREM-1 in the sample with a mean level in a control0 population of individuals not having disease of bacterial or fungal origin, to indicate the presence or extent of disease of bacterial or fungal origin in the patient.
5. The method of any one of claims 1 to 4, further comprising the steps of5 measuring the level of sTREM-1 in a second or further sample from the patient, the first and second or further samples being obtained at different times; and comparing the levels in the samples to indicate the progression or remission of the disease of bacterial or fungal origin. Θ
6. The method of- any one of claims 1 to- 5 wherein said disease of bacterial or fungal origin is pneumonia.
7. The method of any one of claims 1 to 5 wherein said disease of bacterial or fungal origin is sepsis.
8. The method of any one of claims 1 to 7, wherein the sample is selected from the group consisting of whole blood, blood serum, blood plasma, urine and bronchoalveolar lavage fluid.
9. The method of claim 6 wherein the sample is from bronchoalveolar lavage fluid.
10. The method of claim 7 wherein the sample is from blood serum or blood plasma.
11. The method of any one of claims 1 to 10 wherein the sample is a human sample.
12. A compound capable of binding sTREM-1 for use in the diagnosis, prognosis, monitoring of the treatment of disease of bacterial or fungal origin.
13. Use of a compound capable of binding sTREM-1 in a method of diagnosis of disease of bacterial or fungal origin.
14. A method of identifying agonists or antagonists of sTREM-1 said method comprising comparing the level of binding in a sample containing said sTREM-1 and a compound capable of binding sTREM-1, in the presence and absence of a compound to be tested.
15. An agonist or antagonist of sTREM-1 identified according to the method of claim 14.
16. A kit comprising at least one compound capable of binding sTREM-1 and reagents for detecting binding of said compound to sTREM-1 for use in the diagnosis of disease of bacterial or fungal origin
17. A kit comprising at least one compound capable of binding sTREM-1 and means for contacting said compound with a sample containing sTREM-1 for use in the diagnosis of disease of bacterial or fungal origin.
18. The method, compound, use or kit of any of the preceding claims wherein said compound specifically binds sTREM-1.
19. The method, compound, use or kit of any of the preceding claims wherein said compound capable of binding sTREM-1 is an antibody raised against all or part of the TREM-1 receptor.
20. In a method of screening a patient for presence or susceptibility to disease, comprising performing a plurality of diagnostic tests on a tissue sample from the patient for a plurality of diseases, the improvement wherein one of the diagnostic tests comprises measuring the level of sTREM-1.
21. A method, compound or kit for diagnosis, prognosis or monitoring the treatment of disease of bacterial or fungal origin substantially as herein described with reference to the accompanying figures.
22. The method of any one of claims 1 to 11 wherein the level of sTREM-1 is measured by an immunochemical technique.
23. The method of any one of claims 1 to 11 comprising the additional step of measuring the level of TREM-1 -Ligand is one or more biological samples obtained from said subject.
PCT/GB2005/000273 2004-01-27 2005-01-27 Method of diagnosing infectious disease by measuring the level of soluble trem-1 in a sample WO2005071408A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2006550296A JP4911600B2 (en) 2004-01-27 2005-01-27 Method for diagnosing infectious diseases by measuring soluble TREM-1 levels in a sample
EP05702028A EP1709444B1 (en) 2004-01-27 2005-01-27 Method of diagnosing infectious disease by measuring the level of soluble trem-1 in a sample
US10/587,356 US8021836B2 (en) 2004-01-27 2005-01-27 Method of diagnosing infectious disease by measuring the level of soluble TREM-1 in a sample
AU2005207111A AU2005207111A1 (en) 2004-01-27 2005-01-27 Method of diagnosing infectious disease by measuring the level of soluble TREM-1 in a sample
ES05702028T ES2386966T3 (en) 2004-01-27 2005-01-27 Method to diagnose infectious diseases by measuring the level of soluble TREM-1 in a sample
CA002554769A CA2554769A1 (en) 2004-01-27 2005-01-27 Method of diagnosing infectious disease by measuring the level of soluble trem-1 in a sample

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0401730.7A GB0401730D0 (en) 2004-01-27 2004-01-27 Diagnosis method
GB0401730.7 2004-01-27

Publications (1)

Publication Number Publication Date
WO2005071408A1 true WO2005071408A1 (en) 2005-08-04

Family

ID=31971514

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2005/000273 WO2005071408A1 (en) 2004-01-27 2005-01-27 Method of diagnosing infectious disease by measuring the level of soluble trem-1 in a sample

Country Status (8)

Country Link
US (1) US8021836B2 (en)
EP (1) EP1709444B1 (en)
JP (1) JP4911600B2 (en)
AU (1) AU2005207111A1 (en)
CA (1) CA2554769A1 (en)
ES (1) ES2386966T3 (en)
GB (1) GB0401730D0 (en)
WO (1) WO2005071408A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009141359A1 (en) 2008-05-23 2009-11-26 Pronota N.V. New biomarker for diagnosis, prediction and/or prognosis of sepsis and uses thereof
GR1007327B (en) * 2007-08-08 2011-06-27 Ευαγγελος Γιαμαρελλος-Μπουρμπουλης Soluble triggering receptor s-trem-1 used for activating the myeloid cells and controlling the efficiency of the peptic ulcer treatment
US8106165B2 (en) 1997-03-07 2012-01-31 Human Genome Sciences, Inc. Antibodies to HNFIP24 polypeptides
WO2014006022A1 (en) 2012-07-02 2014-01-09 Université De Lorraine Glycoside derivatives, preparation thereof and use thereof as prosthetic groups
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US10179814B2 (en) 2014-07-17 2019-01-15 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
WO2020065044A1 (en) * 2018-09-28 2020-04-02 Inotrem Use of soluble trem-1 levels for identifying subjects susceptible to respond to an anti-inflammatory therapy
US11155618B2 (en) 2018-04-02 2021-10-26 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
WO2021245293A1 (en) * 2020-06-05 2021-12-09 Inotrem Soluble trem-1 as a marker of severity or complications for a subject suffering from a coronavirus infection

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006053442A1 (en) * 2006-11-12 2008-05-15 Brahms Aktiengesellschaft Diagnosis and risk stratification of infections and chronic diseases of the respiratory and lungs by means of proVasopressin, in particular copeptin or neurophysin II
DE102006060112A1 (en) 2006-12-20 2008-06-26 Brahms Aktiengesellschaft Diagnosis and risk stratification using the new marker CT-proADM
WO2008145701A1 (en) * 2007-05-29 2008-12-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for predicting the outcome of a critically ill patient
US20090214469A1 (en) * 2008-02-25 2009-08-27 Jeffrey Gold Methods and Compositions for the Treatment of Sepsis
US9709565B2 (en) 2010-04-21 2017-07-18 Memed Diagnostics Ltd. Signatures and determinants for distinguishing between a bacterial and viral infection and methods of use thereof
ES2679107T3 (en) 2012-02-09 2018-08-22 Memed Diagnostics Ltd. Badges and determinants to diagnose infections and methods to use them
WO2015031996A1 (en) * 2013-09-05 2015-03-12 University Health Network Biomarkers for early determination of a critical or life threatening response to illness and/or treatment response
CN102636645A (en) * 2012-04-24 2012-08-15 广州吉赛生物科技有限公司 sTERM-1 enzyme-linked immunodetection kit and method thereof
MX362388B (en) * 2013-02-06 2019-01-15 Geissler Companies Llc System and method for determining antibiotic effectiveness in respiratory diseased using auscultation analysis.
EP2835641A1 (en) * 2013-08-09 2015-02-11 Inotrem Methods and kits for predicting the risk of having a cardiovascular disease or event
EP3180621B1 (en) 2014-08-14 2020-04-01 Memed Diagnostics Ltd. Computational analysis of biological data using manifold and a hyperplane
US20170234873A1 (en) 2014-10-14 2017-08-17 Memed Diagnostics Ltd. Signatures and determinants for diagnosing infections in non-human subjects and methods of use thereof
WO2017149548A1 (en) 2016-03-03 2017-09-08 Memed Diagnostics Ltd. Rna determinants for distinguishing between bacterial and viral infections
CN109804245B (en) 2016-07-10 2022-10-25 米密德诊断学有限公司 Early diagnosis of infection
EP4141448A1 (en) 2016-07-10 2023-03-01 MeMed Diagnostics Ltd. Protein signatures for distinguishing between bacterial and viral infections
WO2018060999A1 (en) 2016-09-29 2018-04-05 Memed Diagnostics Ltd. Methods of risk assessment and disease classification
US11385241B2 (en) 2016-09-29 2022-07-12 Memed Diagnostics Ltd. Methods of prognosis and treatment
JOP20190248A1 (en) 2017-04-21 2019-10-20 Amgen Inc Trem2 antigen binding proteins and uses thereof
US10209260B2 (en) 2017-07-05 2019-02-19 Memed Diagnostics Ltd. Signatures and determinants for diagnosing infections and methods of use thereof
US10835185B2 (en) * 2018-03-08 2020-11-17 General Electric Company System and method for detecting ventilator-associated pneumonia (VAP)
WO2020036987A1 (en) 2018-08-13 2020-02-20 Signablok, Inc. Peptides and compositions for targeted treatment and imaging
US10836828B2 (en) 2019-02-06 2020-11-17 Pionyr Immunotherapeutics, Inc. Anti-TREM1 antibodies and related methods

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6420526B1 (en) * 1997-03-07 2002-07-16 Human Genome Sciences, Inc. 186 human secreted proteins
US6420326B1 (en) 1997-08-13 2002-07-16 The Procter & Gamble Company Glass cleaner compositions having good surface lubricity and alkaline buffer
WO2002058721A1 (en) 2000-12-08 2002-08-01 Baylor College Of Medicine Trem-1 splice variant for use in modifying immune responses
US20030165875A1 (en) 2001-03-20 2003-09-04 Marco Colonna Novel receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
WO2004035732A2 (en) * 2002-08-29 2004-04-29 Five Prime Therapeutics, Inc. Human polypeptides encoded by polynucleotides and methods of their use
WO2004081233A1 (en) * 2003-03-10 2004-09-23 Bioxell S.P.A. Diagnostic and prognostic compounds and method

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6420526B1 (en) * 1997-03-07 2002-07-16 Human Genome Sciences, Inc. 186 human secreted proteins
US6420326B1 (en) 1997-08-13 2002-07-16 The Procter & Gamble Company Glass cleaner compositions having good surface lubricity and alkaline buffer
WO2002058721A1 (en) 2000-12-08 2002-08-01 Baylor College Of Medicine Trem-1 splice variant for use in modifying immune responses
US20030165875A1 (en) 2001-03-20 2003-09-04 Marco Colonna Novel receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
WO2004035732A2 (en) * 2002-08-29 2004-04-29 Five Prime Therapeutics, Inc. Human polypeptides encoded by polynucleotides and methods of their use
WO2004081233A1 (en) * 2003-03-10 2004-09-23 Bioxell S.P.A. Diagnostic and prognostic compounds and method

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BOUCHON A ET AL., NATURE, vol. 230, 2001, pages 1103 - 7
BOUCHON A ET AL: "CUTTING EDGE: INFLAMMATORY RESPONSES CAN BE TRIGGERED BY TREM-1, A NOVEL RECEPTOR EXPRESSED ON NEUTROPHILS AND MONOCYTES", JOURNAL OF IMMUNOLOGY, THE WILLIAMS AND WILKINS CO. BALTIMORE, US, vol. 164, 2000, pages 4991 - 4995, XP002951620, ISSN: 0022-1767 *
BOUCHON A ET AL: "TREM-1 amplifies inflammation and is a crucial mediator of septic shock", NATURE, MACMILLAN JOURNALS LTD. LONDON, GB, vol. 410, no. 6832, 26 April 2001 (2001-04-26), pages 1103 - 1107, XP002285055, ISSN: 0028-0836 *
COLONNA M ET AL: "TREM-1 (TRIGGERING RECEPTOR EXPRESSED ON MYELOID CELLS): A NEW PLAYER IN ACUTE INFLAMMATORY RESPONSES", JOURNAL OF INFECTIOUS DISEASES, CHICAGO, IL, US, vol. 187, no. SUPPL 2, 15 June 2003 (2003-06-15), pages S397 - S401, XP008033778, ISSN: 0022-1899 *
GINGRAS MARIE-CLAUDE ET AL: "TREM-1, MDL-1, and DAP12 expression is associated with a mature stage of myeloid development", MOLECULAR IMMUNOLOGY, vol. 38, no. 11, March 2002 (2002-03-01), pages 817 - 824, XP002332587, ISSN: 0161-5890 *
NATHAN C ET AL: "TREM-1: A new regulator of innate immunity in sepsis syndrome", NATURE MEDICINE, NATURE PUBLISHING, CO, US, vol. 7, no. 5, 2001, pages 530 - 532, XP002291851, ISSN: 1078-8956 *
NOCHI HITOSHI ET AL: "Modulation of hepatic granulomatous responses by transgene expression of DAP12 or TREM-1-Ig molecules", AMERICAN JOURNAL OF PATHOLOGY, PHILADELPHIA, PA, US, vol. 162, no. 4, April 2003 (2003-04-01), pages 1191 - 1201, XP002285056, ISSN: 0002-9440 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8106165B2 (en) 1997-03-07 2012-01-31 Human Genome Sciences, Inc. Antibodies to HNFIP24 polypeptides
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US10603357B2 (en) 2004-11-29 2020-03-31 Bristol-Myers Squibb Company Therapeutic TREM-1 peptides
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
GR1007327B (en) * 2007-08-08 2011-06-27 Ευαγγελος Γιαμαρελλος-Μπουρμπουλης Soluble triggering receptor s-trem-1 used for activating the myeloid cells and controlling the efficiency of the peptic ulcer treatment
US8697370B2 (en) 2008-05-23 2014-04-15 Pronota N.V. Biomarker for diagnosis, prediction and/or prognosis of sepsis and uses thereof
EP2796878A1 (en) * 2008-05-23 2014-10-29 Biocartis NV New biomarkers for diagnosis, prediction and/or prognosis of sepsis and uses thereof
WO2009141359A1 (en) 2008-05-23 2009-11-26 Pronota N.V. New biomarker for diagnosis, prediction and/or prognosis of sepsis and uses thereof
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10906965B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation wtih antibodies that bind peptidoglycan recognition protein 1
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US10150809B2 (en) 2012-02-15 2018-12-11 Bristol-Myers Squibb Company Antibodies that bind peptidoglycan recognition protein 1
US10189904B2 (en) 2012-02-15 2019-01-29 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10906975B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation with antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
WO2014006022A1 (en) 2012-07-02 2014-01-09 Université De Lorraine Glycoside derivatives, preparation thereof and use thereof as prosthetic groups
US10179814B2 (en) 2014-07-17 2019-01-15 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
US11072654B2 (en) 2014-07-17 2021-07-27 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
US11155618B2 (en) 2018-04-02 2021-10-26 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
US11919954B2 (en) 2018-04-02 2024-03-05 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
US11952420B2 (en) 2018-04-02 2024-04-09 Bristol-Myers Squibb Company Nucleic acids encoding anti-TREM-1 antibodies
WO2020065044A1 (en) * 2018-09-28 2020-04-02 Inotrem Use of soluble trem-1 levels for identifying subjects susceptible to respond to an anti-inflammatory therapy
US20210364532A1 (en) * 2018-09-28 2021-11-25 Inotrem Use of soluble trem-1 levels for identifying subjects susceptible to respond to an anti-inflammatory therapy
WO2021245293A1 (en) * 2020-06-05 2021-12-09 Inotrem Soluble trem-1 as a marker of severity or complications for a subject suffering from a coronavirus infection

Also Published As

Publication number Publication date
ES2386966T3 (en) 2012-09-07
US20070281319A1 (en) 2007-12-06
EP1709444A1 (en) 2006-10-11
AU2005207111A1 (en) 2005-08-04
CA2554769A1 (en) 2005-08-04
US8021836B2 (en) 2011-09-20
GB0401730D0 (en) 2004-03-03
EP1709444B1 (en) 2012-06-06
JP4911600B2 (en) 2012-04-04
JP2007519916A (en) 2007-07-19

Similar Documents

Publication Publication Date Title
US8021836B2 (en) Method of diagnosing infectious disease by measuring the level of soluble TREM-1 in a sample
US9945873B2 (en) Methods and kits for predicting the risk of respiratory failure, renal failure or thrombopenia in a septic patient by measuring endocan levels in blood
EP1587409A2 (en) Diagnosis and monitoring of inflammation, ischemia and appendicitis
KR20180118822A (en) Method for prediction of prognosis of sepsis
EP1295127B1 (en) Inter-alpha-trypsin as a marker for sepsis
EP2674755B1 (en) Method for diagnosing acute lung injury
US8097424B2 (en) Method for predicting the outcome of a critically ill patient
CN110678757B (en) Method for diagnosing or monitoring renal function or diagnosing renal dysfunction
US8697368B2 (en) Diagnostic marker for lung cancer comprising HPαR as active ingredient
JP2007502427A (en) Cirrhosis diagnostic kit containing an antibody specific for human proto-oncoprotein
US20210285949A1 (en) Pro-adrenomedullin or fragment thereof in patients infected with corona virus and treatments with binder against adrenomedullin
AU2012208532B2 (en) Methods and kits for predicting the risk of respiratory failure, renal failure or thrombopenia in a septic patient by measuring endocan levels in blood
SİPAHİ et al. Relationship between inflammatory markers and infection in chronic kidney disease
Grover et al. Biomarkers in the Diagnosis of Ventilator-Associated Pneumonia

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005702028

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006550296

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2005207111

Country of ref document: AU

Ref document number: 2554769

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2005207111

Country of ref document: AU

Date of ref document: 20050127

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005207111

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2005702028

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10587356

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10587356

Country of ref document: US