US20140314775A1 - Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment of anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease - Google Patents

Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment of anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease Download PDF

Info

Publication number
US20140314775A1
US20140314775A1 US14/358,400 US201214358400A US2014314775A1 US 20140314775 A1 US20140314775 A1 US 20140314775A1 US 201214358400 A US201214358400 A US 201214358400A US 2014314775 A1 US2014314775 A1 US 2014314775A1
Authority
US
United States
Prior art keywords
adm
antibody
adrenomedullin
scaffold
fragment
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/358,400
Other languages
English (en)
Inventor
Andreas Bergmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Adrenomed AG
Original Assignee
Adrenomed AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adrenomed AG filed Critical Adrenomed AG
Assigned to ADRENOMED AG reassignment ADRENOMED AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BERGMANN, ANDREAS
Publication of US20140314775A1 publication Critical patent/US20140314775A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/54F(ab')2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/70Mechanisms involved in disease identification
    • G01N2800/7095Inflammation

Definitions

  • Subject matter of the present invention is an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease or acute condition.
  • ADM anti-Adrenomedullin
  • Subject matter of the present invention is a method for regulating the fluid balance in a patient having a chronic or acute disease or acute condition.
  • ADM peptide adrenomedullin
  • the precursor peptide which comprises, inter alia, a signal sequence of 21 amino acids at the N-terminus, is referred to as “preproadrenomedullin” (pre-proADM).
  • pre-proADM preproadrenomedullin
  • all amino acid positions specified usually relate to the pre-proADM which comprises the 185 amino acids.
  • the peptide adrenomedullin (ADM) is a peptide which comprises 52 amino acids (SEQ ID NO: 21) and which comprises the amino acids 95 to 146 of pre-proADM, from which it is formed by proteolytic cleavage.
  • ADM may be regarded as a polyfunctional regulatory peptide. It is released into the circulation in an inactive form extended by glycine (Kitamura, K., et al., “The intermediate form of glycine-extended adrenomedullin is the major circulating molecular form in human plasma”, Biochem. Biophys. Res. Commun., Vol. 244(2), pp. 551-555 (1998).
  • ADM is an effective vasodilator, and thus it is possible to associate the hypotensive effect with the particular peptide segments in the C-terminal part of ADM.
  • PAMP further physiologically active peptide formed from pre-proADM likewise exhibits a hypotensive effect, even if it appears to have an action mechanism differing from that of ADM (cf. in addition to the abovementioned review articles (Eto, T., “A review of the biological properties and clinical implications of adrenomedullin and proadrenomedullin N-terminal 20 peptide (PAMP), hypotensive and vasodilating peptides”, Peptides, Vol.
  • the ADM level in patients with congestive heart failure, myocardial infarction, kidney diseases, hypertensive disorders, Diabetes mellitus, in the acute phase of shock and in sepsis and septic shock are significantly increased, although to different extents.
  • the PAMP concentrations are also increased in some of said pathological states, but the plasma levels are reduced relative to ADM ((Eto, T., “A review of the biological properties and clinical implications of adrenomedullin and proadrenomedullin N-terminal 20 peptide (PAMP), hypotensive and vasodilating peptides”, Peptides, Vol. 22, pp. 1693-1711 (2001)); page 1702).
  • the midregional partial peptide of the proadrenomedullin which contains amino acids (45-92) of the entire preproadrenomedullin, is measured, in particular, with an immunoassay which works with at least one labeled antibody that specifically recognizes a sequence of the mid-proADM (WO2004/090546).
  • WO-A1 2004/097423 describes the use of an antibody against adrenomedullin for diagnosis, prognosis, and treatment of cardiovascular disorders.
  • Treatment of diseases by blocking the ADM receptor are also described in the art, (e.g. WO-A1 2006/027147, PCT/EP2005/012844) said diseases may be sepsis, septic shock, cardiovascular diseases, infections, dermatological diseases, endocrinological diseases, metabolic diseases, gastroenterological diseases, cancer, inflammation, hematological diseases, respiratory diseases, muscle skeleton diseases, neurological diseases, urological diseases.
  • ADM-neutralizing antibodies neutralize the before mentioned effects during the early phase of sepsis (Wang, P., “Adrenomedullin and cardiovascular responses in sepsis”, Peptides, Vol. 22, pp. 1835-1840 (2001).
  • ADM constitutes a risk factor that is strongly associated with the mortality of patients in septic shock.
  • Schütz et al. “Circulating Precursor levels of endothelin-1 and adrenomedullin, two endothelium-derived, counteracting substances, in sepsis”, Endothelium, 14:345-351, (2007)).
  • adrenomedullin antagonists i.e. molecules which prevent or attenuate the vasodilating action of adrenomedulin, e.g. by blocking its relevant receptors, or substances preventing the binding of adrenomedullin to its receptor (e.g.
  • binders as e.g. antibodies binding to adrenomedullin and blocking its receptor bindings sites; “immunological neutralization”).
  • immunological neutralization Such use, or combined use, including a subsequent or preceding separate use, has been described in certain cases to be desirable for example to improve the therapeutic success, or to avoid undesirable physiological stress or side effects.
  • neutralizing ADM antibodies may be used for the treatment of sepsis in the late stage of sepsis.
  • ADM binding protein complement factor H
  • ADM binding protein is present in the circulation of said organism in high concentrations (Pio et al.: Identification, characterization, and physiological actions of factor H as an Adrenomedullin binding Protein present in Human Plasma; Microscopy Res. and Technique, 55:23-27 (2002) and Martinez et al.; Mapping of the Adrenomedullin-Binding domains in Human Complement factor H; Hypertens Res Vol. 26, Suppl (2003), S56-59).
  • the ADM-binding-Protein-1 may be also referred to as ADM-binding-Protein-1 (complement factor H).
  • Patients having a chronic or acute disease or acute condition may suffer from fluid imbalance. This may cause severe adverse events such as kidney failure and mortality.
  • the expression “regulating fluid balance” with the context of the instant invention is directed to any correction of a manifested—imbalance—of a patient's fluid balance due to an underlying chronic or acute disease or acute condition. Said correction is in favour of re-establishing normotension in said patients.
  • the person skilled in the art is fully aware that blood pressure in general, as well as hyper- and hypotension is closely related to the fluid balance of a patient.
  • Fluid balance is the balance of the input and the output of fluids in the body to allow metabolic processes to function. Dehydration is defined as a 1% or greater loss of body mass as a result of fluid loss.
  • the three elements for assessing fluid balance and hydration status are: clinical assessment, body weight and urine output; review fluid balance charts and review of blood chemistry. All this is very well known to a man skilled in the art (Alison Shepherd, Nursing Tomes 19.07.11/Vol 107 No 28, pages 12 to 16).
  • a person in need of regulating the fluid balance and/or improving the fluid balance of such patients is a person that has a 1% or greater loss of body mass as a result of fluid loss.
  • the fluid balance may be assessed according to Scales and Pilsworth (2008) Nursing Standard 22:47, 50-57.
  • normal urine output is in the range of 0.5 to 2 ml/kg of body weight per hour.
  • the minimum acceptable urine output for a patient with normal renal function is 0.5 ml/kg per hour. All these standards may be used to assess whether a patient is in need for regulating the fluid balance and/or improving the fluid balance.
  • antibodies capable to bind ADM, and thus are directed against ADM, and thus can be referred to as “anti-ADM antibodies”, “anti-ADM antibody fragments”, or “anti-ADM non-Ig scaffolds”.
  • the anti-ADM antibodies, anti-ADM antibody fragments, or anti-ADM non-Ig scaffolds in accordance with the invention are capable to bind circulating ADM, and thus are directed against circulating ADM.
  • an anti-Adrenomedullin (ADM) antibody or an anti-ADM antibody fragment thereof or and ADM non-Ig scaffold is to be used in combination with fluids administered intravenously, wherein said combination is for use in therapy of an acute disease or acute condition of a patient for the regulation of fluid balance.
  • ADM anti-Adrenomedullin
  • an anti-Adrenomedullin (ADM) antibody or an anti-ADM antibody fragment thereof or an ADM non-Ig scaffold is to be used in combination with vasopressor agents, e.g. catecholamine, wherein said combination is for use in therapy of an acute disease or acute condition of a patient for the regulation of fluid balance.
  • vasopressor agents e.g. catecholamine
  • Subject matter of the present invention is an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment or an anti-ADM non-Ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease or acute condition.
  • ADM anti-adrenomedullin
  • Subject matter of the present invention is a method for regulating the fluid balance in a patient having a chronic or acute disease or acute condition.
  • said patient is a patient in need of regulating the fluid balance.
  • Subject matter of the present invention is an anti-ADM antibody or anti-ADM antibody fragment or an anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for the regulation of fluid balance.
  • An anti-adrenomedullin (ADM) antibody is an antibody that binds specifically to ADM
  • anti-adrenomedullin antibody fragment is a fragment of an anti-ADM antibody, wherein said fragment binds specifically to ADM.
  • An anti-ADM non-Ig scaffold is a non-Ig scaffold that binds specifically to ADM.
  • binding to ADM allows binding to other antigens as well. This means, this specificity would not exclude that the antibody may cross-react with other polypeptides that against it has been raised.
  • Patient in status of fluid imbalance may get fluid administered intravenously as a standard measure of care, especially in an ICU setting. It is, however, desirable to reduce or avoid the additional fluid administration because of complications that might occur as e.g. the occurrence of edema (acroedema).
  • Edema means swelling caused by fluid in the body's tissues. It may occur in feet and legs, but can involve the entire body and can involve organs as e.g. lung, heart, eye.
  • anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold may be administered at a point of time when the patient is in need of fluid administration.
  • said patient is a patient in need of regulating the fluid balance.
  • subject matter of the present invention is also an anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for the regulation of fluid balance which includes but is not limited to the prevention or reduction of edema.
  • the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold may be also administered preventively before the patient exhibits any signs of fluid imbalance.
  • fluid imbalance problems may be expected, e.g. comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages by chemotherapy.
  • the antibody or fragment or scaffold according to the present invention for reducing the risk of mortality during sepsis and septic shock, i.e. late phases of sepsis.
  • At least one sign of end-organ dysfunction as mentioned under 3) is manifested.
  • Septic shock is indicated, if there is refractory hypotension that does not respond to treatment and intravenous fluid administration alone is insufficient to maintain a patient's blood pressure from becoming hypotensive also provides for an administration of an anti-ADM antibody or an anti-ADM antibody fragment or an anti-ADM non-Ig scaffold in accordance with the present invention.
  • acute disease or acute conditions may be selected from the group but are not limited to the group comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrome (SIRS), or sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy.
  • severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrome (SIRS), or sepsis
  • other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis
  • shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy.
  • the patient is not suffering from SIRS, a severe infection, sepsis, shock as e.g. septic shock.
  • Said severe infection denotes e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), sepsis, severe sepsis, and shock as e.g. septic shock.
  • SIRS Systemic inflammatory Response-Syndrom
  • sepsis severe sepsis
  • shock as e.g. septic shock.
  • a severe sepsis is characterized in that sepsis is manifested in said patient, and additionally a clinical suspicion of any organ dysfunction is present, being it:
  • said acute disease or acute condition is not sepsis, or not severe sepsis, or not SIRS, or not shock, or not septic shock.
  • said acute disease or acute condition is not sepsis.
  • said acute disease or acute condition is selected from the group comprising meningitis, diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy.
  • meningitis e.g. diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis
  • shock e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy.
  • the patient is referred to as being in negative fluid balance.
  • physiological fluid is often given intravenously by a physician to compensate for that loss.
  • a positive fluid balance where fluid gain is greater than fluid loss may provides for information to a problem with either the renal or cardiovascular system.
  • fluid therapy in general denotes the therapeutic administration of fluids (such as physiologic saline solution or water for injection (WFI)) to a patient as a treatment or preventative measure. It can be administered via intravenous, intraperitoneal, intraosseous, subcutaneous and oral routes.
  • fluids such as physiologic saline solution or water for injection (WFI)
  • WFI water for injection
  • Fluid therapy is indicated either when there is a loss of fluid or there is a risk of loss of fluid due to an underlying disease or condition.
  • the medicaments provided by the present invention being anti-ADM antibodies, anti-ADM antibody fragments, or anti-ADM non-Ig scaffolds are only intended to be used for sake of regulating the fluid balance and thus not for any methods of primary treatment to a chronic or acute disease or condition itself.
  • This means the present invention does not provide for a therapy of healing/curing e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), or sepsis, or severe sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, or damages induced by chemotherapy within the scope of the invention.
  • SIRS Systemic inflammatory Response-Syndrom
  • the fluid regulating effect of the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold is thus supporting the primary therapy of said chronic or acute disease or acute condition.
  • a chronic or acute disease or acute condition like severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), sepsis or the like
  • the primary therapy would be e.g. the administration of antibiotics.
  • the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold would regulate the fluid balance and would help to prevent worsening of the critical condition of said patient until the e.g. antibiotic administration takes effect.
  • the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-IG scaffold may be administered in a preventive way or in a therapeutic way, this means in order to prevent fluid imbalance problems or in order to reduce fluid imbalance when fluid imbalance problems are present in said patient. Edema is included in the term fluid imbalance problems.
  • the patients may have a chronic or acute disease or acute condition as primary or underlying disease such as e.g. cancer, or diabetes mellitus.
  • primary or underlying diseases are not prima facie targeted by the therapeutic treatment according to the invention.
  • the therapeutic treatment pursuant to the invention is solely directed against acute symptoms that are diagnosed or indicated for fluid therapy.
  • the invention does not provide for a primary therapy for cancer, diabetes mellitus, meningitis, Systemic inflammatory Response-Syndrom (SIRS), sepsis or the like, but for a therapy of patients that suffer from fluid imbalance that is due to an acute disease or acute condition, and thus they are in need of fluid administration.
  • SIRS Systemic inflammatory Response-Syndrom
  • an antiADM antibody or an anti-ADM antibody fragment or an anti-ADM non-Ig scaffold is to be used in combination with fluids administered intravenously, wherein said combination is for use in therapy of an acute disease or acute condition of a patient for the regulation of fluid balance of said patient.
  • said patient having a chronic or acute disease or condition being in need for regulation of fluid balance is characterized by the need of said patient to get intravenous fluids.
  • said patient having a chronic or acute disease or condition being in need for regulation of fluid balance is characterized by the risk of said patient of getting edema or by the presence of edema in said patient.
  • Subject matter of the invention in one specific embodiment is, thus, an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of a patient in need of intravenous fluids or for use in therapy of a patient having a risk of getting edema or by the presence of edema in said patient.
  • ADM anti-Adrenomedullin
  • an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is to be used in combination with vasopressor agents, e.g. catecholamine, wherein said combination is for use in therapy of an acute disease or acute condition of a patient for regulation of fluid balance.
  • vasopressor agents e.g. catecholamine
  • said patient having a chronic or acute disease or condition being in need for regulation of fluid balance is characterized by the need of said patient to get vasopressor agents, e.g. catecholamine, administration.
  • vasopressor agents e.g. catecholamine
  • Subject matter of the invention in one specific embodiment is, thus, an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold for use in therapy of a patient in need of a vasopressor agent, e.g. catecholamine treatment.
  • ADM anti-Adrenomedullin
  • an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold is monospecific.
  • Monospecific anti-Adrenomedullin (ADM) antibody or monospecific anti-adrenomedullin antibody fragment or monospecific anti-ADM non-Ig scaffold means that said antibody or antibody fragment or non-Ig scaffold binds to one specific region encompassing at least 5 amino acids within the target ADM.
  • Monospecific anti-Adrenomedullin (ADM) antibody or monospecific anti-adrenomedullin antibody fragment or monospecific anti-ADM non-Ig scaffold are anti-Adrenomedullin (ADM) antibodies or anti-adrenomedullin antibody fragments or anti-ADM non-Ig scaffolds that all have affinity for the same antigen.
  • the present invention provides for a monospecific anti-Adrenomedullin (ADM) antibody or monospecific anti-adrenomedullin antibody fragment or monospecific anti-ADM non-Ig scaffold, characterized in that said antibody or antibody fragment or non-Ig scaffold binds to one specific region encompassing at least 4 amino acids within the target ADM.
  • ADM monospecific anti-Adrenomedullin
  • the anti-ADM antibody or the antibody fragment binding to ADM is a monospecific antibody.
  • Monospecific means that said antibody or antibody fragment binds to one specific region encompassing preferably at least 4, or at least 5 amino acids within the target ADM.
  • Monospecific antibodies or fragments are antibodies or fragments that all have affinity for the same antigen.
  • Monoclonal antibodies are monospecific, but monospecific antibodies may also be produced by other means than producing them from a common germ cell.
  • An antibody according to the present invention is a protein including one or more polypeptides substantially encoded by immunoglobulin genes that specifically binds an antigen.
  • the recognized immunoglobulin genes include the kappa, lambda, alpha (IgA), gamma (IgG 1 , IgG 2 , IgG 3 , IgG 4 ), delta (IgD), epsilon (IgE) and mu (IgM) constant region genes, as well as the myriad immunoglobulin variable region genes.
  • Full-length immunoglobulin light chains are generally about 25 Kd or 214 amino acids in length.
  • Full-length immunoglobulin heavy chains are generally about 50 Kd or 446 amino acid in length.
  • Light chains are encoded by a variable region gene at the NH2-terminus (about 110 amino acids in length) and a kappa or lambda constant region gene at the COOH-terminus.
  • Heavy chains are similarly encoded by a variable region gene (about 116 amino acids in length) and one of the other constant region genes.
  • the basic structural unit of an antibody is generally a tetramer that consists of two identical pairs of immunoglobulin chains, each pair having one light and one heavy chain. In each pair, the light and heavy chain variable regions bind to an antigen, and the constant regions mediate effector functions.
  • Immunoglobulins also exist in a variety of other forms including, for example, Fv, Fab, and (Fab′) 2 , as well as bifunctional hybrid antibodies and single chains (e.g., Lanzavecchia et al., Eur. J. Immunol. 17:105, 1987; Huston et al., Proc. Natl. Acad. Sci.
  • An immunoglobulin light or heavy chain variable region includes a framework region interrupted by three hypervariable regions, also called complementarity determining regions (CDR's) (see, Sequences of Proteins of Immunological Interest , E. Kabat et al., U.S. Department of Health and Human Services, 1983). As noted above, the CDRs are primarily responsible for binding to an epitope of an antigen.
  • An immune complex is an antibody, such as a monoclonal antibody, chimeric antibody, humanized antibody or human antibody, or functional antibody fragment, specifically bound to the antigen.
  • Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin variable and constant region genes belonging to different species.
  • the variable segments of the genes from a mouse monoclonal antibody can be joined to human constant segments, such as kappa and gamma 1 or gamma 3.
  • a therapeutic chimeric antibody is thus a hybrid protein composed of the variable or antigen-binding domain from a mouse antibody and the constant or effector domain from a human antibody, although other mammalian species can be used, or the variable region can be produced by molecular techniques. Methods of making chimeric antibodies are well known in the art, e.g., see U.S. Pat. No. 5,807,715.
  • a “humanized” immunoglobulin is an immunoglobulin including a human framework region and one or more CDRs from a non-human (such as a mouse, rat, or synthetic) immunoglobulin.
  • the non-human immunoglobulin providing the CDRs is termed a “donor” and the human immunoglobulin providing the framework is termed an “acceptor.”
  • all the CDRs are from the donor immunoglobulin in a humanized immunoglobulin.
  • Constant regions need not be present, but if they are, they must be substantially identical to human immunoglobulin constant regions, i.e., at least about 85-90%, such as about 95% or more identical.
  • a “humanized antibody” is an antibody comprising a humanized light chain and a humanized heavy chain immunoglobulin.
  • a humanized antibody binds to the same antigen as the donor antibody that provides the CDRs.
  • the acceptor framework of a humanized immunoglobulin or antibody may have a limited number of substitutions by amino acids taken from the donor framework. Humanized or other monoclonal antibodies can have additional conservative amino acid substitutions which have substantially no effect on antigen binding or other immunoglobulin functions.
  • Humanized immunoglobulins can be constructed by means of genetic engineering (e.g., see U.S. Pat. No. 5,585,089).
  • a human antibody is an antibody wherein the light and heavy chain genes are of human origin. Human antibodies can be generated using methods known in the art. Human antibodies can be produced by immortalizing a human B cell secreting the antibody of interest.
  • Immortalization can be accomplished, for example, by EBV infection or by fusing a human B cell with a myeloma or hybridoma cell to produce a trioma cell.
  • Human antibodies can also be produced by phage display methods (see, e.g., Dower et al., PCT Publication No. WO91/17271; McCafferty et al., PCT Publication No. WO92/001047; and Winter, PCT Publication No. WO92/20791), or selected from a human combinatorial monoclonal antibody library (see the Morphosys website).
  • Human antibodies can also be prepared by using transgenic animals carrying a human immunoglobulin gene (for example, see Lonberg et al., PCT Publication No. WO93/12227; and Kucherlapati, PCT Publication No. WO91/10741).
  • the anti-ADM antibody may have the formats known in the art.
  • Examples are human antibodies, monoclonal antibodies, humanized antibodies, chimeric antibodies, CDR-grafted antibodies.
  • antibodies according to the present invention are recombinantly produced antibodies as e.g. IgG, a typical full-length immunoglobulin, or antibody fragments containing at least the F-variable domain of heavy and/or light chain as e.g. chemically coupled antibodies (fragment antigen binding) including but not limited to Fab-fragments including Fab minibodies, single chain Fab antibody, monovalent Fab antibody with epitope tags, e.g.
  • bivalent Fab-V5Sx2 bivalent Fab (mini-antibody) dimerized with the CH3 domain
  • bivalent Fab or multivalent Fab e.g. formed via multimerization with the aid of a heterologous domain, e.g. via dimerization of dHLX domains, e.g. Fab-dHLX-FSx2; F(ab′)2-fragments, scFv-fragments, multimerized multivalent or/and multispecific scFv-fragments, bivalent and/or bispecific diabodies, BITE® (bispecific T-cell engager), trifunctional antibodies, polyvalent antibodies, e.g. from a different class than G; single-domain antibodies, e.g. nanobodies derived from camelid or fish immunoglobulines and numerous others.
  • biopolymer scaffolds are well known in the art to complex a target molecule and have been used for the generation of highly target specific biopolymers. Examples are aptamers, spiegelmers, anticalins and conotoxins. For illustration of antibody formats please see FIGS. 1 a , 1 b and 1 c.
  • the ADM antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein.
  • the antibody format is selected from the group comprising scFab fragment, Fab fragment, scFv fragment and bioavailability optimized conjugates thereof, such as PEGylated fragments.
  • One of the most preferred formats is the scFab format.
  • Non-Ig scaffolds may be protein scaffolds and may be used as antibody mimics as they are capable to bind to ligands or antigenes.
  • Non-Ig scaffolds may be selected from the group comprising tetranectin-based non-Ig scaffolds (e.g. described in US 2010/0028995), fibronectin scaffolds (e.g. described in EP 1266 025; lipocalin-based scaffolds ((e.g. described in WO 2011/154420); ubiquitin scaffolds (e.g. described in WO 2011/073214), transferring scaffolds (e.g. described in US 2004/0023334), protein A scaffolds (e.g. described in EP 2231860), ankyrin repeat based scaffolds (e.g.
  • microproteins preferably microproteins forming a cystine knot
  • Fyn SH3 domain based scaffolds e.g. described in WO 2011/023685
  • EGFR-A-domain based scaffolds e.g. described in WO 2005/040229
  • Kunitz domain based scaffolds e.g. described in EP 1941867.
  • a Balb/c mouse was immunized with ADM-100 ⁇ g Peptide-BSA-Conjugate at day 0 and 14 (emulsified in 100 ⁇ l complete Freund's adjuvant) and 50 ⁇ g at day 21 and 28 (in 100 ⁇ l incomplete Freund's adjuvant).
  • the animal received 50 ⁇ g of the conjugate dissolved in 100 ⁇ l saline, given as one intraperitoneal and one intravenous injection.
  • Spenocytes from the immunized mouse and cells of the myeloma cell line SP2/0 were fused with lml 50% polyethylene glycol for 30 s at 37° C. After washing, the cells were seeded in 96-well cell culture plates. Hybrid clones were selected by growing in HAT medium [RPMI 1640 culture medium supplemented with 20% fetal calf serum and HAT-Supplement]. After two weeks the HAT medium is replaced with HT Medium for three passages followed by returning to the normal cell culture medium.
  • the cell culture supernatants were primary screened for antigen specific IgG antibodies three weeks after fusion.
  • the positive tested microcultures were transferred into 24-well plates for propagation. After retesting, the selected cultures were cloned and recloned using the limiting-dilution technique and the isotypes were determined (see also Lane, R. D. (1985).
  • Glutamate decarboxylase (1996) Glutamate decarboxylase (GAD) is not detectable on the surface of rat islet cells examined by cytofluorometry and complement-dependent antibody-mediated cytotoxicity of monoclonal GAD antibodies, Horm. Metab. Res. 28: 11-15).
  • the human naive antibody gene libraries HALT/8 were used for the isolation of recombinant single chain F-Variable domains (scFv) against adrenomedullin peptide.
  • the antibody gene libraries were screened with a panning strategy comprising the use of peptides containing a biotin tag linked via two different spacers to the adrenomedullin peptide sequence.
  • a mix of panning rounds using non-specifically bound antigen and streptavidin bound antigen were used to minimize background of non-specific binders.
  • the eluted phages from the third round of panning have been used for the generation of monoclonal scFv expressing E. coli strains.
  • the antibody sequence is analyzed for the structural interaction of framework regions (FR) with the complementary determining regions (CDR) and the antigen. Based on structural modeling an appropriate FR of human origin is selected and the murine CDR sequences are transplanted into the human FR. Variations in the amino acid sequence of the CDRs or FRs may be introduced to regain structural interactions, which were abolished by the species switch for the FR sequences. This recovery of structural interactions may be achieved by random approach using phage display libraries or via directed approach guided by molecular modeling (see Almagro J C, Fransson J., 2008. Humanization of antibodies. Front Biosci. 2008 Jan. 1; 13:1619-33).
  • the ADM antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, F(ab) 2 fragment and scFv-Fc Fusion protein.
  • the antibody format is selected from the group comprising scFab fragment, Fab fragment, scFv fragment and bioavailability optimized conjugates thereof, such as PEGylated fragments.
  • One of the most preferred formats is scFab format.
  • the anti-ADM antibody, anti-ADM antibody fragment, or anti-ADM non-Ig scaffold is a full length antibody, antibody fragment, or non-Ig scaffold.
  • the anti-ADM antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold is directed to and can bind to an epitope of at least 5 amino acids in length contained in ADM.
  • the anti-ADM antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold is directed to and can bind to an epitope of at least 4 amino acids in length contained in ADM.
  • the anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin is provided for use in therapy of an acute disease or acute condition of a patient wherein said antibody or fragment or scaffold is not ADM-binding-Protein-1 (complement factor H).
  • the anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin is provided for use in therapy of an acute disease or acute condition of a patient wherein said antibody or antibody fragment or non-Ig scaffold binds to a region of preferably at least 4, or at least 5 amino acids within the sequence of aa 1-42 of mature human ADM:
  • the anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin is provided for use in therapy of an acute disease or acute condition of a patient wherein said antibody or fragment or scaffold binds to a region of preferably at least 4, or at least 5 amino acids within the sequence of aa 1-21 of mature human ADM:
  • said anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold binds to a region of ADM of preferably at least 4, or at least 5 amino acids that is located in the N-terminal part (aa 1-21) of adrenomedullin, (see FIG. 2 ).
  • the anti-adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold is directed to and can bind to an epitope of at least 5 amino acids in length contained in ADM, preferably in human ADM.
  • the anti-adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold is directed to and can bind to an epitope of at least 4 amino acids in length contained in ADM, preferably in human ADM.
  • said anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold recognizes and binds to the N-terminal end (aa 1) of adrenomedullin.
  • N-terminal end means that the amino acid 1, that is “Y” of SEQ ID No. 21 or 23; is mandatory for antibody binding.
  • Said antibody or fragment or scaffold would neither bind N-terminal extended nor N-terminal modified adrenomedullin nor N-terminal degraded adrenomedullin.
  • anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold does not bind to the C-terminal portion of ADM, i.e. the aa 43-52 of ADM (SEQ ID NO: 25):
  • an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention, wherein said adrenomedullin antibody or said adrenomedullin antibody fragment or non-Ig scaffold is an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-Ig scaffold that enhances the half life (t 1/2 ; half retention time) of adrenomedullin in serum, blood, plasma at least 10%, preferably at least 50%, more preferably >50%, most preferably >100%.
  • the half life (half retention time) of ADM may be determined in human plasma in absence and presence of an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-Ig scaffold, respectively, using an immunoassay for the quantification of ADM.
  • a two-fold increase of the half life of ADM is an enhancement of half life of 100%.
  • Half Life is defined as the period over which the concentration of a specified chemical or drug takes to fall to half its baseline concentration in the specified fluid or blood.
  • ADM For some diseases blocking of ADM may be beneficial to a certain extent. However, it might also be detrimental if ADM is totally neutralized as a certain amount of ADM may be required for several physiological functions. In many reports it was emphasized that the administration of ADM may be beneficial in certain diseases. In contrast thereto in other reports ADM was reported as being life threatening when administered in certain conditions.
  • said anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold is a non-neutralizing antibody, fragment or non-Ig scaffold.
  • a neutralizing anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold would block the bioactivity of ADM to nearly 100%, to at least more than 90%, preferably to at least more than 95%.
  • a non-neutralizing anti-ADM antibody, or anti-ADM antibody fragment or anti-ADM non-Ig scaffold blocks the bioactivity of ADM less than 100%, preferably to less than 95%, preferably to less than 90%, more preferred to less than 80% and even more preferred to less than 50%.
  • the residual bioactivity of ADM bound to the non-neutralizing anti-ADM antibody, or anti-ADM antibody fragment or anti-ADM non-Ig scaffold would be more than 0%, preferably more than 5%, preferably more than 10%, more preferred more than 20%, more preferred more than 50%.
  • molecule(s) being it an antibody, or an antibody fragment or a non-Ig scaffold with “non-neutralizing anti-ADM activity”, collectively termed here for simplicity as “non-neutralizing” anti-ADM antibody, antibody fragment, or non-Ig scaffold, that e.g. blocks the bioactivity of ADM to less than 80%, is defined as
  • an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is used, wherein said antibody or an adrenomedullin antibody fragment blocks the bioactivity of ADM to less than 80%, preferably less than 50% (of baseline values). This is in the sense of blocking the circulating ADM of no more than 80% or no more than 50%, respectively.
  • said limited blocking of the bioactivity of ADM occurs even at excess concentration of the antibody, fragment or scaffold, meaning an excess of the antibody, fragment or scaffold in relation to ADM.
  • Said limited blocking is an intrinsic property of the ADM binder itself. This means that said antibody, fragment or scaffold has a maximal inhibition of 80% or 50% respectively.
  • said anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold would block the bioactivity of ADM to at least 5%.
  • the provided anti-ADM antibodies, anti-ADM antibody fragments, and anti-ADM non-Ig scaffolds do not neutralize the respective circulating ADM bioactivity.
  • bioactivity is defined as the effect that a substance takes on a living organism or tissue or organ or functional unit in vivo or in vitro (e.g. in an assay) after its interaction.
  • ADM bioactivity this may be the effect of ADM in a human recombinant Adrenomedullin receptor cAMP functional assay.
  • bioactivity is defined via an Adrenomedullin receptor cAMP functional assay. The following steps may be performed in order to determine the bioactivity of ADM in such an assay:
  • a maximal inhibition in said ADM bioassay of 50% means that said anti-ADM antibody or said anti-adrenomedullin antibody fragment or said anti-adrenomedullin non-Ig scaffold, receptively, blocks the bioactivity to 50% of baseline values.
  • a maximal inhibition in said ADM bioassay of 80% means that said anti-ADM antibody or said anti-adrenomedullin antibody fragment or said anti-adrenomedullin non-Ig scaffold, respectively, blocks the bioactivity of ADM to 80%. This is in the sense of blocking the ADM bioactivity to not more than 80%. This means approximately 20% residual ADM bioactivity remains present.
  • the expression “blocks the bioactivity of ADM” in relation to the herein disclosed anti-ADM antibodies, anti-ADM antibody fragments, and anti-ADM non-Ig scaffolds should be understood as mere decreasing the bioactivity of ADM, preferably decreasing circulating ADM bioactivity from 100% to 20% remaining ADM bioactivity at maximum, preferably decreasing the ADM bioactivity from 100% to 50% remaining ADM bioactivity; but in any case there is ADM bioactivity remaining that can be determined as detailed above.
  • the bioactivity of ADM may be determined in a human recombinant Adrenomedullin receptor cAMP functional assay (Adrenomedullin Bioassay) according to Example 2.
  • a modulating anti-ADM antibody or a modulating anti-ADM adrenomedullin antibody fragment or a modulating anti-ADM adrenomedullin non-Ig scaffold is used in therapy of acute disease or acute condition of a patient for regulation of fluid balance.
  • Such a modulating anti-ADM antibody or a modulating anti-ADM adrenomedullin antibody fragment or a modulating anti-ADM adrenomedullin non-Ig scaffold may be especially useful in the treatment of sepsis.
  • a modulating anti-ADM antibody or a modulating anti-ADM adrenomedullin antibody fragment or a modulating anti-adrenomedullin non-Ig scaffold enhances the bioactivity of ADM in the early phase of sepsis and reduces the damaging effects of ADM in the late phase of sepsis.
  • a “modulating” antibody or a modulating adrenomedullin antibody fragment or a modulating adrenomedullin non-Ig scaffold is an antibody or an adrenomedullin antibody fragment or non-Ig scaffold that enhances the half life (t 1/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10%, preferably at least, 50%, more preferably >50%, most preferably >100% and blocks the bioactivity of ADM to less than 80%, preferably less than 50%.
  • blocking the ADM bioactivity is in the sense of no more than 80%, and thus 20% residual ADM bioactivity. The same applies to blocking the ADM bioactivity to no more than 50%, and thus residual 50% ADM bioactivity.
  • Such a modulating anti-ADM antibody or a modulating anti-ADM adrenomedullin antibody fragment or a modulating anti-adrenomedullin non-Ig scaffold offers the advantage that the dosing of the administration is facilitated.
  • the combination of partially blocking or partially reducing Adrenomedullin bioactivity and increase of the in vivo half life (increasing the Adrenomedullin bioactivity) leads to beneficial simplificity of anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold dosing.
  • the activity lowering effect is the major impact of the antibody or fragment or scaffold, limiting the (negative) effect of Adrenomedullin.
  • the biological effect of anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is a combination of lowering (by partially blocking) and increase by increasing the Adrenomedullin half life.
  • the non-neutralizing and modulating anti-Adrenomedullin antibody or anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold acts like an ADM bioactivity buffer in order to keep the bioactivity of ADM within a certain physiological range.
  • the dosing of the anti-ADM antibody/fragment/scaffold in e.g. sepsis may be selected from an excessive concentration, because both sepsis phases (early and late) benefit from excessive anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold treatment in case of a modulating effect.
  • This means, in case of a modulating anti-ADM antibody or modulating anti-ADM antibody fragment or modulating anti-ADM scaffold dosing in sepsis may be as follows:
  • Adrenomedullin in septic shock is 226+/ ⁇ 66 fmol/ml (Nishio et al., “Increased plasma concentrations of adrenomedullin correlate with relaxation of vascular tone in patients with septic shock.”, Crit Care Med. 1997, 25(6):953-7), an equimolar concentration of antibody or fragment or scaffold is 42.5 ⁇ g/l blood, (based on 6 l blood volume/80 kg body weight) 3.2 ⁇ g/kg body weight.
  • Excess means at least double (mean) septic shock Adrenomedullin concentration, at least >3 ⁇ g anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold/kg body weight, preferred at least 6.4 ⁇ g anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold/kg body weight.
  • the anti-ADM antibody is a monoclonal antibody or an anti-ADM antibody fragment thereof.
  • the anti-ADM antibody or the anti-ADM antibody fragment is a human or humanized antibody or derived therefrom.
  • one or more (murine) CDR's are grafted into a human antibody or antibody fragment.
  • Subject matter of the present invention in one aspect is a human CDR-grafted antibody or antibody fragment thereof that binds to ADM, wherein the human CDR-grafted antibody or antibody fragment thereof comprises an antibody heavy chain (H chain) comprising
  • SEQ ID NO: 4 QSIVYSNGNTY SEQ ID NO: 5 RVS and/or SEQ ID NO: 6 FQGSHIPYT.
  • the anti-ADM antibody has a sequence selected from the group comprising: SEQ ID NO 7, 8, 9, 10, 11, 12, 13 and 14.
  • the anti-ADM antibody or anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention exhibits an affinity towards human ADM in such that affinity constant is greater than 10 ⁇ 7 M, preferred 10 ⁇ 8 M, preferred affinity is greater than 10 ⁇ 9 M, most preferred higher than 10 ⁇ 10 M.
  • affinity constants may be determined according to the method as described in Example 1.
  • the anti-ADM antibody or the anti-ADM antibody fragment or the anti-ADM non-Ig scaffold is used for reducing the risk of mortality during said chronic or acute disease or acute condition of a patient.
  • Chronic or acute disease or acute condition may be a disease or condition selected from the group comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, artheriosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction or capillary leakage, trauma, poisoning, surgery.
  • severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), sepsis
  • other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, artheriosclerosis
  • shock e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction or capillary leakage, trauma, poisoning, surgery.
  • the patient may be has a chronic or acute disease or condition as primary and underlying disease as outlined in the above paragraph; however, the anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold pursuant to the invention are not intended for primary therapy of said diseases, but rather for regulating the fluid balance of a patient that is in need of administration of fluids, which can thus be considered as an acute disease or acute condition besides the primary disease.
  • said need for fluid administration may be associated with a primary underlying disease but this is not mandatory within the scope of the instant invention.
  • the anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is used in therapy of an acute disease or acute condition of a patient according to the present invention wherein said patient is an ICU patient.
  • the anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is used in therapy of an acute disease of a patient according to the present invention, wherein said patient is critically ill. Critically ill means that the patient is having a disease or state in which death is possible or imminent.
  • Subject of the present invention is further an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease of a patient according to the present invention, wherein said antibody or fragment is to be used in combination of ADM binding protein.
  • ADM binding protein is also naturally present in the circulation of said patient.
  • ADM binding protein also denotes ADM-binding-protein-1 (complement factor H), which however is not a non-neutralizing and modulating anti-ADM antibody, anti-ADM antibody fragment, or anti-ADM non-Ig scaffold as in accordance with the invention.
  • Subject of the present invention is further an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient according to the present invention wherein said antibody or fragment or scaffold is to be used in combination with further active ingredients.
  • Subject matter of the invention is also an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold to be used in combination with a primary medicament wherein said combination is for use in therapy of an acute disease or acute condition of a patient for regulating the fluid balance of said patient.
  • ADM anti-Adrenomedullin
  • Primary medicament means a medicament that acts against the primary cause of said disease or condition.
  • Said primary medicament may be antibiotics in case of infections.
  • said primary cause is related to the primary and underlying disease or condition, and is not related to the acute disease or acute condition that is associated with fluid imbalance of a patient, for which the herein provided therapy of regulating the fluid balance is intended.
  • said combinations are to be used in combination with vasopressors e.g. catecholamine wherein said further combination is for use in therapy of an acute disease or condition of a patient for regulating the fluid balance.
  • vasopressors e.g. catecholamine
  • said patient having a chronic or acute disease or chronic condition being in need for regulating the fluid balance is characterized by the need of the patient to get administration of vasopressors e.g. of catecholamine.
  • said patient is having a chronic or acute disease or chronic condition such as cancer, or diabetes, and thus this can be considered as primary, underlying disease, but in addition said patient is in acute need for regulating the fluid balance that is may be due to another acute disease or acute condition such as e.g. SIRS, sepsis, severe sepsis, or shock, or septic shock.
  • a chronic or acute disease or chronic condition such as cancer, or diabetes
  • another acute disease or acute condition such as e.g. SIRS, sepsis, severe sepsis, or shock, or septic shock.
  • an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold to be used in combination with ADM binding protein and/or further active ingredients for use in therapy of a patient in need of a treatment of vasopressors e.g. catecholamine treatment.
  • ADM anti-Adrenomedullin
  • said combinations are to be used in combination with fluids administered intravenously, wherein said combination is for use in therapy of an acute disease or condition of a patient for regulating the fluid balance.
  • said patient having a chronic or acute disease or acute condition being in need for regulating the fluid balance is characterized by the need of the patient to get intravenous fluids.
  • Subject matter of the invention in one specific embodiment is, thus, an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold in combination with ADM binding protein and/or further active ingredients for use in therapy of a patient in need of intravenous fluids.
  • ADM anti-Adrenomedullin
  • Said anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold or combinations thereof with ADM binding protein and/or further active ingredients may be used in combination with vasopressors e.g. catecholamine and/or with fluids administered intravenously for use in therapy of an acute disease or acute condition of a patient for regulating the fluid balance.
  • vasopressors e.g. catecholamine and/or with fluids administered intravenously for use in therapy of an acute disease or acute condition of a patient for regulating the fluid balance.
  • Subject matter of the invention is also an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention to be used in combination with TNF-alpha-antibodies.
  • TNF-alpha-antibodies are commercially available for the treatment of patients.
  • Subject of the present invention is further a pharmaceutical formulation comprising an anti-ADM antibody or anti-ADM antibody fragment or anti-ADM antibody scaffold according to the present invention.
  • Subject of the present invention is further a pharmaceutical formulation according to the present invention wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution.
  • Said pharmaceutical formulation may be administered intra-muscular.
  • Said pharmaceutical formulation may be administered intra-vascular.
  • Said pharmaceutical formulation may be administered via infusion.
  • the pharmaceutical formulation in accordance with the invention as may be administered intra-muscular, intra-vascular, or via infusion is preferably administered to a patient for regulating the systemic fluid balance with the proviso that said patient is in need of regulating the fluid balance.
  • the pharmaceutical formulation according to the present invention is to be administered to a patient for regulating the systemic fluid balance with the proviso that said patient is in need of regulating the fluid balance.
  • the expression “regulating fluid balance” with the context of the instant invention is directed to any correction of a manifested—imbalance—of a patient's fluid balance due to an underlying chronic or acute disease or acute condition. Said correction is in favour of re-establishing normotension in said patients.
  • the person skilled in the art is fully aware that blood pressure in general, as well as hyper- and hypotension is closely related to the fluid balance of a patient.
  • Fluid balance is the balance of the input and the output of fluids in the body to allow metabolic processes to function. Dehydration is defined as a 1% or greater loss of body mass as a result of fluid loss.
  • the three elements for assessing fluid balance and hydration status are: clinical assessment, body weight and urine output; review fluid balance charts and review of blood chemistry. All this is very well known to a man skilled in the art (Alison Shepherd, Nursing Tomes 19.07.11/Vol 107 No 28, pages 12 to 16).
  • a person in need of regulating the fluid balance and/or improving the fluid balance of such patients is a person that has a 1% or greater loss of body mass as a result of fluid loss.
  • the fluid balance may be assessed according to Scales and Pilsworth (2008) Nursing Standard 22:47, 50-57.
  • normal urine output is in the range of 0.5 to 2 ml/kg of body weight per hour.
  • the minimum acceptable urine output for a patient with normal renal function is 0.5 ml/kg per hour. All these standards may be used to assess whether a patient is in need for regulating the fluid balance and/or improving the fluid balance.
  • subject of the present invention is further a pharmaceutical formulation according to the present invention wherein said pharmaceutical formulation is in a dried state to be reconstituted before use.
  • subject of the present invention is further a pharmaceutical formulation according to the present invention wherein said pharmaceutical formulation is in a freeze-dried state.
  • the antibodies, antibody fragments and non-Ig scaffolds of the example portion in accordance with the invention are binding to ADM, and thus should be considered as anti-ADM antibodies/antibody fragments/non-Ig scaffolds.
  • Peptides for immunization were synthesized, see Table 1, OPT Technologies, Berlin, Germany) with an additional N-terminal Cystein (if no Cystein is present within the selected ADM-sequence) residue for conjugation of the peptides to Bovine Serum Albumin (BSA).
  • BSA Bovine Serum Albumin
  • the peptides were covalently linked to BSA by using Sulfolink-coupling gel (Perbio-science, Bonn, Germany). The coupling procedure was performed according to the manual of Perbio.
  • the murine antibodies were generated according to the following method:
  • a Balb/c mouse was immunized with 100 ⁇ g Peptide-BSA-Conjugate at day 0 and 14 (emulsified in 100 ⁇ l complete Freund's adjuvant) and 50 ⁇ g at day 21 and 28 (in 100 ⁇ l incomplete Freund's adjuvant).
  • the animal received 50 ⁇ g of the conjugate dissolved in 100 ⁇ l saline, given as one intraperitoneal and one intra-venous injection.
  • Spenocytes from the immunized mouse and cells of the myeloma cell line SP2/0 were fused with 1 ml 50% polyethylene glycol for 30 s at 37° C. After washing, the cells were seeded in 96-well cell culture plates. Hybrid clones were selected by growing in HAT medium [RPMI 1640 culture medium supplemented with 20% fetal calf serum and HAT-Supplement]. After two weeks the HAT medium is replaced with HT Medium for three passages followed by returning to the normal cell culture medium.
  • the cell culture supernatants were primary screened for antigen specific IgG antibodies three weeks after fusion.
  • the positive tested microcultures were transferred into 24-well plates for propagation. After retesting, the selected cultures were cloned and recloned using the limiting-dilution technique and the isotypes were determined.
  • Antibodies were produced via standard antibody production methods (Marx et al, Monoclonal Antibody Production, ATLA 25, 121, 1997), and purified via Protein A. The antibody purities were >95% based on SDS gel electrophoresis analysis.
  • Human Antibodies were produced by means of phage display according to the following procedure:
  • the human naive antibody gene libraries HALT/8 were used for the isolation of recombinant single chain F-Variable domains (scFv) against adrenomedullin peptide.
  • the antibody gene libraries were screened with a panning strategy comprising the use of peptides containing a biotin tag linked via two different spacers to the adrenomedullin peptide sequence.
  • a mix of panning rounds using non-specifically bound antigen and streptavidin bound antigen were used to minimize background of non-specific binders.
  • the eluted phages from the third round of panning have been used for the generation of monoclonal scFv expressing E. coli strains.
  • Positive clones have been selected based on positive ELISA signal for antigen and negative for streptavidin coated micro titer plates.
  • the scFv open reading frame has been cloned into the expression plasmid pOPE107 (Hust et al., J. Biotechn. 2011), captured from the culture supernatant via immobilised metal ion affinity chromatography and purified by a size exclusion chromatography.
  • the monoclonal antibodies were raised against the below depicted ADM regions of human and murine ADM, respectively.
  • the following table represents a selection of obtained antibodies used in further experiments. Selection was based on target region:
  • Fab and F(ab) 2 fragments were done by enzymatic digestion of the murine full length antibody NT-M.
  • Antibody NT-M was digested using a) the pepsin-based F(ab) 2 Preparation Kit (Pierce 44988) and b) the papain-based Fab Preparation Kit (Pierce 44985).
  • the fragmentation procedures were performed according to the instructions provided by the supplier. Digestion was carried out in case of F(ab) 2 -fragmentation for 8 h at 37° C.
  • the Fab-fragmentation digestion was carried out for 16 h, respectively.
  • the immobilized papain was equilibrated by washing the resin with 0.5 ml of Digestion Buffer and centrifuging the column at 5000 ⁇ g for 1 minute. The buffer was discarded afterwards.
  • the desalting column was prepared by removing the storage solution and washing it with digestion buffer, centrifuging it each time afterwards at 1000 ⁇ g for 2 minutes.
  • 0.5 ml of the prepared IgG sample where added to the spin column tube containing the equilibrated Immobilized Papain. Incubation time of the digestion reaction was done for 16 h on a tabletop rocker at 37° C. The column was centrifuged at 5000 ⁇ g for 1 minute to separate digest from the Immobilized Papain.
  • the resin was washed with 0.5 ml PBS and centrifuged at 5000 ⁇ g for 1 minute.
  • the wash fraction was added to the digested antibody that the total sample volume was 1.0 ml.
  • the NAb Protein A Column was equilibrated with PBS and IgG Elution Buffer at room temperature. The column was centrifuged for 1 minute to remove storage solution (contains 0.02% sodium azide) and equilibrated by adding 2 ml of PBS, centrifuge again for 1 minute and the flow-through discarded.
  • the sample was applied to the column and resuspended by inversion. Incubation was done at room temperature with end-over-end mixing for 10 minutes.
  • the column was centrifuged for 1 minute, saving the flow-through with the Fab fragments.
  • the immobilized Pepsin was equilibrated by washing the resin with 0.5 ml of Digestion Buffer and centrifuging the column at 5000 ⁇ g for 1 minute. The buffer was discarded afterwards.
  • the desalting column was prepared by removing the storage solution and washing it with digestion buffer, centrifuging it each time afterwards at 1000 ⁇ g for 2 minutes.
  • 0.5 ml of the prepared IgG sample where added to the spin column tube containing the equilibrated Immobilized Pepsin. Incubation time of the digestion reaction was done for 16 h on a tabletop rocker at 37° C. The column was centrifuged at 5000 ⁇ g for 1 minute to separate digest from the Immobilized Papain.
  • the resin was washed with 0.5 mL PBS and centrifuged at 5000 ⁇ g for 1 minute.
  • the wash fraction was added to the digested antibody that the total sample volume was 1.0 ml.
  • the NAb Protein A Column was equilibrated with PBS and IgG Elution Buffer at room temperature. The column was centrifuged for 1 minute to remove storage solution (contains 0.02% sodium azide) and equilibrated by adding 2 mL of PBS, centrifuge again for 1 minute and the flow-through discarded.
  • the sample was applied to the column and resuspended by inversion. Incubation was done at room temperature with end-over-end mixing for 10 minutes.
  • the column was centrifuged for 1 minute, saving the flow-through with the Fab fragments.
  • the antibody fragment was humanized by the CDR-grafting method (Jones, P. T., Dear, P. H., Foote, J., Neuberger, M. S., and Winter, G. (1986) Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature 321, 522-525).
  • Total RNA extraction Total RNA was extracted from NT-H hybridomas using the Qiagen kit.
  • RT-PCR QIAGEN® OneStep RT-PCR Kit (Cat No. 210210) was used. RT-PCR was performed with primer sets specific for the heavy and light chains. For each RNA sample, 12 individual heavy chain and 11 light chain RT-PCR reactions were set up using degenerate forward primer mixtures covering the leader sequences of variable regions. Reverse primers are located in the constant regions of heavy and light chains. No restriction sites were engineered into the primers.
  • Second-round semi-nested PCR The RT-PCR products from the first-round reactions were further amplified in the second-round PCR. 12 individual heavy chain and 11 light chain RT-PCR reactions were set up using semi-nested primer sets specific for antibody variable regions.
  • Annotation for the antibody fragment sequences (SEQ ID NO: 7-14): bold and underline are the CDR 1, 2, 3 in chronologically arranged; italic are constant regions; hinge regions are highlighted with bold letters and the histidine tag with bold and italic letters; framework point mutation have a grey letter-background.
  • A-VH-C SEQ ID NO: 7 QVQLQQSGAELMKPGASVKISCKAT GYTFSRYW IEWVKQRPGHGLEWIGE ILPGSGST NYNEKFKGKATITADTSS NTAYMQLSSLTSEDSAVYYC WGQGTTLTVSSAS TKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLS SVVTVPSSSLGTQTYICNVNHKPSNTKVDKRV EPK (AM-VH1) SEQ ID NO: 8 QVQLVQSGAEVKKPGSSVKVSCKAS GYTFSRYW ISWVRQAPGQGLEWMGR ILPGSGS T NYAQKFQGRVTITADE STSTAYMELSSLRSEDTAVYYC WGQGTTVTVSSA STKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLS
  • Adrenomedullin Bioassay The effect of selected ADM-antibodies on ADM-bioactivity was tested in an human recombinant Adrenomedullin receptor cAMP functional assay (Adrenomedullin Bioassay).
  • Adrenomedullin (CRLR+RAMP3)
  • CHO-K1 cells expressing human recombinant adrenomedullin receptor (FAST-027C) grown prior to the test in media without antibiotic were detached by gentle flushing with PBS-EDTA (5 mM EDTA), recovered by centrifugation and resuspended in assay buffer (KRH: 5 mM KCl, 1.25 mM MgSO4, 124 mM NaCl, 25 mM HEPES, 13.3 mM Glucose, 1.25 mM KH2PO4, 1.45 mM CaCl2, 0.5 g/1 BSA).
  • PBS-EDTA 5 mM EDTA
  • Dose response curves were performed in parallel with the reference agonists (hADM or mADM).
  • hADM 22-52 was used as reference antagonist.
  • the anti-h-ADM antibodies (NT-H, MR-H, CT-H) were tested for antagonist activity in human recombinant adrenomedullin receptor (FAST-027C) cAMP functional assay in the presence of 5.63 nM Human ADM 1-52, at the following final antibody concentrations: 100 ⁇ g/ml, 20 ⁇ g/ml, 4 ⁇ g/ml, 0.8 ⁇ g/ml, 0.16 ⁇ g/ml.
  • the anti-m-ADM antibodies (NT-M, MR-M, CT-M) were tested for antagonist activity in human recombinant adrenomedullin receptor (FAST-027C) cAMP functional assay in the presence of 0.67 nM Mouse ADM 1-50, at the following final antibody concentrations: 100 ⁇ g/ml, 20 ⁇ g/ml, 4 ⁇ g/ml, 0.8 ⁇ g/ml, 0.16 ⁇ g/ml. Data were plotted relative inhibition vs. antagonist concentration (see FIGS. 3 a to 3 l ). The maximal inhibition by the individual antibody is given in table 3.
  • the stabilizing effect of human ADM by human ADM antibodies was tested using a hADM immunoassay.
  • the technology used was a sandwich coated tube luminescence immunoassay, based on Acridinium ester labelling.
  • CT-H 100 ⁇ g (100 ul) CT-H (1 mg/ml in PBS, pH 7.4, AdrenoMed AGGermany) was mixed with 10 ⁇ l Acridinium NHS-ester (1 mg/ml in acetonitrile, InVent GmbH, Germany) (EP 0353971) and incubated for 20 min at room temperature.
  • Labelled CT-H was purified by Gel-filtration HPLC on Bio-Sil® SEC 400-5 (Bio-Rad Laboratories, Inc., USA) The purified CT-H was diluted in (300 mmol/L potassium phosphate, 100 mmol/L NaCl, 10 mmol/L Na-EDTA, 5 g/L Bovine Serum Albumin, pH 7.0). The final concentration was approx.
  • Polystyrene tubes (Greiner Bio-One International AG, Austria) were coated (18 h at room temperature) with MR-H (AdrenoMed AG, Germany) (1.5 ⁇ g MR-H/0.3 mL 100 mmol/L NaCl, 50 mmol/L TRIS/HCl, pH 7.8). After blocking with 5% bovine serum albumine, the tubes were washed with PBS, pH 7.4 and vacuum dried.
  • the assay was calibrated, using dilutions of hADM
  • sample 50 ⁇ l was pipetted into coated tubes, after adding labeleld CT-H (200 ⁇ l), the tubes were incubated for 4 h at 4° C. Unbound tracer was removed by washing 5 times (each 1 ml) with washing solution (20 mM PBS, pH 7.4, 0.1% Triton X-100).
  • FIG. 4 shows a typical hADM dose/signal curve. And an hADM dose signal curve in the presence of 100 ⁇ g/mL antibody NT-H.
  • NT-H did not affect the described hADM immunoassay.
  • Human ADM was diluted in human Citrate plasma (final concentration 10 nM) and incubated at 24° C. At selected time points, the degradation of hADM was stopped by freezing at ⁇ 20° C. The incubation was performed in absence and presence of NT-H (100 ⁇ g/ml). The remaining hADM was quantified by using the hADM immunoassay described above.
  • FIG. 5 shows the stability of hADM in human plasma (citrate) in absence and in the presence of NT-H antibody.
  • the half life of hADM alone was 7.8 h and in the presence of NT-H, the half life was 18.3 h. (2.3 times higher stability).
  • CLP cecal ligation and puncture
  • mice were tested versus: vehicle and versus control compound treatment.
  • Each group contained 5 mice for blood drawing after 1 day for BUN (serum blood urea nitrogen test) determination. Ten further mice per each group were followed over a period of 4 days.
  • BUN serum blood urea nitrogen test
  • BUN Blood urea nitrogen
  • mice After 4 days 70% of the mice survived when treated with NT-M antibody. When treated with MR-M antibody 30% of the animals survived and when treated with CT-M antibody 10% of the animals survived after 4 days. In contrast thereto all mice were dead after 4 days when treated with unspecific mouse IgG. The same result was obtained in the control group where PBS (phosphate buffered saline) was administered to mice.
  • PBS phosphate buffered saline
  • the blood urea nitrogen or BUN test is used to evaluate kidney function, to help diagnose kidney disease, and to monitor patients with acute or chronic kidney dysfunction or failure.
  • NT-M FAB2 was tested versus: vehicle and versus control compound treatment. Treatment was performed after full development of sepsis, 6 hours after CLP (late treatment). Each group contained 4 mice and were followed over a period of 4 days.
  • mice were anesthetized by intraperitoneal injection of 120 ⁇ g/g Ketamin, 1.25 ⁇ g/g Midazolam and 0.25 ⁇ g/g Fentanyl.
  • body temperature was kept at 37-38° C.
  • a 1 cm midline abdominal section was performed to get access to the cecum.
  • the cecum then was ligated with 3-0 silk tie close to the basis and a single puncture with a 18-gauge needle was applied. The cecum was returned and the incision was closed again (4-0 tie).
  • mice For the compensation of perioperative loss of liquids, 0.5 ml lacted Ringer's solution with 1 ⁇ g/g Buprenorphin as analgetic was injected subcutaneously in dorsal dermis. For antibiosis the mice received Ceftriaxon 30 ⁇ g/g and Clindamycin 30 ⁇ g/g subcutaneously via the lower extremities. After CLP surgery the animal were kept in an adequately heated environment with water and food ad libitum.
  • test substance antibody NT-M was applied in a concentration of 500 ⁇ g/ml in phosphate buffered saline (PBS) via injection into the penis vein for a dose of 2 mg per kg body weight (dose volume 88-120 ⁇ l) (5 animals).
  • PBS phosphate buffered saline
  • the control group (6 animals) received a corresponding amount of the vehicle PBS solution without antibody (4 ⁇ l/g, 88-120 ⁇ l) immediately after CLP surgery.
  • Group 1 (5 animals) received the antibody NT-M 15.5 h after CLP
  • group 2 received the antibody NT-M immediately after CLP surgery
  • group 3 received a comparable amount of PBS (40/g).
  • 16 hour incubation post CLP to allow the polymicrobial sepsis to progress
  • the experiment was continued with monitoring and interventions comparable to an intensive medical care regime. Therefore, after weighing the animals were anesthetized as described in the CLP surgery part (except the late treated animals, which were anesthized before treatment). Body temperature was maintained at 37-38° C. for the rest of the experiment.
  • vena jugularis externa dextra with a continuous infusion of Ketamin 30 ⁇ g/g ⁇ h and Fentanyl 0.3 ⁇ g/g ⁇ h.
  • the right aorta carotis communis was cannulated for continuous monitoring of heart rate and the mean arterial pressure (MAP).
  • the mean arterial pressure was maintained at MAP>65 mmHg via intravenous (V. jugularis) infusion of colloids (80 ⁇ L/g ⁇ h, Hextend®) and, if needed, Noradrenalin dissolved in colloids as vasopressor. Blood samples (120 ⁇ l) were taken via the cannulated A.
  • the catecholamine requirement was measured after administration of either non specific mouse IgG to a total of 6 mice as control group, NT-murine antibody to a group of 5 mice immediately after CLP (early treatment) or NT-murine antibody to a group of 3 mice 15.5 h after CLP (late treatment).
  • the reduction of the catecholamine requirement is a measure for the stabilization of the circulation.
  • the data show that the ADM antibody, especially the NT-M antibody, leads to a considerable stabilization of the circulation and to a considerable reduction of the catecholamine requirement.
  • the circulation-stabilizing effect was given in early treatment (immediately after CLP) and treatment after full sepsis development (late treatment) (see FIG. 7 ).
  • the fluid balance was improved by about 20-30%, also in both, early and late treatment.
  • the data show that the use of ADM antibody, especially the use of NT ADM antibody, is favorable for regulating the fluid balance in patients. (see table 8 and FIGS. 8 and 9 ).
  • Kidney function creatinine mean creatinine concentration ( ⁇ g/mL) clearance ( ⁇ L/min) control mouse IgG (MW) 2.6 ⁇ g/ml 174 ⁇ l/min NT-M (MW) 1.5 ⁇ g/ml 373 ⁇ l/min Relative change ⁇ 42% (42%) +114% (114%) (amelioration)
  • NF- ⁇ B nuclear factor kappa-light-chain gene enhancer in B cells
  • mice 12-15 week old male C57Bl/6 mice (Charles River Laboratories, Germany) were used for the study. 6 mice were treated with (10 ul/g bodyweight) dose of NT-M, 0.2 mg/ml. As control, 6 mice were treated with (10 ⁇ l/g body weight) PBS. Survival and physical condition was monitored for 14 days. The mortality was 0 in both groups, there were no differences in physical condition between NT-M and control group.
  • a non-septic acute kidney injury model has been established, which makes use of the nephrotoxicity induced by Gentamicin (Chiu P J S. Models used to assess renal functions. Drug Develop Res 32:247-255, 1994). This model was used to assess whether treatment with anti-Adrenomedullin antibody can improve kidney function.
  • c NT-M at 4 mg/kg was injected intravenously (i.v.) 5 min before gentamicin on Day 0, followed by 2 mg/kg i.v. on Days 2, 4, and 6.
  • d Plasma samples were collected in EDTA tubes (Days 1 and 3 before Test and Control article: 100 ⁇ l; Day 7: 120 ⁇ l. 24 h urine collection on ice is initiated after gentamicin on Day 0, followed by Days 2 and 6; blood collection on days 1, 3, and 7.
  • Groups of 8 male Sprague-Dawley rats weighing 250 ⁇ 20 g were employed. Animals were challenged with gentamicin at 120 mg/kg i.m. for seven consecutive days (Groups 1 and 2). Test compound (anti-adrenomedullin antibody NT-M) and vehicle (phosphate buffered saline) were injected intravenously 5 min before gentamicin on day 0, followed by injection on days 2, 4, and 6. Body weights and clinical signs were monitored daily. Twenty-four (24) hour urine collections on ice were performed on Days 0, 2, and 6. Urine specimens were assayed for concentrations of Na+ and K+, and creatinine.
  • Test compound anti-adrenomedullin antibody NT-M
  • vehicle phosphate buffered saline
  • Plasma samples for clinical chemistry were collected on Days 1 (before gentamicin), 3 (before gentamicin), and 7.
  • Serum electrolytes (Na+ and K+), creatinine, and BUN were the primary analytes that were monitored for assessing renal function.
  • Plasma samples were collected in EDTA tubes (Days 1 and 3: 100 ⁇ l; Day 7: 120 ⁇ l). Creatinine clearance was calculated.
  • Urine volume, urinary electrolytes, and creatinine are expressed as amount excreted per 100 g of animal body weight. All animals were sacrificed on Day 7. Kidneys were weighed.
  • Urine collection The animals were placed in individual cages where urine was collected for 24 h on Day 0, Day 2, and Day 6. Urine volume, urinary Na+, K+, and creatinine were measured.
  • CCr (ml/24 h) [ UCr (mg/ml) ⁇ V (ml/24 h)]/ SCr (mg/ml)
  • Treatment with anti-Adrenomedullin antibody improved several measures of kidney function on day 7 as compared to vehicle: serum creatinine 1.01 mg/dL (NT-M) vs 1.55 mg/dL (vehicle) ( FIG. 11 ), BUN 32.08 mg/dL (NT-M) vs. 52.41 mg/dL (vehicle) ( FIG. 12 ), endogenous creatinine clearance 934.43 mL/24 h (NT-M) vs. 613.34 mL/24 h (vehicle) ( FIG. 13 ), fractional secretion of Na + 0.98% (NT-M) vs. 1.75% (vehicle) ( FIG. 14 ).
  • mice CLP model In the mice CLP model described above, the effect of treatment with anti-adrenomedullin antibody NT-M on several parameters of kidney function was investigated.
  • NT-M caused a three- and two-fold higher diuresis and creatinine clearance, respectively, ultimately resulting in lower creatinine, urea, and NGAL blood concentrations at the end of the experiment (see Table 10). Moreover, keratinocyte-derived chemokine (KC) concentrations in the kidney were significantly lowered by treatment with NT-M ( FIG. 15 ).
  • Blood NGAL concentrations were measured using a commercial ELISA (mouse NGAL, RUO 042, BioPorto Diagnostics A/S, Denmark, Gentofte).
  • Urea and creatinine concentrations were measured with a capillary column (Optima-5MS, Macherey-Nagel, Düren, Germany) gas chromatography/mass spectrometry system (Agilent 5890/5970, Boblingen, Germany) using 2 H 3 -creatinine (CDN isotopes, Pointe-Claire, QU, Canada) and methyl-urea (FlukaChemikalien, Buchs, Switzerland) as internal standards.
  • Ions m/z 231 and 245, and m/z 329 and 332 were monitored for urea and creatinine analytes and internal standards, respectively. From the urine output and the plasma and urine creatinine concentrations creatinine clearance was calculated using the standard formula.
  • the kidney which was stored at ⁇ 80° C. was disrupted with a homogenizer in PBS and lysed with a 2-fold concentrated buffer for a whole cell lysate (100 mM Tris pH 7.6; 500 mM NaCl; 6 mM EDTA; 6 mM EGTA; 1% Triton-X-100; 0.5% NP 40; 10% Glycerol; Protease-Inhibitors ( ⁇ -Glycerolphosphate 2 mM; DTT 4 mM; Leupeptine 20 ⁇ M; Natriumorthovanadate 0.2 mM)) and subsequently centrifuged. The whole cell lysate was obtained out of the supernatant; the pellet consisting of cell remnants was discarded.
  • a whole cell lysate 100 mM Tris pH 7.6; 500 mM NaCl; 6 mM EDTA; 6 mM EGTA; 1% Triton-X-100; 0.5% NP 40; 10% Gly
  • the amount of protein was determined photometrically with a commercially available protein assay (Bio-Rad, Hercules, Calif.) and the specimens were adjusted in the way that the final protein concentration was 4
  • the samples for the Multiplex- and EMSA analysis were diluted 1:1 with EMSA buffer (10 mM Hepes; 50 mM KCl; 10% Glycerol; 0.1 mM EDTA; 1 mM DTT), the samples for the immuno blots 1:1 with 2-fold Sample Buffer (2% SDS; 125 mM Tris-HCL (pH 6.8 at 25° C.); 10% Glycerol; 50 mM DTT; 0.01% Bromophenol blue).
  • KC keratinocyte-derived chemokine
  • mice CLP model In the mice CLP model described above, the effect of treatment with anti-adrenomedullin antibody NT-M on the liver was investigated.
  • NT-M caused a significant lowering of keratinocyte-derived chemokine (KC) concentrations in the liver ( FIG. 16 ).
  • KC keratinocyte-derived chemokine
  • mice CLP model In the mice CLP model described above, the effect of treatment with anti-adrenomedullin antibody NT-M on several cytokines and chemokinesin the blood circulation (plasma) was investigated.
  • Plasma levels of tumor necrosis factor (TNF)- ⁇ , interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1, and keratinocyte-derived chemokine (KC) concentrations were determined using a mouse multiplex cytokine kit (Bio-Plex Pro Cytokine Assay, Bio-Rad, Hercules, Calif.), the assay was performed by using the Bio-plex suspension array system with the manufacturer's instructions (see also Wagner F, Wagner K, Weber S, Stahl B, Knöferl M W, Huber-Lang M, Seitz D H, Asfar P, Calzia E, Senftleben U, Gebhard F, Georgieff M, Raderraum P, Hysa V.
  • the samples were incubated with antibodies chemically attached to fluorescent-labeled micro beads. Thereafter, premixed detection antibodies were added to each well, and subsequently, streptavidin-phycoerythrin was added. Beads were then re-suspended, and the cytokines reaction mixture was quantified using the Bio-Plex protein array reader. Data were automatically processed and analyzed by Bio-Plex Manager Software 4.1 using the standard curve produced from recombinant cytokine standards. Levels below the detection limit of the assays were set to zero for statistical purposes.
  • Plasma levels and kidney tissue concentrations of tumor necrosis factor (TNF)- ⁇ , interleukin (IL)-6 and IL-10, monocyte chemoattractant protein (MCP)-1, and keratinocyte-derived chemokine (KC) were determined using a commercially available “Multiplex Cytokine Kit” (Bio-Plex Pro Precision Pro Cytokine Assay, Bio-Rad, Hercules, Calif.), which allows to collect several parameters out of one single sample.
  • the fluorescence-labeled microspheres (“beads”) were added to a 96-well plate, followed by two washing steps, the addition of internal standards and the addition of plasma- and kidney homogenate samples. During the subsequent incubation the single cytokines bind to the antibodies attached to polystyrene-beads. After the addition of the cytokine-specific biotin-labeled antibodies, which are for the detection of the single cytokines, and an additional incubation time, subsequently phycoerythrin-labeled streptavidine was added.
  • NT-M caused a significant lowering of plasma concentrations of IL-6 ( FIG. 17 ), IL-10 ( FIG. 18 ), keratinocyte-derived chemokine (KC) ( FIG. 19 ), monocyte chemoattractant protein-1 (MCP-1) ( FIG. 20 ), TNF-alpha ( FIG. 21 ).
  • b NT-M at 4 mg/kg was injected intravenously (i.v.) 5 min before reperfusion on day 0, followed by 2 mg/kg i.v. each on days 1 and 2.
  • Groups of 8 male Sprague-Dawley rats weighing 250 to 280 g were used. The animals were kept on a 12-hr light/dark cycle and receive a standard diet with distilled water ad libitum. The animals receive fluid supplements (0.9% NaCl and 5% dextrose/1:1, 10 ml/kg p.o.) 30 min prior to surgery (day 0). The rats were anaesthetized with pentobarbital (50 mg/kg, i.p.). The abdominal cavity was exposed via a midline incision, followed by intravenous administration of heparin (100 U/kg, i.v.) and both renal arteries were occluded for 45 min by using vascular clamps.
  • pentobarbital 50 mg/kg, i.p.
  • test compound N-M
  • vehicle phosphate buffered saline
  • Urine collection The 24-h urine collection on ice was initiated at 24 h before ischemia/reperfusion on day ⁇ 1 ( ⁇ 24 h to 0 h), and day 0 (0-24 h), day 1 (24-48 h) and day 2 (48-72 h) after reperfusion,
  • Blood collection 0.4 ml blood was collected through the tail vein into EDTA tubes at 0 h (before I RI surgery), 24 h (before vehicle or TA), 48 h (before vehicle or TA) and 72 h for determination of plasma creatinine/Na+/K+, and BUN; 2 ml blood was collected through venal cava terminally.
  • the creatinine clearance (CCr) was calculated as follows:
  • CCr (ml/24 h) [ UCr (mg/ml) ⁇ V (ml/24 h)]/ PCr (mg/ml)
  • fractional excretion of Na+ (FENa), or percentage of the filtered sodium that is excreted into the final urine, is a measure of tubular Na+ reabsorptive function. It was computed as follows:
  • FENa (%) 100 ⁇ [UNa ( ⁇ Eq/ml) ⁇ V (ml/24 h)]/ PNa ( ⁇ Eq/ml) ⁇ CCr (ml/24 h)
  • Blood urea nitrogen (BUN) showed a strong increase in the vehicle group (0 h: 17.49 mg/dL, 24 h: 98.85 mg/dL, 48 h: 109.84 mg/dL, 72 h: 91.88 mg/dL), which was less pronounced with NT-M treatment (0 h: 16.33 mg/dL, 24 h: 84.2 mg/dL, 48 h: 82.61 mg/dL, 72 h: 64.54 mg/dL) ( FIG. 22 ).
  • Serum creatinine developed similarly: Vehicle group (0 h: 0.61 mg/dL, 24 h: 3.3 mg/dL, 48 h: 3.16 mg/dL, 72 h: 2.31 mg/dL), NT-M group: (0 h: 0.59 mg/dL, 24 h: 2.96 mg/dL, 48 h: 2.31 mg/dL, 72 h: 1.8 mg/dL) ( FIG. 23 ).
  • FIG. 1 a
  • FIG. 1 b
  • FIG. 1 c
  • FIG. 2
  • FIG. 3 is a diagrammatic representation of FIG. 3 :
  • FIG. 4
  • This figure shows a typical hADM dose/signal curve. And an hADM dose signal curve in the presence of 100 ⁇ g/mL antibody NT-H.
  • FIG. 5
  • This figure shows the stability of hADM in human plasma (citrate) in absence and in the presence of NT-H antibody.
  • FIG. 6 is a diagrammatic representation of FIG. 6 :
  • FIG. 7
  • FIG. 8
  • FIG. 9 is a diagrammatic representation of FIG. 9 .
  • FIG. 10 is a diagrammatic representation of FIG. 10 :
  • NF- ⁇ B nuclear factor kappa-light-chain gene enhancer in B cells
  • ESA electophoretic mobility shift assay
  • FIG. 11 is a diagrammatic representation of FIG. 11 :
  • FIG. 12
  • FIG. 13 is a diagrammatic representation of FIG. 13 :
  • FIG. 14
  • FIG. 15
  • Keratinocyte-derived chemokine (KC) levels determined in relation to the total kidney protein extracted.
  • the white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 16
  • Keratinocyte-derived chemokine (KC) levels determined in relation to the total liver protein extracted.
  • the white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 17 is a diagrammatic representation of FIG. 17 :
  • Plasma IL-6 levels Plasma IL-6 levels.
  • the white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 18 is a diagrammatic representation of FIG. 18 :
  • Plasma IL-10 levels The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 19 is a diagrammatic representation of FIG. 19 :
  • Plasma keratinocyte-derived chemokine (KC) levels The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 20
  • Plasma monocyte chemoattractant protein-1 (MCP-1) levels The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 21 is a diagrammatic representation of FIG. 21 :
  • Plasma TNF-alpha levels The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
  • FIG. 22
  • FIG. 23 is a diagrammatic representation of FIG. 23 :
  • FIG. 24

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Emergency Medicine (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • Cardiology (AREA)
  • Communicable Diseases (AREA)
  • Mycology (AREA)
US14/358,400 2011-11-16 2012-11-16 Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment of anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease Abandoned US20140314775A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP11189452 2011-11-16
EP11189452.3 2011-11-16
EP12160015.9 2012-03-16
EP12160015 2012-03-16
PCT/EP2012/072933 WO2013072514A1 (en) 2011-11-16 2012-11-16 Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/072933 A-371-Of-International WO2013072514A1 (en) 2011-11-16 2012-11-16 Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/045,576 Continuation US10227405B2 (en) 2011-11-16 2016-02-17 Methods of modulating the activity of adrenomedullin in a subject in need of regulation of fluid balance by administering an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment

Publications (1)

Publication Number Publication Date
US20140314775A1 true US20140314775A1 (en) 2014-10-23

Family

ID=48429017

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/358,400 Abandoned US20140314775A1 (en) 2011-11-16 2012-11-16 Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment of anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease
US15/045,576 Active 2032-12-19 US10227405B2 (en) 2011-11-16 2016-02-17 Methods of modulating the activity of adrenomedullin in a subject in need of regulation of fluid balance by administering an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/045,576 Active 2032-12-19 US10227405B2 (en) 2011-11-16 2016-02-17 Methods of modulating the activity of adrenomedullin in a subject in need of regulation of fluid balance by administering an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment

Country Status (15)

Country Link
US (2) US20140314775A1 (pt)
EP (1) EP2780371B1 (pt)
JP (2) JP6193872B2 (pt)
AU (1) AU2012338734B2 (pt)
DK (1) DK2780371T3 (pt)
ES (1) ES2707878T3 (pt)
HR (1) HRP20190122T1 (pt)
HU (1) HUE042477T2 (pt)
LT (1) LT2780371T (pt)
PL (1) PL2780371T3 (pt)
PT (1) PT2780371T (pt)
RS (1) RS58340B1 (pt)
SG (2) SG11201402354YA (pt)
WO (1) WO2013072514A1 (pt)
ZA (1) ZA201403550B (pt)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020102422A1 (en) * 2018-11-14 2020-05-22 Arch Oncology, Inc. THERAPEUTIC SIRPα ANTIBODIES
WO2024017331A1 (zh) * 2022-07-20 2024-01-25 上海济煜医药科技有限公司 一种抗肾上腺髓质素非中和抗体、其制备方法及应用

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180348235A1 (en) * 2015-11-27 2018-12-06 B.R.A.H.M.S Gmbh MR-proADM as marker for the extracellular volume status of a subject
EP3339324A1 (en) 2016-12-22 2018-06-27 sphingotec GmbH Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
BR112019011713A2 (pt) * 2016-12-16 2019-10-15 Adrenomed Ag anticorpo anti-adrenomedulina (adm) ou fragmento de anticorpo anti-adm ou arcabouço anti-adm não ig para uso na intervenção e terapia da congestão em um paciente com necessidade da mesma
BR112020005682A2 (pt) * 2017-10-18 2020-10-20 Adrenomed Ag monitoração de terapia sob tratamento com um aglutinante de antiadrenomedulina (adm)
EP3871689A1 (en) 2020-02-26 2021-09-01 sphingotec GmbH Anti-adm-antibodies binding to the free n-terminus for accelerated transition of adm-gly to bio-adm in patients with adm-gly/ bio-adm ratio above a threshold and combination with vitamin c
EP4111204A1 (en) 2020-02-27 2023-01-04 4TEEN4 Pharmaceuticals GmbH Dpp3 for therapy guidance, monitoring and stratification of nt-adm antibodies in patients with shock
CA3169447A1 (en) 2020-02-27 2021-09-02 Andreas Bergmann Anti-adrenomedullin (adm) binder for use in therapy of patients in shock
JP2023515042A (ja) 2020-02-27 2023-04-12 アドレノメト アクチェンゲゼルシャフト ショックの治療若しくは予防において使用するための抗アドレノメデュリン(ADM)抗体若しくは抗ADM抗体フラグメント又は抗ADM非Ig足場
US20210285949A1 (en) 2020-03-16 2021-09-16 Sphingotec Gmbh Pro-adrenomedullin or fragment thereof in patients infected with corona virus and treatments with binder against adrenomedullin
WO2023175035A1 (en) 2022-03-15 2023-09-21 Adrenomed Ag Stable aqueous formulation of an anti-adrenomedullin (adm) antibody or anti-adm antibody fragment
WO2024023369A1 (en) 2022-07-29 2024-02-01 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock
WO2024023368A1 (en) 2022-07-29 2024-02-01 4TEEN4 Pharmaceuticals GmbH Prediction of an increase of dpp3 in a patient with septic shock

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7659081B2 (en) * 1999-09-10 2010-02-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Determination of AM-binding proteins and the association of adrenomedullin (AM) therewith

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
AU634716B2 (en) 1988-08-01 1993-03-04 Ciba Corning Diagnostics Corp. Method for detection of an analyte using acridinium esters and liposomes
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
SG48759A1 (en) 1990-01-12 2002-07-23 Abgenix Inc Generation of xenogenic antibodies
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
CA2109602C (en) 1990-07-10 2002-10-01 Gregory P. Winter Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
AU3328493A (en) 1991-12-17 1993-07-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
JP2774769B2 (ja) 1993-04-26 1998-07-09 賢治 寒川 アドレノメデュリン
JPH11512087A (ja) 1995-08-18 1999-10-19 アメリカ合衆国 ヒトの病理学及び生理学における、アドレノメジュリン(am)及び遺伝子関連産物(pamp)の機能的役割
CA2242308A1 (en) 1997-12-08 1999-06-08 Smithkline Beecham Laboratoires Pharmaceutiques Novel compounds
DE19847690A1 (de) 1998-10-15 2000-04-20 Brahms Diagnostica Gmbh Verfahren und Substanzen für die Diagnose und Therapie von Sepsis und sepsisähnlichen systemischen Infektionen
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
ATE439588T1 (de) 2001-05-04 2009-08-15 Biosite Inc Diagnostische marker für akute herzerkrankungen und verfahren des gebrauchs
MXPA04001804A (es) 2001-08-30 2005-03-07 Biorexis Pharmaceutical Corp Proteinas de fusion de transferrina modificada.
US6864237B2 (en) 2002-05-17 2005-03-08 Ping Wang Treatment of shock using adrenomedullin and adrenomedullin binding protein-1
ATE528014T1 (de) 2002-06-07 2011-10-15 Dyax Corp Polypeptid mit modifizierten kunitz domains
DE10316583A1 (de) 2003-04-10 2004-10-28 B.R.A.H.M.S Aktiengesellschaft Bestimmung eines midregionalen Proadrenomedullin-Teilpeptids in biologischen Flüssigkeiten zu diagnostischen Zwecken, sowie Immunoassays für die Durchführung einer solchen Bestimmung
EP1620734A1 (en) 2003-04-25 2006-02-01 Genova Ltd. Secreted polypeptide species reduced cardiovascular disorders
US6884781B2 (en) 2003-05-16 2005-04-26 Ping Wang Treatment of shock using adrenomedullin binding protein-1
US20050164301A1 (en) 2003-10-24 2005-07-28 Avidia Research Institute LDL receptor class A and EGF domain monomers and multimers
US20100028995A1 (en) 2004-02-23 2010-02-04 Anaphore, Inc. Tetranectin Trimerizing Polypeptides
EP1800131A2 (en) 2004-09-09 2007-06-27 Bayer HealthCare AG Diagnostics and therapeutics for diseases associated with adrenomedullin receptor (amdr)
EP1793847A2 (en) 2004-09-21 2007-06-13 NascaCell IP GmbH Use of microproteins as tryptase inhibitors
WO2007062676A1 (en) 2005-12-01 2007-06-07 B.R.A.H.M.S. Aktiengesellschaft Methods for the diagnosis and treatment of critically ill patients with endothelin, endothelin agonists and adrenomedullin antagonists
US7825217B2 (en) 2006-09-15 2010-11-02 University Of Kansas Medical Center Polypeptides for bone mineralization
KR101686247B1 (ko) * 2007-10-31 2016-12-14 메디뮨 엘엘씨 단백질 스캐폴드
DK2231860T3 (da) 2007-12-19 2011-12-05 Affibody Ab Polypeptid afledt protein A og i stand til at binde PDGF
CA2742241C (en) 2008-11-03 2019-12-10 Molecular Partners Ag Binding proteins inhibiting the vegf-a receptor interaction
CA2772162C (en) 2009-08-27 2018-05-22 Covagen Ag Anti-il-17a fynomers and medical uses thereof
US8748351B2 (en) 2009-12-14 2014-06-10 Scil Proteins Gmbh Method for identifying hetero-multimeric modified ubiquitin proteins with binding capability to ligands
DE102010040035A1 (de) 2010-03-04 2011-09-08 Robert Bosch Gmbh Verbesserungen der Rückwärts-Analyse zur Bestimmung von Fehlermaskierungsfaktoren
ES2729652T3 (es) 2010-06-08 2019-11-05 Pieris Pharmaceuticals Gmbh Muteína de lipocalina lacrimal unidas a IL-4R alpha
FR2964103B1 (fr) 2010-08-30 2018-11-23 Universite D'aix-Marseille Anticorps se liant a l'adrenomedulline et aux recepteurs de l'adrenomedulline et leurs utilisations comme medicament
PL2780717T3 (pl) 2011-11-16 2017-06-30 Sphingotec Gmbh Oznaczenia adrenomeduliny i sposoby do oznaczania dojrzałej adrenomeduliny

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7659081B2 (en) * 1999-09-10 2010-02-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Determination of AM-binding proteins and the association of adrenomedullin (AM) therewith

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020102422A1 (en) * 2018-11-14 2020-05-22 Arch Oncology, Inc. THERAPEUTIC SIRPα ANTIBODIES
US11202828B2 (en) 2018-11-14 2021-12-21 Arch Oncology, Inc. Therapeutic SIRP-α antibodies
WO2024017331A1 (zh) * 2022-07-20 2024-01-25 上海济煜医药科技有限公司 一种抗肾上腺髓质素非中和抗体、其制备方法及应用

Also Published As

Publication number Publication date
EP2780371A1 (en) 2014-09-24
DK2780371T3 (en) 2019-02-25
SG10201802054TA (en) 2018-05-30
ES2707878T3 (es) 2019-04-05
JP6193872B2 (ja) 2017-09-06
PL2780371T3 (pl) 2019-06-28
LT2780371T (lt) 2019-05-27
WO2013072514A1 (en) 2013-05-23
AU2012338734B2 (en) 2017-08-24
US20160159900A1 (en) 2016-06-09
JP2017155050A (ja) 2017-09-07
PT2780371T (pt) 2019-01-30
AU2012338734A1 (en) 2014-05-29
SG11201402354YA (en) 2014-06-27
RS58340B1 (sr) 2019-03-29
JP2014533672A (ja) 2014-12-15
EP2780371B1 (en) 2018-10-24
HUE042477T2 (hu) 2019-07-29
HRP20190122T1 (hr) 2019-03-08
US10227405B2 (en) 2019-03-12
NZ624877A (en) 2016-07-29
ZA201403550B (en) 2021-05-26

Similar Documents

Publication Publication Date Title
US11673949B2 (en) Method of modulating the adrenomedullin (ADM) activity of a patient by administering to the patient an anti-ADM antibody or fragment thereof that specifically binds to mature human ADM
US10227405B2 (en) Methods of modulating the activity of adrenomedullin in a subject in need of regulation of fluid balance by administering an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment
US9140696B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-IG scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
EP2594588B1 (en) Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or an anti-ADM non-Ig protein scaffold for use in therapy
US9402900B2 (en) Methods of modulating adrenomedullin by administering an anti-adrenomedullin (ADM) antibody
EP2594587B1 (en) Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig protein scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
CA2856150A1 (en) Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease

Legal Events

Date Code Title Description
AS Assignment

Owner name: ADRENOMED AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BERGMANN, ANDREAS;REEL/FRAME:033095/0871

Effective date: 20140606

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION