US20040106155A1 - Method for kidney disease treatment by drug intervention - Google Patents

Method for kidney disease treatment by drug intervention Download PDF

Info

Publication number
US20040106155A1
US20040106155A1 US10/721,351 US72135103A US2004106155A1 US 20040106155 A1 US20040106155 A1 US 20040106155A1 US 72135103 A US72135103 A US 72135103A US 2004106155 A1 US2004106155 A1 US 2004106155A1
Authority
US
United States
Prior art keywords
disease
albumin
protein
sample
renal
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
US10/721,351
Inventor
Wayne Comper
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.)
Monash University
Original Assignee
Monash University
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
Priority claimed from AUPP7843A external-priority patent/AUPP784398A0/en
Application filed by Monash University filed Critical Monash University
Priority to US10/721,351 priority Critical patent/US20040106155A1/en
Assigned to NORTHSHORE ASSET MANAGEMENT, LLC reassignment NORTHSHORE ASSET MANAGEMENT, LLC SECURITY AGREEMENT Assignors: AUSAM BIOTECHNOLOGIES, INC.
Publication of US20040106155A1 publication Critical patent/US20040106155A1/en
Assigned to TRIDENT GROWTH FUND, LP reassignment TRIDENT GROWTH FUND, LP SECURITY AGREEMENT Assignors: AUSAM BIOTECHNOLOGIES, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/811Test for named disease, body condition or organ function

Definitions

  • the present invention relates to methods of detecting an early stage of renal disease and/or renal complications of a disease.
  • the invention also relates to preventing and treating the disease.
  • Specific proteinuria and in particular, albuminuria (micro- and macro-), is a marker of disease including renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, diabetic nephropathy, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, Pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycystic kidney disease, autosomal rece
  • Kidney disease may result from bacterial infection, allergies, congenital defects, stones, antibiotics, immunosuppressives, antineoplastics, nonsteroidal anti-inflammatory drugs, analgesics, heavy metals, tumors, chemicals.
  • proteins including albumin
  • proteins are normally excreted as a mixture of native protein and fragments that are specifically produced during renal passage. Osicka T. M. et al., Nephrology 2:199-212 (1996). Proteins are heavily degraded during renal passage by post-glomerular (basement membrane) cells, which may include tubular cells. Lysosomes in renal tubular cells may be responsible for the breakdown of proteins excreted during renal passage (see, FIG. 1). The breakdown products are excreted into the tubular lumen. In normal individuals, most of the albumin in the urine is fragmented.
  • the invention provides for the use of a lysosome-activating compound in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome.
  • a lysosome-activating compound in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome.
  • lysosome activity or intracellular processes directing substrates to lysosomes is reduced as in kidney disease, more of the high molecular weight, and substantially full length filtered protein including albumin appears in the urine.
  • conventional drugs, the ACE inhibitor ramipril and the anti-glycation agent aminoguanidine are able to prevent changes in the renal lysosomal processing of protein.
  • the invention is directed to a method for treating a person suffering from a disease in which a diseased state is indicated by specific proteinuria, by administering a therapeutically effective amount of the treatment agent, obtained according to the above method, to a person in need thereof.
  • the treatment agent is a lysosome-activating compound.
  • a method of preventing or treating kidney disease including administering an effective amount of lysosome-activating compound to a subject in need thereof.
  • composition comprising a lysosome-activating compound and a carrier.
  • a method of screening a multiplicity of compounds to identify a compound capable of activating or processes that direct substrates to the lysosome or products away from the lysosome including the steps of:
  • the disease sought to be treated includes nephropathy, diabetes insipidus, diabetes type I, diabetes II, renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, Pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycys
  • the invention can be used in conjunction with a method of diagnosing early stage of renal disease and/or renal complications of a disease, comprising:
  • the invention can be used for determining a treatment agent (such as a lysosome-activating compound) for renal disease and/or renal complications of a disease, comprising:
  • FIG. 1 illustrates the progress of filtered intact albumin into tubular cells and breakdown of albumin to provide excreted albumin fragments.
  • FIG. 2 illustrate a representative profile of( 3 H) HSA in (a) urine and (b) plasma collected from normal, healthy volunteers by size exclusion chromatography.
  • Urine contains mostly fragmented albumin.
  • plasma contains mostly intact albumin.
  • FIG. 3 illustrates urine from normal, healthy volunteer showing a fragmented albumin peak, but no intact albumin peak from size exclusion chromatography.
  • FIG. 4 illustrates urine from a diabetic patient showing both intact and fragmented albumin peaks from size exclusion chromatography.
  • FIG. 5 illustrates a HPLC profile of albumin alone.
  • FIG. 6 illustrates the HPLC profile of plasma from normal, healthy volunteer showing albumin peaks.
  • FIG. 7 shows the HPLC profile of urine from normal, healthy volunteer with fragmented products of albumin but no intact albumin peak.
  • FIG. 8 shows the HPLC profile of a urine sample from a normoalbuminuric diabetic patient showing albumin breakdown products and a small-modified albumin peak at approximately 39-44 minutes retention time.
  • FIG. 9 shows the HPLC profile of urine from a normoalbuminuric diabetic patient showing signs of kidney failure and the presence of the characteristic spiked albumin peak at approximately 39-44 minutes retention time.
  • FIG. 10 illustrates a HPLC profile of a normoalbuminuric diabetic patient showing signs of kidney failure and the presence of the characteristic spiked modified albumin peak at approximately 39-44 minutes retention time.
  • FIG. 11 illustrates a HPLC of a macroalbuminuric diabetic patient showing high levels of the normal albumin as well as the characteristic spiked appearance at approximately 39-44 minutes retention time.
  • FIG. 12 illustrates a longitudinal study of a patient in which the modified protein was detected at a time prior to onset of diabetic nephropathy, indicating predisposition to diabetic nephropathy, and the delay in treatment caused by relying on conventional RIA methods.
  • FIG. 13 illustrates a longitudinal study of a patient in which the modified protein was detected at a time prior to onset of diabetic nephropathy, indicating predisposition to diabetic nephropathy, and the delay in treatment caused by relying on conventional RIA methods.
  • FIG. 14 illustrates a longitudinal study of a patient in which the modified protein was detected at a time prior to onset of diabetic nephropathy, indicating predisposition to diabetic nephropathy, and the delay in treatment caused by relying on conventional RIA methods.
  • FIG. 15 shows the HPLC chromatogram used as a criterion of purity of the modified albumin of Example 4.
  • FIG. 16 illustrates ( 3 H)HSA in urine collected from a microalbuminuric type I diabetic patient by size exclusion chromatography.
  • Urine contains mostly fragmented albumin.
  • Plasma contains only intact albumin.
  • Conventional RIA human albumin ordinate measures only intact albumin and not fragments.
  • FIG. 17 illustrates the size exclusion profiles of ( 3 H)HSA in urine from four different patients with varying albumin excretion rate (AER) as measured by conventional RIA.
  • AER albumin excretion rate
  • proteins including major plasma proteins such as albumin and immunoglobulin
  • tubular cells lie beyond the kidney filter and come in direct contact with the primary filtrate.
  • proteins are internalized by the tubular cells, they are directed towards the lysosomes, where they are partially degraded to various size fragments, and then regurgitated to outside the cell. These regurgitated fragments, of which there may be at least 60 different fragments generated from any one particular type of protein, are then excreted into the urine.
  • the drugs may also be useful in other renal diseases where lysosomal activities are affected, or in diabetes without renal complications in situations where lysosomal activity is turned off in non-renal tissues.
  • Such drugs include antiproliferative drugs, such as anticancer drugs or antibodies to neutralize TGF-beta.
  • the invention is also directed to a method of determining a treatment agent for renal disease and/or renal complications of a disease, comprising: (a) administering to a person an agent that is suspected of being able to treat the disease; (b) obtaining a urine sample from the person; and (c) assaying for the protein in the sample, wherein decreasing amount of the protein over time indicates that the agent is a treatment agent for the disease.
  • the treatment agent may be a lysosome-activating agent that may act directly or indirectly to activate lysosome, and thereby cause the lysosome to digest post-glomerular filtered proteins, which is a sign of a healthy kidney.
  • PKC protein kinase C
  • a lysosome-activating compound for use in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome.
  • composition comprising a lysosome-activating compound and a carrier.
  • a method of preventing or treating kidney disease including administering an effective amount of a lysosome-activating compound to a subject.
  • a method of screening a multiplicity of compounds to identify a compound capable of activating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome said method including the steps of: (a) exposing said compound to a lysosome and assaying said compound for the ability to activate a lysosome wherein said lysosome when activated has a changed activity; (b) assaying for the ability to restore a cellular process to substantially normal levels in kidney tissue, wherein said kidney tissue has a low lysosome activity; and/or (c) assaying for the ability to restore tissue turnover to substantially normal levels in kidney tissue, wherein said kidney tissue has low lysosome activity.
  • Lysosomes are associated with the breakdown of proteins, particularly albumin, in the kidney (see, FIG. 1). In cases of microalbuminuria, substantial amounts of albumin escape lysosomal breakdown possibly due to a deactivated lysosome. Restoration of lysosomal breakdown restores the balance in the kidney of cellular processes and tissue turnover.
  • a lysosome-activating compound is a compound that acts directly or indirectly on the lysosome. By acting indirectly, the compound may act on a component that influences the activity of the lysosome. Nevertheless, the outcome results in an activation of the lysosome, thereby providing enhanced protein breakdown.
  • composition comprising a lysosome-activating compound and a carrier.
  • the composition may be a physiologically acceptable or pharmaceutically acceptable composition. However, it will be a composition that allows for stable storage of the lysosome-activating compound. Where the composition is a pharmaceutically acceptable composition, it may be suitable for use in a method of preventing or treating kidney disease.
  • a method of preventing or treating kidney disease including administering an effective amount of a lysosome-activating compound to a subject.
  • the lysosome-activating compound may act by reactivating the lysosome so that cellular processes and tissue turnover are restored fully or in part, thereby resulting in the kidney being restored partially or fully.
  • administering a lysosome-activating compound to an animal having kidney disease may restore lysosome activity fully or in part.
  • Methods of administering may be oral or parenteral.
  • Oral may include administering with tablets, capsules, powders, syrups, etc.
  • Parenteral administration may include intravenous, intramuscular, subcutaneous or intraperitoneal routes. Dosages for administration of the compounds of the invention may be calculated by those of skill in the art (see, Goodman & Gilman, The Pharmacological Basis of Therapeutics , 8th Ed. (Pergamon Press, NY, 1990); and The Merck Index , 11th Ed. (Merck and Co., Inc., Rahway, N.J. 1989); both incorporated herein by reference).
  • the changed activity of the lysosome is preferably a change that enhances the activity of the lysosome so that albumin breakdown is improved.
  • the ability to not only activate lysosome but also improve cellular processes and/or tissue turnover is a characteristic of the most desirable lysosome-activating compound.
  • “Fragmented protein or fragment albumin” includes post-glomerular breakdown products after chemical, enzymatic or physical breakdown that occurs during renal passage. These components have a reduced size and/or may have changed hydrophobicity.
  • “Intact albumin, modified albumin, or modified form of albumin” as used herein means a compound having similar size and structural characteristics to native albumin, wherein the amino acid sequence is substantially the same as the native albumin. It is preferably a filtered intact protein. It elutes at or near the same position as native albumin on high-pressure liquid chromatography (HPLC) (FIG. 5).
  • HPLC high-pressure liquid chromatography
  • the structure has been modified biochemically either by minor enzyme mediated modification or addition to its basic structure and/or physically through a change in its three dimensional structure so that it escapes detection by conventionally used anti-albumin antibodies.
  • Biochemical modification may be made by enzymes such as endo- or exo- peptidases.
  • the 3D structure of albumin may have been altered in some way. Ligands may have bound to the albumin, or it may be any combination of these.
  • the modified albumin detected in the method of the invention is not detectable by current and conventional radioimmunoassays using
  • Conventional anti-albumin antibodies can be purchased from any purveyor of immunochemicals.
  • monoclonal antibody catalog numbers A6684 clone no. HSA-11
  • A2672 clone no. HSA-9
  • liquid whole serum, lyophilized fractionates, liquid IgG fraction, and the monoclonal antibodies in liquid ascites fluids form can be obtained from Sigma, St. Louis, Mo., as found in the Immunochemicals section at pages 1151-1152 in the 1994 Sigma—Biochemicals Organic Compounds for Research and Diagnostic Reagents catalog.
  • intact/modified albumin includes albumin that is substantially full-length, fragmented, chemically modified, or physically modified.
  • intact/modified albumin is meant to indicate albumin that is less than, equal to, or greater in molecular weight than the full-length albumin, and elutes at or near the native albumin position in a separation medium, such as chromatography, preferably HPLC, and most preferably hydrophobicity HPLC.
  • fragmented albumin is meant to refer to the fragment of albumin that is not detected by conventional anti-albumin antibody, and its presence is detected in diagnosing an early stage of renal disease and/or renal complications of a disease. The detection of the presence of intact/modified albumin is an indication of a predisposition to renal disease.
  • “Intact protein, modified protein or modified form of a protein” as used herein includes those forms of substantially full-length protein which are undetectable by conventional radioimmunoassay.
  • the protein includes, but is not limited to, albumins, globulins ( ⁇ -globulin( ⁇ 1 -globulin, ⁇ 2 -globulin), ⁇ -globulins, ⁇ -globulins), euglobulins, pseudoglobulin I and II, fibrinogen, ⁇ 1 , acid glycoprotein (orosomucoid), ⁇ 1 glycoprotein, ⁇ 1 lipoproteins, ceruloplasmin, ⁇ 2 19S glycoprotein, ⁇ 1 transferrin, ⁇ 1 lipoprotein, immunoglobulins A, E ,G, and M, protein hormones including growth hormone, insulin, parathyroid hormone and other proteins including horseradish peroxidase, lactate dehydrogenase, glucose oxidase, myoglobin, and lysozyme.
  • Kidney disease as used herein includes any malfunction of the kidney. Kidney disease may be identified by the presence of intact or modified albumin in the urine. Preferably, an early diagnosis of the kidney disease may be made by detecting the presence of modified protein in the urine, or an increase in the modified protein in the urine over time.
  • Low lysosome activity as used herein is compared against normal levels of lysosome activity and/or lysosome machinery that traffics protein to the lysosome in a normal individual. The activity is insufficient for the lysosome to fragment proteins so that intact protein is excreted at a greater amount than at normally low levels.
  • “Lysosome-activating compound” as used herein refers to a compound that is beneficial to reactivation of the lysosome.
  • the compound may work directly or indirectly on the lysosome resulting in activation of lysosomal function.
  • These compounds may be selected from the group including, but not limited to, anticancer compounds, antiproliferation compounds, paracetamol, insulin vitamin A (retinoic acid) or derivatives of retinol, or compounds, including antibodies, to neutralize TGF-beta.
  • Aminoguanidine and ramipril which have been previously thought to have different mechanism of action, seem to prevent similar diabetes-induced changes in lysosomal processing either through their similar effects on enzyme activity within the lysosome or through their similar effects on the trafficking of molecules to and from the lysosome.
  • Microalbuminuria is a condition where an individual excretes greater than 200 ⁇ g albumin/min in the urine as measured by conventional radioimmunoassay (RIA).
  • Microalbuminuria is a condition where an individual excretes at least 20 ⁇ g albumin/min in the urine as measured by conventional radioimmunoassay (RIA). RIA measures down to 15.6 ng/ml and is able to measure albumin in urine of normal subjects who have clearance of less than 6 ⁇ g/min. However, when albumin excretion exceeds 20 ⁇ g/min, treatment of the kidney disease is limited and full recovery is difficult from this point.
  • RIA radioimmunoassay
  • “Microalbuminuric” as used herein is a condition when albumin is detected in the urine at an excretion rate of at least 20 ⁇ g/min as measured by conventional RIA.
  • “native” and “unmodified” are used interchangeably to describe a protein that is naturally found in an organism, preferably a human, which has not been modified by the filtering process of the renal glomeruli.
  • Normal individual as used herein is an individual who does not have a disease in which intact protein found in urine is an indicator of the disease.
  • the disease is kidney disease.
  • Normal levels of lysosome activity are levels of lysosome activity found in undiseased kidney of a normal individual.
  • Normal albuminuric as used herein means a condition where albumin is excreted in the urine and is not detectable by RIA, or less than 20 ⁇ g/min (as measured by RIA) is excreted.
  • Propensity for a disease means that a disease may result in an individual as judged by a determination of the presence and excretion rate of a modified protein such as modified albumin.
  • Proteinuria as used herein is the existence of protein in the urine, usually in the form of albumin, a protein that is soluble in water and can be coagulated by heat.
  • specific proteinuria refers to the existence of a particular protein in the urine.
  • Radioimmunoassay as used herein is a method for detection and measurement of substances using radioactively labeled specific antibodies or antigens.
  • Reactivation of the lysosome includes an activation of lysosome activity preferably so that breakdown of proteins, particularly albumin, is increased compared with an inactivated state of the lysosome.
  • Restore means to restore in full or in part so that the component being restored has an improved function compared with its previous function.
  • the “sum of intact and intact modified protein” as used herein refers to the total amount of intact protein, and intact modified protein present in a biological sample.
  • Total protein refers to a particular filtered protein present in native, unmodified, modified or fragmented form that is excreted in urine. It includes protein that is not detected by conventional radioimmunoassay or conventional methods, which are currently available to detect the protein. Preferably the protein is albumin.
  • the diseases to be treated include, but are not limited to renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, diabetic nephropathy, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycystic kidney disease, autosomal recessive polycystic kidney
  • Kidney disease may result from bacterial infection, allergies, congenital defects, stones, antibiotics, immunosuppressives, antineoplastics, nonsteroidal antiinflammatory drugs, analgesics, heavy metals, tumors, chemicals.
  • a method for determining a propensity for or early diagnosis of renal disease and/or renal complications of a disease includes determining a change in the albumin content in a urine sample.
  • the disease may be a kidney disease, although not necessarily limited to a kidney disease.
  • albumin is used herein only as an example of a protein to be detected in urine.
  • albumin in a patient is analyzed by conventional RIA, it is expected that a normoalbuminuric patient or normal individual would have albumin in the urine in the range of 3-10 ⁇ g/min in young people and greater in older people.
  • normoalbuminuric patients also show levels of albumin in the urine if measured by HPLC. Applicant has found that these levels may be in the order of 5 ⁇ g/min.
  • the level of intact/modified albumin will increase to microalbuminuria levels in the order of 20 to 200 ⁇ g/min as determined by RIA.
  • a patient suspected of having diabetic kidney disease will not show signs of kidney degeneration until well after 10 to 15 years when albumin is detected by currently available methods such as RIA methods.
  • Urinary excretion rates of at least 20 ⁇ g/min may be detected by RIA when an individual enters a microalbuminuric state. Again, by observing the excretion of modified albumin, a change in the kidney and possibly onset of a kidney disease may be detected.
  • a normoalbuminuric subject, or normoalbuminuric diabetic patient may continue to have a low albumin excretion rate of less than 20 ⁇ g/min as determined by RIA, for many years.
  • the presence of albumin in the urine is a sign that functions of the kidney may be impaired. Once this level begins to change, treatment may be initiated.
  • albumin In a normal individual a small amount of albumin is detectable in the urine. Total filtered albumin appears mainly as fragmented albumin in urine. Some albumin may be detected in normoalbuminuric individuals. However, the excretion rate of albumin in urine in a normoalbuminuric individual may be as low as 5 ⁇ g/min. This level is generally detectable by RIA.
  • the modified protein of the invention can be detected by a variety of methods that are well-known in the art, including, but not limited to chromatography, electrophoresis and sedimentation, or a combination of these, which are described in Karger B L, Hancock W S (eds.) High Resolution Separation and Analysis of biological Macromolecules. Part A Fundamentals in Methods in Enzymology , Vol. 270, 1996, Academic Press, San Diego, Calif., USA; Karger B L, Hancock W S (eds.) High Resolution Separation and Analysis of biological Macromolecules. Part B Applications in Methods in Enzymology , Vol.
  • the electrophoresis method includes, but is not limited to, moving-boundary electrophoresis, zone electrophoresis, and isoelectric focusing.
  • the chromatography method includes, but is not limited to, partition chromatography, adsorption chromatography, paper chromatography, thin-layer chromatography, gas-liquid chromatography, gel chromatography, ion-exchange chromatography, affinity chromatography, and hydrophobic interaction chromatography.
  • the method is a sizing gel chromatography and hydrophobic interaction chromatography. More preferably, the method is hydrophobic interaction chromatography using a HPLC column.
  • the modified protein can also be detected by the use of specific albumin dyes. Such methods are described by Pegoraro et al., American Journal of Kidney Diseases 35(4): 739-744 (April 2000), the entire disclosure of which is hereby incorporated by reference.
  • the modified albumin, as well as the whole albumin is detectable by this dye method to provide the sum of modified albumin and whole or intact albumin.
  • This detection method may be used with or without an initial separation of the albumin components from urine.
  • Such dyes normally do not detect fragments ⁇ 10,000 in molecular weight, but will detect the modified albumin.
  • a dye such as Albumin Blue 580 is used.
  • Such dyes are naturally non-fluorescent, but fluoresce on binding to intact albumin as well as the modified albumin, but do not bind to globulins. Therefore, globulins do not interfere with the assay so that measurements can be made in unfractionated urine.
  • a normoalbuminuric diabetic patient has almost undetectable levels of modified or fragments of albumin when analyzed by conventional RIA. They appear to be normal. However, when the urine is tested by HPLC, the levels of modified albumin are much greater than found in a normal individual. This difference in albumin may be attributed to the inability of conventional RIA's to adequately detect all albumin (total albumin) in intact or modified forms. Thus, HPLC is preferred for generating a fragmentation profile.
  • a fragmentation profile on HPLC is characterized by a series of peaks representing a number of species of albumin as fragments or in intact or modified forms.
  • the method of determining a propensity for or early diagnosis of a kidney disease in a subject is determined before the subject becomes microalbuminuric.
  • Measuring albumin content in a sample by an HPLC method of the present invention may provide different results from its measurement by conventional RIA.
  • a low level of albumin is observed in normal individuals.
  • the level of modified albumin begins to be detected and its level increases, and progresses toward microalbuminuria then a patient can be determined to have a propensity for kidney disease.
  • the HPLC generated fragmentation profile is characterized by the absence of a peak in a region where full-length native albumin elutes. Instead, multiple fragmented albumin is detectable.
  • a pure protein product (unmodified) produces essentially a single peak.
  • albumin was observed to elute in the range of 39-44 minutes (FIG. 5).
  • a normal individual would provide a distinct fragmentation profile indicative of an absence of kidney disease or no propensity for a kidney disease.
  • an increasing amount of modified albumin first, and then native form later are detectable.
  • the fragmentation profile begins to change and more products in the region of full-length albumin manifests as additional spikes or an enlarged peak indicative of more intact/modified albumin in the urine.
  • the modified albumin may appear in a region where native albumin elutes but may be manifest as multiple peaks indicating the presence of multiple forms of modified albumin.
  • the propensity for kidney disease may be measured by determining the presence of or identifying at least one species of modified albumin. This may be determined or identified by the presence of a specific peak on a HPLC profile, preferably the peak is within the range of position that corresponds to the elution position of the native albumin.
  • a HPLC column for detecting modified albumin or unmodified albumin may be a hydrophobicity column, such as Zorbax 300 SB-CB (4.6 mm ⁇ 150 mm).
  • a 50 ⁇ l sample loop may be used.
  • Elution solvents suitable for HPLC in detecting albumin and its breakdown products may include standard elution solvents such as acetonitrile solvents.
  • a buffer of water/1% trifluoro acetic acid (TFA) followed by a buffer of 60% acetonitrile/0.09% TFA may be used.
  • TFA trifluoro acetic acid
  • a gradient of 0 to 100% of a 60% acetonitrile/0.09% TFA has been found to be suitable.
  • Suitable HPLC conditions for a hydrophobicity column may be as follows:
  • the wavelength used in HPLC may be approximately 214 nm.
  • Modified albumin may elute between 39-44 minutes (FIG. 5). Albumin fragments may elute much earlier, mainly at less than 20 minutes.
  • the method for determining the propensity for kidney disease is applicable to any individual.
  • Kidney disease may be caused by a number of factors including bacterial infection, allergic, congenital defects, stones, tumors, chemicals or from diabetes.
  • the method is applicable for determining a propensity for kidney disease in diabetic patients that may progress to a kidney disease.
  • the individual is a normoalbuminuric diabetic.
  • normal individuals may be monitored for propensity for the disease by determining increased levels of intact or modified albumin in the urine.
  • the method of the invention can be carried out using non-antibody separation procedures as described above.
  • antibody specific for modified protein may also be used to detect the presence of the modified protein.
  • the antibody to the modified protein may be obtained using the following method.
  • the procedure is described specifically for albumin by way of example only, and can be readily applied to antibody production against any other protein in the urine.
  • the method seeks to determine which modified albumin molecule is the most sensitive marker to identify diabetic patients, for example, who will progress to kidney complications.
  • the modified albumin is characterized by carrying out a quantitative separation of the modified albumin molecules, such as by preparative HPLC.
  • the modified proteins are analyzed for ligand binding, such as glycation.
  • amino acid sequence of the individual modified protein is determined, preferably by mass spectrometry using methods described in Karger B L, Hancock W S (eds.) High Resolution Separation and Analysis of biological Macromolecules. Part A Fundamentals in Methods in Enzymology , Vol. 270, 1996, Academic Press, San Diego, Calif., USA; or Karger B L, Hancock W S (eds.) High Resolution Separation and Analysis of biological Macromolecules. Part B Applications in Methods in Enzymology , Vol. 271, 1996, Academic Press, San Diego, Calif., USA, for example, which references are incorporated herein by reference in their entirety. In a preferred embodiment, there may be about 3 to 4 modified albumin species.
  • the method of generating antibody against the modified albumin seeks to develop a diagnostic immunoassay for the modified albumin that predicts those diabetic patients, for example, that progress to kidney complications. To accomplish this, sufficient quantities of modified albumin is prepared by HPLC. Antibodies are made by sequential injection of the modified albumin in an animal such as a rabbit, to generate good titer, and the antibodies are isolated using conventional techniques using methods described in Goding J W, Monoclonal Antibodies: Principles and Practice.
  • the obtained antibodies may be polyclonal antibodies or monoclonal antibodies.
  • At least one species of a modified albumin is isolated and identified for use in determining a propensity for kidney disease.
  • the isolated species may be used to generate antibodies for use in immunoassays.
  • the antibodies may be tagged with an enzymatic, radioactive, fluorescent or chemiluminescent label.
  • the detection method may include, but is not limited to radioimmunoassay, immunoradiometric assay, fluorescent immunoassay, enzyme linked immunoassay, and protein A immunoassay.
  • the assays may be carried out in the manner described in Goding J W, Monoclonal Antibodies: Principles and Practice. Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology .
  • kits for rapidly and accurately determining the presence or absence of modified protein such as modified albumin, in a sample quantitatively or non-quantitatively as desired.
  • modified protein such as modified albumin
  • Each component of the kit(s). may be individually packaged in its own suitable container.
  • the individual container may also be labeled in a manner, which identifies the contents.
  • the individually packaged components may be placed in a larger container capable of holding all desired components.
  • Associated with the kit may be instructions, which explain how to use the kit. These instructions may be written on or attached to the kit.
  • the invention is also directed to a method of determining a treatment agent for renal disease and/or renal complications of a disease, comprising:
  • the treatment agent may be a lysosome activating agent that may act directly or indirectly to activate lysosome, and thereby cause the lysosome to digest post-glomerular filtered proteins, which is a sign of a healthy kidney.
  • PKC protein kinase C
  • a lysosome-activating compound for use in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome.
  • composition comprising a lysosome-activating compound and a carrier.
  • a method of preventing or treating kidney disease including administering an effective amount of a lysosome-activating compound to a subject.
  • a method of screening a multiplicity of compounds to identify a compound capable of activating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome including the steps of:
  • Lysosomes may be associated with the breakdown of proteins, particularly albumin, in the kidney. In cases of microalbuminuria, substantial amounts of albumin escape lysosomal breakdown possibly due to a deactivated lysosome. Restoration of lysosomal breakdown may restore the balance in the kidney of cellular processes and tissue turnover.
  • a lysosome-activating compound may be a compound that acts directly or indirectly on the lysosome. By acting indirectly, the compound may act on a component, which influences the activity of the lysosome. Nevertheless, the outcome results in an activation of the lysosome, thereby providing enhanced protein breakdown.
  • composition comprising a lysosome-activating compound and a carrier.
  • the composition may be a physiologically acceptable or pharmaceutically acceptable composition. However, it will be a composition which allows for stable storage of the lysosome activating compound. Where the composition is a pharmaceutically acceptable composition, it may be suitable for use in a method of preventing or treating kidney disease.
  • the lysosome-activating compound may act by reactivating the lysosome so that cellular processes and tissue turnover are restored fully or in part, thereby resulting in the kidney being restored partially or fully.
  • administering a lysosome activating compound to an animal having kidney disease may restore lysosome activity fully or in part.
  • Methods of administering may be oral or parenteral.
  • Oral may include administering with tablets, capsules, powders, syrups, etc.
  • Parenteral administration may include intravenous, intramuscular, subcutaneous or intraperitoneal routes.
  • the changed activity of the lysosome is preferably a change which enhances the activity of the lysosome so that albumin breakdown is improved.
  • the ability to not only activate lysosome but also improve cellular processes and/or tissue turnover is a characteristic of the most desirable lysosome activating compound.
  • a method for preventing kidney disease in a subject including:
  • H[HSA] Human Serum Albumin
  • Tritium radioactivity was determined in 1 ml aqueous samples with 3 ml scintillant and measured on a Wallac 1410 liquid scintillation counter (Wallac Turku, Finland).
  • FIG. 2 illustrates the distribution of albumin in urine and in plasma.
  • Example 1 3 H[HSA] as used in Example 1 was injected into a normal, healthy volunteer and a diabetic patient. Samples of urine were collected and 3 H[HSA] was determined as in Example 1.
  • the normal, healthy volunteer (FIG. 3) shows the excretion of fragments of albumin on a size exclusion chromatography as performed in Example 1.
  • the diabetic patient (FIG. 4) shows the presence of substantially full-length and fragmented albumin on size exclusion chromatography.
  • excretion rates of albumin detectable by these methods were in the order of 5 ⁇ g/min (control) and 1457 ⁇ g/min (diabetic).
  • Urine samples were collected from normal, healthy volunteer, normoalbuminuric diabetic patients and from macroalbuminuric patients. Urine was collected midstream in 50 ml urine specimen containers. The urine was frozen until further use. Prior to HPLC analysis the urine was centrifuged at 5000 g.
  • a wavelength of 214 nm was used.
  • Urine from microalbuminuric patient which had an intact albumin concentration of 43.5 mg/L as determined by turbitimer (involving conventional immunochemical assay) was initially filtered through a 30 kDa membrane to separate the modified albumin from low molecular weight ( ⁇ 30,000) protein fragments in urine.
  • the material that was retained by the filter gave a yield of intact albumin of 27.4 mg/L as determined by turbitimer assay.
  • This retained material was then subjected to size exclusion chromotography on Sephadex G100.
  • the material collected was the peak fraction that coelutes with intact albumin. This material gave a yield of 15.2 ml/L of albumin as determined by the turbitimer method.
  • FIG. 5 illustrates a HPLC profile of albumin alone. Essentially a single peak which elutes at approximately 39-44 minutes retention time was obtained.
  • FIG. 6 illustrates a HPLC profile of plasma showing a distinct albumin peak at approximately 39-44 minutes as well as other peaks corresponding to other plasma proteins.
  • FIG. 7 illustrates a HPLC profile of a normal, healthy volunteer showing no albumin peak in the urine sample. This individual breaks down the albumin excreted into the urine possibly via an active lysosome. Substantial fragmented products were evident showing prominence of some species, particularly of a species at approximately less than 14.5 minutes retention time.
  • a further urine sample from a normoalbuminuric diabetic patient (with an albumin excretion rate of 4.37 ⁇ g/min) was analyzed, and the HPLC profile is illustrated in FIG. 10. Again, modified albumin was detected at approximately 39-44 minutes retention time showing multiple peaks. This patient again did register normal albumin by RIA. Whereas conventional test indicates the presence of ⁇ 6 mg/l of albumin in the urine sample, the method of the invention showed that the true albumin content in the urine sample was 491 mg/l. Treatment for the disease should have begun on this individual. It is clear that modified albumin assessment is necessary to identify these changes. This patient would be determined to have a propensity for kidney disease. As kidney disease progresses, the modified albumin peak will continue to increase.
  • the method of the invention results in early detection of a propensity for a renal disease as illustrated by the longitudinal studies in FIGS. 12 - 14 .
  • FIGS. 12 - 14 show situations in which the ACE inhibitor treatment for diabetes was begun later than it should have had the modified albumin detection method of the invention been used. Detecting modified protein using the method according to the invention is a more effective method for predicting the onset of a renal disease than using conventional RIA.
  • FIG. 17 shows that in renal disease in type I diabetic patients the severity of the disease as measured by albuminuria is associated with the increased presence of intact albumin in urine.
  • FIG. 17 illustrates the size exclusion profiles of( 3 H)HSA in urine from four different patients with varying albumin excretion rate (AER) as measured by conventional RIA.
  • AER albumin excretion rate

Abstract

A method is disclosed for diagnosing early stage of a disease in which an intact protein found in urine is an indicator of the disease, followed by early drug intervention to prevent and treat the disease are also disclosed. The drug treatment involves the use of a lysosome activating compound.

Description

    CLAIM OF PRIORITY
  • This application is a continuation-in-part of and claims priority to parent application Ser. No. ______, filed Jun. 22, 2001, which is a continuation-in-part of and claims priority to Ser. No. 09/415,217, filed Oct. 12, 1999.[0001]
  • FIELD OF THE INVENTION
  • The present invention relates to methods of detecting an early stage of renal disease and/or renal complications of a disease. The invention also relates to preventing and treating the disease. [0002]
  • BACKGROUND OF THE INVENTION
  • The appearance of excess protein such as albumin in the urine is indicative of kidney disease. Diabetic nephropathy is such a disease. By the time the excess albumin is detected, kidney disease has progressed, possibly to a stage where it is irreversible and treatment has little effect. Therefore it is an object of the invention to provide a test that is more sensitive than the currently known radioimmunoassay to detect such a disease as early as possible so that the disease can be either prevented or a treatment protocol commenced early on in the disease. [0003]
  • Specific proteinuria, and in particular, albuminuria (micro- and macro-), is a marker of disease including renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, diabetic nephropathy, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, Pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycystic kidney disease, autosomal recessive polycystic kidney disease, tuborous sclerosis), von Hippel-Lindau disease, familial thin-glomerular basement membrane disease, collagen III glomerulopathy, fibronectin glomerulopathy, Alport's syndrome, Fabry's disease, Nail-Patella Syndrome, congenital urologic anomalies), monoclonal gammopathies (multiple myeloma, amyloidosis and related disrders), febrile illness (familial Mediterannean fever, HIV infection -AIDS), inflammatory disease (systemic vasculitides (polyarteritis nodosa, Wegener's granulomatosis, polyarteritis, necrotizing and crescentic glomerulonephritis), polymyositis-dermatomyositis, pancreatitis, rheumatoid arthritis, systemic lupus erythematosus, gout), blood disorders (sickle cell disease, thrombotic thrombocytopenia purpura, hemolytic-uremic syndrome, acute corticol necrosis, renal thromboembolism), trauma and surgery (extensive injury, burns, abdominal and vascular surgery, induction of anesthesia), drugs (penicillamine, steroids) and drug abuse, malignant disease (epithelial (lung, breast), adenocarcinoma (renal), melanoma, lymphoreticular, multiple myeloma), circulatory disease (myocardial infarction, cardiac failure, peripheral vascular disease, hypertension, coronary heart disease, non-atherosclerotic cardiovascular disease, atherosclerotic cardiovascular disease), skin disease (psoriasis, systemic sclerosis), respiratory disease (COPD, obstructive sleep apnoea, hypoia at high altitude) and endocrine disease (acromegaly, diabetes mellitus, and diabetes insipidus). [0004]
  • Kidney disease may result from bacterial infection, allergies, congenital defects, stones, antibiotics, immunosuppressives, antineoplastics, nonsteroidal anti-inflammatory drugs, analgesics, heavy metals, tumors, chemicals. [0005]
  • The applicant has found that proteins, including albumin, are normally excreted as a mixture of native protein and fragments that are specifically produced during renal passage. Osicka T. M. et al., [0006] Nephrology 2:199-212 (1996). Proteins are heavily degraded during renal passage by post-glomerular (basement membrane) cells, which may include tubular cells. Lysosomes in renal tubular cells may be responsible for the breakdown of proteins excreted during renal passage (see, FIG. 1). The breakdown products are excreted into the tubular lumen. In normal individuals, most of the albumin in the urine is fragmented.
  • SUMMARY OF THE INVENTION
  • The invention provides for the use of a lysosome-activating compound in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome. When lysosome activity or intracellular processes directing substrates to lysosomes is reduced as in kidney disease, more of the high molecular weight, and substantially full length filtered protein including albumin appears in the urine. The applicant has found that conventional drugs, the ACE inhibitor ramipril and the anti-glycation agent aminoguanidine, are able to prevent changes in the renal lysosomal processing of protein. [0007]
  • The invention is directed to a method for treating a person suffering from a disease in which a diseased state is indicated by specific proteinuria, by administering a therapeutically effective amount of the treatment agent, obtained according to the above method, to a person in need thereof. Preferably, the treatment agent is a lysosome-activating compound. [0008]
  • In one aspect of the invention, there is provided a method of preventing or treating kidney disease, the method including administering an effective amount of lysosome-activating compound to a subject in need thereof. [0009]
  • In another aspect of the present invention, there is provided a composition comprising a lysosome-activating compound and a carrier. [0010]
  • In yet another aspect of the present invention, there is provided a method of screening a multiplicity of compounds to identify a compound capable of activating or processes that direct substrates to the lysosome or products away from the lysosome, said method including the steps of: [0011]
  • (a) exposing said compound to a lysosome and assaying said compound for the ability to activate a lysosome wherein said lysosome when activated has a changed activity; [0012]
  • (b) assaying for the ability to restore a cellular process to substantially normal levels in kidney tissue, wherein said kidney has a low lysosome activity; and/or [0013]
  • (c) assaying for the ability to restore tissue turnover to substantially normal levels in kidney tissue, wherein said kidney tissue has low lysosome activity. [0014]
  • Although not limited to any particular disease, according to the method of the invention, the disease sought to be treated includes nephropathy, diabetes insipidus, diabetes type I, diabetes II, renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, Pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycystic kidney disease, autosomal recessive polycystic kidney disease, tuborous sclerosis), von Hippel-Lindau disease, familial thin-glomerular basement membrane disease, collagen III glomerulopathy, fibronectin glomerulopathy, Alport's syndrome, Fabry's disease, Nail-Patella Syndrome, congenital urologic anomalies), monoclonal gammopathies (multiple myeloma, amyloidosis and related disorders), febrile illness (familial Mediterranean fever, HIV infection—AIDS), inflamrnatory disease (systemic vasculitides (polyarteritis nodosa, Wegener's granulomatosis, polyarteritis, necrotizing and crescentic glomerulonephritis), polymyositis-dermatomyositis, pancreatitis, rheumatoid arthritis, systemic lupus erythematosus, gout), blood disorders (sickle cell disease, thrombotic thrombocytopenia purpura, hemolytic-uremic syndrome, acute cortical necrosis, renal thromboembolism), trauma and surgery (extensive injury, burns, abdominal and vascular surgery, induction of anesthesia), drugs (penicillamine, steroids) and drug abuse, malignant disease (epithelial (lung, breast), adenocarcinoma (renal), melanoma, lymphoreticular, multiple myeloma), circulatory disease (myocardial infarction, cardiac failure, peripheral vascular disease, hypertension, coronary heart disease, non-atherosclerotic cardiovascular disease, atherosclerotic cardiovascular disease), skin disease (psoriasis, systemic sclerosis), respiratory disease (COPD, obstructive sleep apnoea, hypoia at high altitude) and endocrine disease (acromegaly, diabetes mellitus, diabetes insipidus). [0015]
  • The invention can be used in conjunction with a method of diagnosing early stage of renal disease and/or renal complications of a disease, comprising: [0016]
  • (a) separating all of the proteins in a urine sample; and [0017]
  • (b) detecting a modified form of a protein in the sample, wherein detection of the modified protein is indicative of an early stage of the renal disease and/or renal complications of a disease. [0018]
  • The invention can be used for determining a treatment agent (such as a lysosome-activating compound) for renal disease and/or renal complications of a disease, comprising: [0019]
  • (a) administering to a person in need thereof an agent that is suspected of being able to treat the disease; [0020]
  • (b) obtaining a urine sample from the person; and [0021]
  • (c) assaying for a modified form of the protein in the sample, wherein either presence or lack of presence of the modified form of the protein in the urine or decreasing amount of the modified form of the protein over time indicates that the agent is a treatment agent for the renal disease and/or renal complications of a disease. [0022]
  • These and other objects of the invention will be more fully understood from the following description of the invention, the referenced drawings attached hereto and the claims appended hereto.[0023]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates the progress of filtered intact albumin into tubular cells and breakdown of albumin to provide excreted albumin fragments. [0024]
  • FIG. 2 ([0025] 2 a and 2 b) illustrate a representative profile of(3H) HSA in (a) urine and (b) plasma collected from normal, healthy volunteers by size exclusion chromatography. Urine contains mostly fragmented albumin. And plasma contains mostly intact albumin.
  • FIG. 3 illustrates urine from normal, healthy volunteer showing a fragmented albumin peak, but no intact albumin peak from size exclusion chromatography. [0026]
  • FIG. 4 illustrates urine from a diabetic patient showing both intact and fragmented albumin peaks from size exclusion chromatography. [0027]
  • FIG. 5 illustrates a HPLC profile of albumin alone. [0028]
  • FIG. 6 illustrates the HPLC profile of plasma from normal, healthy volunteer showing albumin peaks. [0029]
  • FIG. 7 shows the HPLC profile of urine from normal, healthy volunteer with fragmented products of albumin but no intact albumin peak. [0030]
  • FIG. 8 shows the HPLC profile of a urine sample from a normoalbuminuric diabetic patient showing albumin breakdown products and a small-modified albumin peak at approximately 39-44 minutes retention time. [0031]
  • FIG. 9 shows the HPLC profile of urine from a normoalbuminuric diabetic patient showing signs of kidney failure and the presence of the characteristic spiked albumin peak at approximately 39-44 minutes retention time. [0032]
  • FIG. 10 illustrates a HPLC profile of a normoalbuminuric diabetic patient showing signs of kidney failure and the presence of the characteristic spiked modified albumin peak at approximately 39-44 minutes retention time. [0033]
  • FIG. 11 illustrates a HPLC of a macroalbuminuric diabetic patient showing high levels of the normal albumin as well as the characteristic spiked appearance at approximately 39-44 minutes retention time. [0034]
  • FIG. 12 illustrates a longitudinal study of a patient in which the modified protein was detected at a time prior to onset of diabetic nephropathy, indicating predisposition to diabetic nephropathy, and the delay in treatment caused by relying on conventional RIA methods. [0035]
  • FIG. 13 illustrates a longitudinal study of a patient in which the modified protein was detected at a time prior to onset of diabetic nephropathy, indicating predisposition to diabetic nephropathy, and the delay in treatment caused by relying on conventional RIA methods. [0036]
  • FIG. 14 illustrates a longitudinal study of a patient in which the modified protein was detected at a time prior to onset of diabetic nephropathy, indicating predisposition to diabetic nephropathy, and the delay in treatment caused by relying on conventional RIA methods. [0037]
  • FIG. 15 shows the HPLC chromatogram used as a criterion of purity of the modified albumin of Example 4. [0038]
  • FIG. 16 illustrates ([0039] 3H)HSA in urine collected from a microalbuminuric type I diabetic patient by size exclusion chromatography. Urine contains mostly fragmented albumin. Plasma contains only intact albumin. Conventional RIA (human albumin ordinate) measures only intact albumin and not fragments.
  • FIG. 17 illustrates the size exclusion profiles of ([0040] 3H)HSA in urine from four different patients with varying albumin excretion rate (AER) as measured by conventional RIA. As intact albuminuria increases the amount of intact albumin in the urine increases and the relative amount of fragments decreases. The total excretion represents the excretion of intact albumin plus albumin derived fragments.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The applicant has discovered that when proteins, including major plasma proteins such as albumin and immunoglobulin, are filtered by the kidney they are subsequently degraded by cells in the kidney prior to the material being excreted. It is likely that filtered proteins are taken up by tubular cells. Tubular cells lie beyond the kidney filter and come in direct contact with the primary filtrate. When proteins are internalized by the tubular cells, they are directed towards the lysosomes, where they are partially degraded to various size fragments, and then regurgitated to outside the cell. These regurgitated fragments, of which there may be at least 60 different fragments generated from any one particular type of protein, are then excreted into the urine. [0041]
  • The applicant has discovered that in renal disease fragmentation of proteins is inhibited. This means that substantially full-length filtered proteins will be excreted in a person suffering from renal disease. This transition from fragmentation to inhibition of fragmentation of excreted proteins is a basis for the development of new drugs and diagnostic assays. For example, initial changes that occur with the onset of renal complications in diabetes are associated with a change in the fragmentation profile of excreted albumin. This leads to an apparent microalbuminuria, which is synonymous with the development of diabetic nephropathy. It is likely that this is due to an inhibition in the lysosomal activity of tubular cells in diabetes. Thus, drugs can be formulated to turn on lysosomal activity in diabetes where renal complications are occurring. The drugs may also be useful in other renal diseases where lysosomal activities are affected, or in diabetes without renal complications in situations where lysosomal activity is turned off in non-renal tissues. Such drugs include antiproliferative drugs, such as anticancer drugs or antibodies to neutralize TGF-beta. [0042]
  • The invention is also directed to a method of determining a treatment agent for renal disease and/or renal complications of a disease, comprising: (a) administering to a person an agent that is suspected of being able to treat the disease; (b) obtaining a urine sample from the person; and (c) assaying for the protein in the sample, wherein decreasing amount of the protein over time indicates that the agent is a treatment agent for the disease. The treatment agent may be a lysosome-activating agent that may act directly or indirectly to activate lysosome, and thereby cause the lysosome to digest post-glomerular filtered proteins, which is a sign of a healthy kidney. [0043]
  • The process of trafficking of proteins to the lysosomes plays a role in the mechanism of albuminuria in diabetes. An intracellular molecule that is involved in trafficking is protein kinase C (PKC). It is contemplated that a drug or agent can be formulated that activates lysosomal trafficking or inhibits PKC. [0044]
  • Accordingly, in one aspect of the present invention, there is provided a lysosome-activating compound for use in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome. [0045]
  • In another aspect of the present invention, there is provided a composition comprising a lysosome-activating compound and a carrier. [0046]
  • In yet another aspect of the invention there is provided a method of preventing or treating kidney disease, said method including administering an effective amount of a lysosome-activating compound to a subject. [0047]
  • In yet another aspect of the present invention, there is provided a method of screening a multiplicity of compounds to identify a compound capable of activating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome, said method including the steps of: (a) exposing said compound to a lysosome and assaying said compound for the ability to activate a lysosome wherein said lysosome when activated has a changed activity; (b) assaying for the ability to restore a cellular process to substantially normal levels in kidney tissue, wherein said kidney tissue has a low lysosome activity; and/or (c) assaying for the ability to restore tissue turnover to substantially normal levels in kidney tissue, wherein said kidney tissue has low lysosome activity. [0048]
  • Lysosomes are associated with the breakdown of proteins, particularly albumin, in the kidney (see, FIG. 1). In cases of microalbuminuria, substantial amounts of albumin escape lysosomal breakdown possibly due to a deactivated lysosome. Restoration of lysosomal breakdown restores the balance in the kidney of cellular processes and tissue turnover. [0049]
  • A lysosome-activating compound is a compound that acts directly or indirectly on the lysosome. By acting indirectly, the compound may act on a component that influences the activity of the lysosome. Nevertheless, the outcome results in an activation of the lysosome, thereby providing enhanced protein breakdown. [0050]
  • In another aspect of the present invention, there is provided a composition comprising a lysosome-activating compound and a carrier. [0051]
  • The composition may be a physiologically acceptable or pharmaceutically acceptable composition. However, it will be a composition that allows for stable storage of the lysosome-activating compound. Where the composition is a pharmaceutically acceptable composition, it may be suitable for use in a method of preventing or treating kidney disease. [0052]
  • In yet another aspect of the invention there is provided a method of preventing or treating kidney disease, said method including administering an effective amount of a lysosome-activating compound to a subject. [0053]
  • As described above, the lysosome-activating compound may act by reactivating the lysosome so that cellular processes and tissue turnover are restored fully or in part, thereby resulting in the kidney being restored partially or fully. In any case, administering a lysosome-activating compound to an animal having kidney disease may restore lysosome activity fully or in part. [0054]
  • Methods of administering may be oral or parenteral. Oral may include administering with tablets, capsules, powders, syrups, etc. Parenteral administration may include intravenous, intramuscular, subcutaneous or intraperitoneal routes. Dosages for administration of the compounds of the invention may be calculated by those of skill in the art (see, Goodman & Gilman, [0055] The Pharmacological Basis of Therapeutics, 8th Ed. (Pergamon Press, NY, 1990); and The Merck Index, 11th Ed. (Merck and Co., Inc., Rahway, N.J. 1989); both incorporated herein by reference). Preparation of aqueous solutions, liposomes, emulsion and suspensions are known to those of ordinary skill in the art (see, Remington's Pharmaceutical Sciences, 18th Ed. (Mack Publishing Co., Easton, Pa., 1990); incorporated herein by reference).
  • The changed activity of the lysosome is preferably a change that enhances the activity of the lysosome so that albumin breakdown is improved. The ability to not only activate lysosome but also improve cellular processes and/or tissue turnover is a characteristic of the most desirable lysosome-activating compound. Preferably, it is desired to use the lysosome-activating compound to restore kidney function. [0056]
  • DEFINITIONS [0057]
  • “Fragmented protein or fragment albumin” includes post-glomerular breakdown products after chemical, enzymatic or physical breakdown that occurs during renal passage. These components have a reduced size and/or may have changed hydrophobicity. [0058]
  • “Intact albumin, modified albumin, or modified form of albumin” as used herein means a compound having similar size and structural characteristics to native albumin, wherein the amino acid sequence is substantially the same as the native albumin. It is preferably a filtered intact protein. It elutes at or near the same position as native albumin on high-pressure liquid chromatography (HPLC) (FIG. 5). However, the structure has been modified biochemically either by minor enzyme mediated modification or addition to its basic structure and/or physically through a change in its three dimensional structure so that it escapes detection by conventionally used anti-albumin antibodies. Biochemical modification may be made by enzymes such as endo- or exo- peptidases. The 3D structure of albumin may have been altered in some way. Ligands may have bound to the albumin, or it may be any combination of these. The modified albumin detected in the method of the invention is not detectable by current and conventional radioimmunoassays using available antibodies and is not a fragment. [0059]
  • Conventional anti-albumin antibodies can be purchased from any purveyor of immunochemicals. For example, monoclonal antibody catalog numbers A6684 (clone no. HSA-11), and A2672 (clone no. HSA-9), as well as liquid whole serum, lyophilized fractionates, liquid IgG fraction, and the monoclonal antibodies in liquid ascites fluids form, can be obtained from Sigma, St. Louis, Mo., as found in the Immunochemicals section at pages 1151-1152 in the 1994 [0060] Sigma—Biochemicals Organic Compounds for Research and Diagnostic Reagents catalog.
  • As used herein, intact/modified albumin includes albumin that is substantially full-length, fragmented, chemically modified, or physically modified. As used herein, intact/modified albumin is meant to indicate albumin that is less than, equal to, or greater in molecular weight than the full-length albumin, and elutes at or near the native albumin position in a separation medium, such as chromatography, preferably HPLC, and most preferably hydrophobicity HPLC. As used herein, fragmented albumin is meant to refer to the fragment of albumin that is not detected by conventional anti-albumin antibody, and its presence is detected in diagnosing an early stage of renal disease and/or renal complications of a disease. The detection of the presence of intact/modified albumin is an indication of a predisposition to renal disease. [0061]
  • “Intact protein, modified protein or modified form of a protein” as used herein includes those forms of substantially full-length protein which are undetectable by conventional radioimmunoassay. The protein includes, but is not limited to, albumins, globulins (α-globulin(α[0062] 1-globulin, α2-globulin),β-globulins,γ-globulins), euglobulins, pseudoglobulin I and II, fibrinogen, α1, acid glycoprotein (orosomucoid), α1 glycoprotein, α1 lipoproteins, ceruloplasmin, α2 19S glycoprotein, β1 transferrin, β1 lipoprotein, immunoglobulins A, E ,G, and M, protein hormones including growth hormone, insulin, parathyroid hormone and other proteins including horseradish peroxidase, lactate dehydrogenase, glucose oxidase, myoglobin, and lysozyme.
  • “Kidney disease” as used herein includes any malfunction of the kidney. Kidney disease may be identified by the presence of intact or modified albumin in the urine. Preferably, an early diagnosis of the kidney disease may be made by detecting the presence of modified protein in the urine, or an increase in the modified protein in the urine over time. [0063]
  • “Low lysosome activity” as used herein is compared against normal levels of lysosome activity and/or lysosome machinery that traffics protein to the lysosome in a normal individual. The activity is insufficient for the lysosome to fragment proteins so that intact protein is excreted at a greater amount than at normally low levels. [0064]
  • “Lysosome-activating compound” as used herein refers to a compound that is beneficial to reactivation of the lysosome. The compound may work directly or indirectly on the lysosome resulting in activation of lysosomal function. These compounds may be selected from the group including, but not limited to, anticancer compounds, antiproliferation compounds, paracetamol, insulin vitamin A (retinoic acid) or derivatives of retinol, or compounds, including antibodies, to neutralize TGF-beta. [0065]
  • Osicka T. M. et al., [0066] Diabetologia 44: 230-236 (2001) examined the time course for the diabetes-related changes in renal lysosomal processing and to determine whether these changes can be prevented by aminoguanidine or ramipril treatment. The percentage desulphation of intravenously injected tritium labelled dextran sulphate ([3H]DSO4) in the urine, as determined by ion-exchange chromatography, was used as a marker of lysosomal sulphatase activity. The amount of totally desulphated [3H]DSO4 was significantly reduced in diabetic rats. The significant decrease in the amount of totally desulphated ([3H]DSO4) in the urine was not affected by drug treatment. However, the increase in totally sulphated ([3H]DSO4) in the urine collected from diabetic rats compared with that of control rats was normalised by treatment with both aminoguanidine or ramipril. These results indicate that the diabetes-induced changes in renal lysosomal processing may be one of the initial events in the development of diabetic nephropathy. Aminoguanidine and ramipril, which have been previously thought to have different mechanism of action, seem to prevent similar diabetes-induced changes in lysosomal processing either through their similar effects on enzyme activity within the lysosome or through their similar effects on the trafficking of molecules to and from the lysosome.
  • “Macroalbuminuria” is a condition where an individual excretes greater than 200 μg albumin/min in the urine as measured by conventional radioimmunoassay (RIA). [0067]
  • “Microalbuminuria” is a condition where an individual excretes at least 20 μg albumin/min in the urine as measured by conventional radioimmunoassay (RIA). RIA measures down to 15.6 ng/ml and is able to measure albumin in urine of normal subjects who have clearance of less than 6 μg/min. However, when albumin excretion exceeds 20 μg/min, treatment of the kidney disease is limited and full recovery is difficult from this point. [0068]
  • “Microalbuminuric” as used herein is a condition when albumin is detected in the urine at an excretion rate of at least 20 μg/min as measured by conventional RIA. [0069]
  • As used herein, “native” and “unmodified” are used interchangeably to describe a protein that is naturally found in an organism, preferably a human, which has not been modified by the filtering process of the renal glomeruli. [0070]
  • “Normal individual” as used herein is an individual who does not have a disease in which intact protein found in urine is an indicator of the disease. Preferably, the disease is kidney disease. [0071]
  • “Normal levels of lysosome activity” are levels of lysosome activity found in undiseased kidney of a normal individual. [0072]
  • “Normoalbuminuric” as used herein means a condition where albumin is excreted in the urine and is not detectable by RIA, or less than 20 μg/min (as measured by RIA) is excreted. [0073]
  • “Propensity for a disease” as used herein means that a disease may result in an individual as judged by a determination of the presence and excretion rate of a modified protein such as modified albumin. [0074]
  • “Proteinuria” as used herein is the existence of protein in the urine, usually in the form of albumin, a protein that is soluble in water and can be coagulated by heat. Related to this, “specific proteinuria” refers to the existence of a particular protein in the urine. [0075]
  • “Radioimmunoassay” as used herein is a method for detection and measurement of substances using radioactively labeled specific antibodies or antigens. [0076]
  • “Reactivation of the lysosome” as used herein includes an activation of lysosome activity preferably so that breakdown of proteins, particularly albumin, is increased compared with an inactivated state of the lysosome. [0077]
  • “Restore” as used herein means to restore in full or in part so that the component being restored has an improved function compared with its previous function. [0078]
  • The “sum of intact and intact modified protein” as used herein refers to the total amount of intact protein, and intact modified protein present in a biological sample. [0079]
  • “Total protein” as used herein refers to a particular filtered protein present in native, unmodified, modified or fragmented form that is excreted in urine. It includes protein that is not detected by conventional radioimmunoassay or conventional methods, which are currently available to detect the protein. Preferably the protein is albumin. [0080]
  • According to the present invention, the diseases to be treated include, but are not limited to renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, diabetic nephropathy, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycystic kidney disease, autosomal recessive polycystic kidney disease, tuborous sclerosis), von Hippel-Lindau disease, familial thin-glomerular basement membrane disease, collagen III glomerulopathy, fibronectin glomerulopathy, Alport's syndrome, Fabry's disease, Nail-Patella Syndrome, congenital urologic anomalies), monoclonal gammopathies (multiple myeloma, amyloidosis and related disrders), febrile illness (familial Mediterannean fever, HIV infection -AIDS), inflammatory disease (systemic vasculitides (polyarteritis nodosa, Wegener's granulomatosis, polyarteritis, necrotizing and crescentic glomerulonephritis), polymyositis-dermatomyositis, pancreatitis, rheumatoid arthritis, systemic lupus erythematosus, gout), blood disorders (sickle cell disease, thrombotic thrombocytopenia purpura, hemolytic-uremic syndrome, acute corticol necrosis, renal thromboembolism), trauma and surgery (extensive injury, burns, abdominal and vascular surgery, induction of anaesthesia), drugs (penicillamine, steroids) and drug abuse, malignant disease (epithelial (lung, breast), adenocarcinoma (renal), melanoma, lymphoreticular, multiple myeloma), circulatory disease (myocardial infarction, cardiac failure, peripheral vascular disease, hypertension, coronary heart disease, non-atherosclerotic cardiovascular disease, atherosclerotic cardiovascular disease), skin disease (psoriasis, systemic sclerosis), respiratory disease (COPD, obstructive sleep apnoea, hypoia at high altitude) and endocrine disease (acromegaly, diabetes mellitus, and diabetes insipidus). [0081]
  • Kidney disease may result from bacterial infection, allergies, congenital defects, stones, antibiotics, immunosuppressives, antineoplastics, nonsteroidal antiinflammatory drugs, analgesics, heavy metals, tumors, chemicals. [0082]
  • In one aspect of the invention, there is provided a method for determining a propensity for or early diagnosis of renal disease and/or renal complications of a disease. The method includes determining a change in the albumin content in a urine sample. The disease may be a kidney disease, although not necessarily limited to a kidney disease. [0083]
  • In the method of the invention, albumin is used herein only as an example of a protein to be detected in urine. When the albumin in a patient is analyzed by conventional RIA, it is expected that a normoalbuminuric patient or normal individual would have albumin in the urine in the range of 3-10 μg/min in young people and greater in older people. However, normoalbuminuric patients also show levels of albumin in the urine if measured by HPLC. Applicant has found that these levels may be in the order of 5 μg/min. As kidney disease progresses, the level of intact/modified albumin will increase to microalbuminuria levels in the order of 20 to 200 μg/min as determined by RIA. This will be much higher when determined by HPLC or a method that determines the sum of intact albumin and intact modified albumin. By monitoring the increase in intact/modified albumin, early signs of kidney disease may be detected. However, these levels are not detectable by the methods currently available such as radioimmunoassay using antibodies currently commercially in use, possibly for the reason that antibodies detect certain epitopes. If the albumin is modified in any way as described above, the epitope may be destroyed thereby leaving the modified albumin undetectable, [0084]
  • A patient suspected of having diabetic kidney disease will not show signs of kidney degeneration until well after 10 to 15 years when albumin is detected by currently available methods such as RIA methods. Urinary excretion rates of at least 20 μg/min may be detected by RIA when an individual enters a microalbuminuric state. Again, by observing the excretion of modified albumin, a change in the kidney and possibly onset of a kidney disease may be detected. [0085]
  • A normoalbuminuric subject, or normoalbuminuric diabetic patient may continue to have a low albumin excretion rate of less than 20 μg/min as determined by RIA, for many years. The presence of albumin in the urine is a sign that functions of the kidney may be impaired. Once this level begins to change, treatment may be initiated. [0086]
  • In a normal individual a small amount of albumin is detectable in the urine. Total filtered albumin appears mainly as fragmented albumin in urine. Some albumin may be detected in normoalbuminuric individuals. However, the excretion rate of albumin in urine in a normoalbuminuric individual may be as low as 5 μg/min. This level is generally detectable by RIA. [0087]
  • The modified protein of the invention can be detected by a variety of methods that are well-known in the art, including, but not limited to chromatography, electrophoresis and sedimentation, or a combination of these, which are described in Karger B L, Hancock W S (eds.) [0088] High Resolution Separation and Analysis of biological Macromolecules. Part A Fundamentals in Methods in Enzymology, Vol. 270, 1996, Academic Press, San Diego, Calif., USA; Karger B L, Hancock W S (eds.) High Resolution Separation and Analysis of biological Macromolecules. Part B Applications in Methods in Enzymology, Vol. 271, 1996, Acadermic Press, San Diego, Calif., USA; or Harding S E, Rowe, A J, Horton J C (eds.) Analytical Ultracentrifugation in Biochemistry and Polymer Science. 1992, Royal Soc. Chemistry, Cambridge, UK, which references are incorporated herein by reference in their entirety.
  • The electrophoresis method includes, but is not limited to, moving-boundary electrophoresis, zone electrophoresis, and isoelectric focusing. [0089]
  • The chromatography method includes, but is not limited to, partition chromatography, adsorption chromatography, paper chromatography, thin-layer chromatography, gas-liquid chromatography, gel chromatography, ion-exchange chromatography, affinity chromatography, and hydrophobic interaction chromatography. Preferably, the method is a sizing gel chromatography and hydrophobic interaction chromatography. More preferably, the method is hydrophobic interaction chromatography using a HPLC column. [0090]
  • The modified protein can also be detected by the use of specific albumin dyes. Such methods are described by Pegoraro et al., [0091] American Journal of Kidney Diseases 35(4): 739-744 (April 2000), the entire disclosure of which is hereby incorporated by reference. The modified albumin, as well as the whole albumin, is detectable by this dye method to provide the sum of modified albumin and whole or intact albumin. This detection method may be used with or without an initial separation of the albumin components from urine. Such dyes normally do not detect fragments <10,000 in molecular weight, but will detect the modified albumin.
  • In this dye method of detection, a dye such as Albumin Blue 580 is used. Such dyes are naturally non-fluorescent, but fluoresce on binding to intact albumin as well as the modified albumin, but do not bind to globulins. Therefore, globulins do not interfere with the assay so that measurements can be made in unfractionated urine. [0092]
  • Applicant has found that among diabetics, a normoalbuminuric diabetic patient has almost undetectable levels of modified or fragments of albumin when analyzed by conventional RIA. They appear to be normal. However, when the urine is tested by HPLC, the levels of modified albumin are much greater than found in a normal individual. This difference in albumin may be attributed to the inability of conventional RIA's to adequately detect all albumin (total albumin) in intact or modified forms. Thus, HPLC is preferred for generating a fragmentation profile. A fragmentation profile on HPLC is characterized by a series of peaks representing a number of species of albumin as fragments or in intact or modified forms. [0093]
  • In a preferred aspect of the present invention, the method of determining a propensity for or early diagnosis of a kidney disease in a subject is determined before the subject becomes microalbuminuric. [0094]
  • Measuring albumin content in a sample by an HPLC method of the present invention may provide different results from its measurement by conventional RIA. In the HPLC technique, a low level of albumin is observed in normal individuals. When the level of modified albumin begins to be detected and its level increases, and progresses toward microalbuminuria then a patient can be determined to have a propensity for kidney disease. [0095]
  • In a normal individual, the HPLC generated fragmentation profile is characterized by the absence of a peak in a region where full-length native albumin elutes. Instead, multiple fragmented albumin is detectable. A pure protein product (unmodified) produces essentially a single peak. For example, using a hydrophobicity HPLC, albumin was observed to elute in the range of 39-44 minutes (FIG. 5). Thus, a normal individual would provide a distinct fragmentation profile indicative of an absence of kidney disease or no propensity for a kidney disease. However, as kidney disease progresses, an increasing amount of modified albumin first, and then native form later are detectable. The fragmentation profile begins to change and more products in the region of full-length albumin manifests as additional spikes or an enlarged peak indicative of more intact/modified albumin in the urine. [0096]
  • In a HPLC generated fragmentation profile of a urine sample, the modified albumin may appear in a region where native albumin elutes but may be manifest as multiple peaks indicating the presence of multiple forms of modified albumin. [0097]
  • In a further preferred embodiment, the propensity for kidney disease may be measured by determining the presence of or identifying at least one species of modified albumin. This may be determined or identified by the presence of a specific peak on a HPLC profile, preferably the peak is within the range of position that corresponds to the elution position of the native albumin. [0098]
  • A HPLC column for detecting modified albumin or unmodified albumin may be a hydrophobicity column, such as Zorbax 300 SB-CB (4.6 mm×150 mm). A 50 μl sample loop may be used. Elution solvents suitable for HPLC in detecting albumin and its breakdown products may include standard elution solvents such as acetonitrile solvents. Preferably a buffer of water/1% trifluoro acetic acid (TFA) followed by a buffer of 60% acetonitrile/0.09% TFA may be used. A gradient of 0 to 100% of a 60% acetonitrile/0.09% TFA has been found to be suitable. [0099]
  • Suitable HPLC conditions for a hydrophobicity column may be as follows: [0100]
  • Solvent A H[0101] 2O, 1% trifluoro acetic acid
  • [0102] Solvent B 60% acetonitrile, 0.09% TFA
  • Solvent A2 99.96>00.00:49.58 min Pressure 9.014Mpascalls (˜1100 psi) [0103]
  • Solvent B2 0.04>100.0:49.58 min Pressure 7.154Mpascalls [0104]
  • The wavelength used in HPLC may be approximately 214 nm. [0105]
  • Modified albumin may elute between 39-44 minutes (FIG. 5). Albumin fragments may elute much earlier, mainly at less than 20 minutes. [0106]
  • The method for determining the propensity for kidney disease is applicable to any individual. Kidney disease may be caused by a number of factors including bacterial infection, allergic, congenital defects, stones, tumors, chemicals or from diabetes. Preferably, the method is applicable for determining a propensity for kidney disease in diabetic patients that may progress to a kidney disease. Preferably, the individual is a normoalbuminuric diabetic. However, normal individuals may be monitored for propensity for the disease by determining increased levels of intact or modified albumin in the urine. [0107]
  • The method of the invention can be carried out using non-antibody separation procedures as described above. However, antibody specific for modified protein may also be used to detect the presence of the modified protein. [0108]
  • The antibody to the modified protein may be obtained using the following method. The procedure is described specifically for albumin by way of example only, and can be readily applied to antibody production against any other protein in the urine. The method seeks to determine which modified albumin molecule is the most sensitive marker to identify diabetic patients, for example, who will progress to kidney complications. [0109]
  • The modified albumin is characterized by carrying out a quantitative separation of the modified albumin molecules, such as by preparative HPLC. The modified proteins are analyzed for ligand binding, such as glycation. Subsequently, amino acid sequence of the individual modified protein is determined, preferably by mass spectrometry using methods described in Karger B L, Hancock W S (eds.) [0110] High Resolution Separation and Analysis of biological Macromolecules. Part A Fundamentals in Methods in Enzymology, Vol. 270, 1996, Academic Press, San Diego, Calif., USA; or Karger B L, Hancock W S (eds.) High Resolution Separation and Analysis of biological Macromolecules. Part B Applications in Methods in Enzymology, Vol. 271, 1996, Academic Press, San Diego, Calif., USA, for example, which references are incorporated herein by reference in their entirety. In a preferred embodiment, there may be about 3 to 4 modified albumin species.
  • The method of generating antibody against the modified albumin seeks to develop a diagnostic immunoassay for the modified albumin that predicts those diabetic patients, for example, that progress to kidney complications. To accomplish this, sufficient quantities of modified albumin is prepared by HPLC. Antibodies are made by sequential injection of the modified albumin in an animal such as a rabbit, to generate good titer, and the antibodies are isolated using conventional techniques using methods described in Goding J W, Monoclonal Antibodies: Principles and Practice. [0111] Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology, 2nd Edition 1986, Academic Press, London, UK; or Johnstone A, Thorpe R, Immunochemistry in Practice, 3rd edition 1996, Blackwell Science Ltd, Oxford, UK, for example, which references are incorporated herein by reference in their entirety. The obtained antibodies may be polyclonal antibodies or monoclonal antibodies.
  • Preferably, at least one species of a modified albumin is isolated and identified for use in determining a propensity for kidney disease. The isolated species may be used to generate antibodies for use in immunoassays. The antibodies may be tagged with an enzymatic, radioactive, fluorescent or chemiluminescent label. The detection method may include, but is not limited to radioimmunoassay, immunoradiometric assay, fluorescent immunoassay, enzyme linked immunoassay, and protein A immunoassay. The assays may be carried out in the manner described in Goding J W, [0112] Monoclonal Antibodies: Principles and Practice. Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology. 2nd Edition 1986, Academic Press, London, UK; Johnstone A, Thorpe R, Immunochemistry in Practice, 3rd edition 1996, Blackwell Science Ltd, Oxford, UK; or Price C P, Newman D J (eds.) Principles and Practice of Immunoassay, 2nd Edition, 1997 Stockton Press, New York, N.Y., USA, for example, which references are incorporated herein by reference in their entirety.
  • It is an object of this invention to provide an article of matter or a kit for rapidly and accurately determining the presence or absence of modified protein such as modified albumin, in a sample quantitatively or non-quantitatively as desired. Each component of the kit(s).may be individually packaged in its own suitable container. The individual container may also be labeled in a manner, which identifies the contents. Moreover, the individually packaged components may be placed in a larger container capable of holding all desired components. Associated with the kit may be instructions, which explain how to use the kit. These instructions may be written on or attached to the kit. [0113]
  • The invention is also directed to a method of determining a treatment agent for renal disease and/or renal complications of a disease, comprising: [0114]
  • (a) administering to a person an agent that is suspected of being able to treat the disease; [0115]
  • (b) obtaining a urine sample from the person; and [0116]
  • (c) assaying for the modified form of the protein in the sample, wherein either the presence of or lack of presence of a modified form of the protein in the urine or decreasing amount of the modified form of the protein over time indicates that the agent is a treatment agent for the disease. The treatment agent may be a lysosome activating agent that may act directly or indirectly to activate lysosome, and thereby cause the lysosome to digest post-glomerular filtered proteins, which is a sign of a healthy kidney. [0117]
  • The process of trafficking of proteins to the lysosomes plays a role in the mechanism of albuminuria in diabetes. An intracellular molecule that is involved in trafficking is protein kinase C (PKC). It is contemplated that a drug or agent can be formulated that will activate lysosomal trafficking or inhibit PKC. [0118]
  • Accordingly, in one aspect of the present invention, there is provided a lysosome-activating compound for use in reactivating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome. [0119]
  • In another aspect of the present invention, there is provided a composition comprising a lysosome-activating compound and a carrier. [0120]
  • In yet another aspect of the invention there is provided a method of preventing or treating kidney disease, said method including administering an effective amount of a lysosome-activating compound to a subject. [0121]
  • In yet another aspect of the present invention, there is provided a method of screening a multiplicity of compounds to identify a compound capable of activating lysosomes or processes that direct substrates to the lysosome or products away from the lysosome, said method including the steps of: [0122]
  • (a) exposing said compound to a lysosome and assaying said compound for the ability to activate a lysosome wherein said lysosome when activated has a changed activity; [0123]
  • (b) assaying for the ability to restore a cellular process to substantially normal levels in kidney tissue, wherein said kidney tissue has a low lysosome activity; and/or [0124]
  • (c) assaying for the ability to restore tissue turnover to substantially normal levels in kidney tissue, wherein said kidney tissue has low lysosome activity. [0125]
  • Lysosomes may be associated with the breakdown of proteins, particularly albumin, in the kidney. In cases of microalbuminuria, substantial amounts of albumin escape lysosomal breakdown possibly due to a deactivated lysosome. Restoration of lysosomal breakdown may restore the balance in the kidney of cellular processes and tissue turnover. [0126]
  • A lysosome-activating compound may be a compound that acts directly or indirectly on the lysosome. By acting indirectly, the compound may act on a component, which influences the activity of the lysosome. Nevertheless, the outcome results in an activation of the lysosome, thereby providing enhanced protein breakdown. [0127]
  • In another aspect of the present invention, there is provided a composition comprising a lysosome-activating compound and a carrier. [0128]
  • The composition may be a physiologically acceptable or pharmaceutically acceptable composition. However, it will be a composition which allows for stable storage of the lysosome activating compound. Where the composition is a pharmaceutically acceptable composition, it may be suitable for use in a method of preventing or treating kidney disease. [0129]
  • In yet another aspect of the invention there is provided a method of preventing or treating kidney disease, said method including administering an effective amount of a lysosome-activating compound to a subject. [0130]
  • As described above, the lysosome-activating compound may act by reactivating the lysosome so that cellular processes and tissue turnover are restored fully or in part, thereby resulting in the kidney being restored partially or fully. In any case, administering a lysosome activating compound to an animal having kidney disease may restore lysosome activity fully or in part. [0131]
  • Methods of administering may be oral or parenteral. Oral may include administering with tablets, capsules, powders, syrups, etc. Parenteral administration may include intravenous, intramuscular, subcutaneous or intraperitoneal routes. [0132]
  • The changed activity of the lysosome is preferably a change which enhances the activity of the lysosome so that albumin breakdown is improved. The ability to not only activate lysosome but also improve cellular processes and/or tissue turnover is a characteristic of the most desirable lysosome activating compound. Preferably, it is desired to use the lysosome activating compound to restore kidney function. [0133]
  • In another aspect of the present invention there is provided a method for preventing kidney disease in a subject, said method including: [0134]
  • (a) measuring the amount of intact and modified intact albumin content in a urine sample; [0135]
  • (b) determining a change in the amount of intact albumin in the urine that has been modified so as to be not detectable by conventional RIA methods wherein the change is indicative of a propensity for kidney disease; and [0136]
  • (c) treating the animal for a kidney disease when a change is determined. [0137]
  • The following examples are offered by way of illustration of the present invention, and not by way of limitation. [0138]
  • EXAMPLES Example 1 Size Exclusion Chromatography of Human Serum Albumin (HSA)
  • Normal, healthy volunteers were used to provide urine for analyzing the distribution of albumin in their urine. [0139]
  • [0140] 3H[HSA] (Human Serum Albumin) was injected into healthy volunteers and urine and plasma were collected and analyzed by size exclusion chromatography using a G-100 column. The column was eluted with PBS (pH=7.4) at 20 ml/hr at 4° C. The void volume (V0) of the column was determined with blue dextran T2000 and the total volume with tritiated water.
  • Tritium radioactivity was determined in 1 ml aqueous samples with 3 ml scintillant and measured on a Wallac 1410 liquid scintillation counter (Wallac Turku, Finland). [0141]
  • FIG. 2 illustrates the distribution of albumin in urine and in plasma. [0142]
  • Example 2 Albumin Excretion in a Normal, Healthy Volunteer and Diabetic Patient
  • [0143] 3H[HSA] as used in Example 1 was injected into a normal, healthy volunteer and a diabetic patient. Samples of urine were collected and 3H[HSA] was determined as in Example 1.
  • The normal, healthy volunteer (FIG. 3) shows the excretion of fragments of albumin on a size exclusion chromatography as performed in Example 1. [0144]
  • The diabetic patient (FIG. 4) shows the presence of substantially full-length and fragmented albumin on size exclusion chromatography. However, excretion rates of albumin detectable by these methods were in the order of 5 μg/min (control) and 1457 μg/min (diabetic). [0145]
  • Example 3 Determination of Intact Albumin, and Intact/Modified Albumin on HPLC.
  • Urine samples were collected from normal, healthy volunteer, normoalbuminuric diabetic patients and from macroalbuminuric patients. Urine was collected midstream in 50 ml urine specimen containers. The urine was frozen until further use. Prior to HPLC analysis the urine was centrifuged at 5000 g. [0146]
  • Samples were analyzed on HPLC using a hydrophobicity column Zorbax 300 SB-CB (4.6 mm×150 mm). A 50 μl sample loop was used. [0147]
  • Samples were eluted from the columns using the following conditions. [0148]
  • Solvent A H[0149] 2O, 1% trifluoro acetic acid
  • [0150] Solvent B 60% acetonitrile, 0.09% TFA
  • Solvent A2 99.96>00.00:49.58 min Pressure 9.014Mpascalls (˜1100 psi) [0151]
  • Solvent B2 0.04>100.0:49.58 min Pressure 7.154Mpascalls [0152]
  • A wavelength of 214 nm was used. [0153]
  • Example 4 Purification of Modified Albumin for Antibody Production by Standard Techniques
  • Urine from microalbuminuric patient which had an intact albumin concentration of 43.5 mg/L as determined by turbitimer (involving conventional immunochemical assay) was initially filtered through a 30 kDa membrane to separate the modified albumin from low molecular weight (<30,000) protein fragments in urine. The material that was retained by the filter gave a yield of intact albumin of 27.4 mg/L as determined by turbitimer assay. This retained material was then subjected to size exclusion chromotography on Sephadex G100. The material collected was the peak fraction that coelutes with intact albumin. This material gave a yield of 15.2 ml/L of albumin as determined by the turbitimer method. This material was then subjected to affinity chromatography on an intact,albumin antibody column. This column will only bind albumin that has conventional epitopes. The yield of material that eluted from the column was <6 mg/L (lowest sensitivity of the turbitimer). This is expected as the immunoreactive albumin would have bound to the affinity column. The eluate was then subject to reverse phase HPLC chromatography(as described above) to determine the amount of immuno-unreactive albumin in the sample. A 1452 unit area corresponding to 30.91 mg/L of purified modified albumin was noted as shown in FIG. 15. This purified modified albumin can then be used for antibody production by standard means. [0154]
  • Results [0155]
  • FIG. 5 illustrates a HPLC profile of albumin alone. Essentially a single peak which elutes at approximately 39-44 minutes retention time was obtained. [0156]
  • FIG. 6 illustrates a HPLC profile of plasma showing a distinct albumin peak at approximately 39-44 minutes as well as other peaks corresponding to other plasma proteins. [0157]
  • FIG. 7 illustrates a HPLC profile of a normal, healthy volunteer showing no albumin peak in the urine sample. This individual breaks down the albumin excreted into the urine possibly via an active lysosome. Substantial fragmented products were evident showing prominence of some species, particularly of a species at approximately less than 14.5 minutes retention time. [0158]
  • When urine from a normoalbuminuric diabetic patient (with an albumin excretion rate of 8.07 μg/min, as measured by RIA) is analyzed (FIG. 8), small amounts of modified albumin eluting at approximately 39-44 minutes retention time is evident. Whereas conventional test indicates the presence of <6 mg/l of albumin in the urine sample, the method of the invention showed that the true albumin content in the urine sample was 26.7 mg/l. Treatment for the disease should have begun on this individual. Albumin by-products or fragmented albumin is present as in the normal, healthy volunteer. [0159]
  • Another urine sample from normoalbuminuric diabetic patient (with albumin excretion rate of 17.04 μg/min) was analyzed (FIG. 9). RIA tests show albumin excreted in the urine for this patient. However, on HPLC (FIG. 9) an albumin or modified albumin peak is evident at approximately 39-44 minutes retention time. Whereas conventional test indicates the presence of <6 mg/l of albumin in the urine sample, the method of the invention showed that the true albumin content in the urine sample was 81.3 mg/l. Treatment for the disease should have begun on this individual. This peak begins to show a multiple peaked appearance. A smaller peak corresponding to intact albumin shows that modified albumin may represent the peak at 39-44 minutes. The presence of this albumin peak compared with the profile of a normal, healthy volunteer having no albumin peak shows a change in the detectable levels of the amount of intact/modified albumin. This may signal a propensity for a kidney disease. [0160]
  • A further urine sample from a normoalbuminuric diabetic patient (with an albumin excretion rate of 4.37 μg/min) was analyzed, and the HPLC profile is illustrated in FIG. 10. Again, modified albumin was detected at approximately 39-44 minutes retention time showing multiple peaks. This patient again did register normal albumin by RIA. Whereas conventional test indicates the presence of <6 mg/l of albumin in the urine sample, the method of the invention showed that the true albumin content in the urine sample was [0161] 491 mg/l. Treatment for the disease should have begun on this individual. It is clear that modified albumin assessment is necessary to identify these changes. This patient would be determined to have a propensity for kidney disease. As kidney disease progresses, the modified albumin peak will continue to increase.
  • This is shown in FIG. 11 where a urine sample of a macroalbuminuric patient was analyzed. A quite significant albumin peak at approximately 39-44 minutes retention time showing multiple peaks was evident. The patient's albumin content was 1796 mg/l. Treatment for this individual is in progress. [0162]
  • The method of the invention results in early detection of a propensity for a renal disease as illustrated by the longitudinal studies in FIGS. [0163] 12-14. FIGS. 12-14 show situations in which the ACE inhibitor treatment for diabetes was begun later than it should have had the modified albumin detection method of the invention been used. Detecting modified protein using the method according to the invention is a more effective method for predicting the onset of a renal disease than using conventional RIA.
  • Example 5 (3H)HSA in Urine Collected from a Microalbuminuric Type I Diabetic
  • FIG. 16 illustrates ([0164] 3H)HSA in urine collected from a microalbuminuric type I diabetic patient by size exclusion chromatography. Urine contains mostly fragmented albumin. Plasma contains only intact albumin. Conventional RIA (human albumin ordinate) measures only intact albumin and not fragments.
  • Example 4 Size Exclusion Profiles of (3H)HSA in Patient Urine
  • When lysosome activity or intracellular processes directing substrates to lysosomes is reduced as in kidney disease, more of the high molecular weight, and substantially full length filtered protein including albumin appears in the urine. FIG. 17 shows that in renal disease in type I diabetic patients the severity of the disease as measured by albuminuria is associated with the increased presence of intact albumin in urine. [0165]
  • FIG. 17 illustrates the size exclusion profiles of([0166] 3H)HSA in urine from four different patients with varying albumin excretion rate (AER) as measured by conventional RIA. As intact albuminuria increases the amount of intact albumin in the urine increases and the relative amount of fragments decreases. The total excretion represents the excretion of intact albumin plus albumin derived fragments.
  • All of the references cited herein are incorporated by reference in their entirety. [0167]
  • Finally, it is to be understood that various other modifications and/or alterations may be made without departing from the spirit of the present invention as outlined herein. [0168]

Claims (21)

What is claimed is:
1. A method for treating a person with a renal disease and/or renal complications of a disease, comprising:
(a) administering a treatment agent to a person in need thereof;
(b) obtaining a sample of body fluid from the person; and
(c) assaying for a protein in the sample, wherein either presence of or lack of presence of the protein in the urine or decreasing amount of the protein over time in the urine indicates that the treatment agent is therapeutically effective for the renal disease and/or renal complications of a disease.
2. The method according to claim 1, wherein the renal disease and/or renal complications of the disease is selected from the group consisting of nephropathy, diabetes insipidus, diabetes type I, diabetes II, renal disease (glomerulonephritis, bacterial and viral glomerulonephritides, IgA nephropathy and Henoch-Schönlein Purpura, membranoproliferative glomerulonephritis, membranous nephropathy, Sjögren's syndrome, nephrotic syndrome (minimal change disease, focal glomerulosclerosis and related disorders), acute renal failure, acute tubulointerstitial nephritis, pyelonephritis, GU tract inflammatory disease, Pre-clampsia, renal graft rejection, leprosy, reflux nephropathy, nephrolithiasis), genetic renal disease (medullary cystic, medullar sponge, polycystic kidney disease (autosomal dominant polycystic kidney disease, autosomal recessive polycystic kidney disease, tuborous sclerosis), von Hippel-Lindau disease, familial thin-glomerular basement membrane disease, collagen III glomerulopathy, fibronectin glomerulopathy, Alport's syndrome, Fabry's disease, Nail-Patella Syndrome, congenital urologic anomalies), monoclonal gammopathies (multiple myeloma, amyloidosis and related disorders), febrile illness (familial Mediterranean fever, HIV infection -AIDS), inflammatory disease (systemic vasculitides (polyarteritis nodosa, Wegener's granulomatosis, polyarteritis, necrotizing and crescentic glomerulonephritis), polymyositis-dermatomyositis, pancreatitis, rheumatoid arthritis, systemic lupus erythematosus, gout), blood disorders (sickle cell disease, thrombotic thrombocytopenia purpura, hemolytic-uremic syndrome, acute corticol necrosis, renal thromboembolism), trauma and surgery (extensive injury, burns, abdominal and vascular surgery, induction of anesthesia), drugs (penicillamine, steroids) and drug abuse, malignant disease (epithelial (lung, breast), adenocarcinoma (renal), melanoma, lymphoreticular, multiple myeloma), circulatory disease (myocardial infarction, cardiac failure, peripheral vascular disease, hypertension, coronary heart disease, non-atherosclerotic cardiovascular disease, atherosclerotic cardiovascular disease), skin disease (psoriasis, systemic sclerosis), respiratory disease (COPD, obstructive sleep apnoea, hypoia at high altitude) and endocrine disease (acromegaly, diabetes mellitus, diabetes insipidus).
3. The method according to claim 1, wherein the treatment agent is a lysosome-activating compound.
4. The method according to claim 3, wherein the lysosome-activating compound is selected from the group consisting of ACE inhibitors, anti-glycation agents, anticancer compounds, antiproliferation compounds, and compounds that neutralize TGF-beta.
5. The method according to claim 3, wherein the lysosome-activating compound is selected from the group consisting of ramipril, aminoguanidine, paracetamol, vitamin A (retinoic acid), retinol derivatives, and anti-TGF beta antibodies.
6. The method according to claim 1, wherein the sample of body fluid is a urine, blood or laboratory sample.
7. The method according to claim 1, wherein the protein comprises albumin, globulin (α-globulin(α1-globulin, α2-globulin),β-globulin,γ-globulin), euglobulin, pseudoglobulin I and II, fibrinogen, α1 acid glycoprotein (orosomucoid), α1 glycoprotein, α1 lipoprotein, ceruloplasmin, α2 19S glycoprotein, β1 transferrin, β1 lipoprotein, immunoglobulins A, E, G, and M, horseradish peroxidase, lactate dehydrogenase, glucose oxidase, myoglobin, lysozyme, protein hormone, growth hormone, insulin, or parathyroid hormone.
8. The method according to claim 1, wherein the assaying for a protein in the sample comprises a method selected from the group consisting of:
(a) assaying for albumin by a conventional method; and
(b) assaying for intact modified albumin.
9. The method according to claim 8, wherein the conventional method comprises a method selected from the group consisting of:
(a) an antibody method, and
(b) a non-antibody method comprising loading the sample on a chromatography, electrophoresis or sedimentation apparatus to test for native or intact modified albumin.
10. The method according to claim 9, wherein the albumin is detected by an antibody that is specific for both unmodified and modified forms of the protein.
11. The method according to claim 9, wherein the albumin is detected by an antibody that is specific for the modified protein.
12. The method according to claim 9, wherein the albumin is detected by an antibody that is attached to an enzymatic, radioactive, fluorescent or chemiluminescent label, wherein the detecting step comprises radioimmunoassay, immunoradiometric assay, fluorescent immunoassay, enzyme linked immunoassay, or protein A immunoassay.
13. The method according to claim 1, wherein the assaying for a protein in the sample comprises the steps of:
(i) detecting native albumin amount by conventional antibody assay:
(ii) detecting intact modified albumin by a non-antibody method; and
(iii) adding the values obtained in (i) and (ii) to obtain an accurate reading of total albumin content in the sample.
14. The method according to claim 13, wherein the non-antibody method comprises loading the sample on a chromatography, electrophoresis or sedimentation apparatus to test for native or intact modified albumin.
15. The method according to claim 1, wherein the assaying for a protein in the sample is a non-antibody method comprising detecting a sum of native protein and intact modified protein in a sample.
16. The method according to claim 1, wherein the assaying for a protein in the sample is by a method selected from the group consisting of partition chromatography, adsorption chromatography, paper chromatography, thin-layer chromatography, gas-liquid chromatography, gel chromatography, ion-exchange chromatography, affinity chromatography, or hydrophobic interaction chromatography, moving-boundary electrophoresis, zone electrophoresis, or isoelectric focusing.
17. The method according to claim 1, wherein the assaying for a protein in the sample is by hydrophobic interaction chromatography carried out in a high pressure liquid chromatography (HPLC) apparatus.
18. The method according to claim 1, wherein the assaying for a protein in the sample is by detecting albumin in the sample with specific albumin dyes.
19. The method according to claim 1, wherein an early stage of the disease is diagnosed when modified albumin is present in the sample in increasing amounts over time.
20. A method for identifying a treatment agent for renal disease and/or renal complications of a disease, comprising:
(a) administering to a person in need thereof an agent that is suspected of being able to treat the disease;
(b) obtaining a urine sample from the person; and
(c) assaying for a protein in the sample, wherein either presence of or lack of presence of the protein in the urine or decreasing amount of the protein over time in the urine indicates that the agent is a treatment agent for the renal disease and/or renal complications of a disease.
21. The method of claim 19, wherein the for a protein in the sample comprises assaying for a modified form of albumin in the sample, wherein either presence of or a lack of presence of the modified form of the protein in the sample or decreasing amount of the modified form of the protein over time in the urine indicates that the agent is a treatment agent for the renal disease and/or renal complications of a disease.
US10/721,351 1998-12-21 2003-11-26 Method for kidney disease treatment by drug intervention Abandoned US20040106155A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/721,351 US20040106155A1 (en) 1998-12-21 2003-11-26 Method for kidney disease treatment by drug intervention

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
AUPP7843 1998-12-21
AUPP7843A AUPP784398A0 (en) 1998-12-21 1998-12-21 Kidney disease detection and treatment
US09/415,217 US6447989B1 (en) 1998-12-21 1999-10-12 Kidney disease detection and treatment
WOPCT/IB99/02029 1999-12-20
PCT/IB1999/002029 WO2000037944A1 (en) 1998-12-21 1999-12-20 Kidney disease detection and treatment
US09/893,346 US20020012906A1 (en) 1998-12-21 2001-06-28 Method for kidney disease treatment by drug intervention
US10/721,351 US20040106155A1 (en) 1998-12-21 2003-11-26 Method for kidney disease treatment by drug intervention

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/893,346 Continuation US20020012906A1 (en) 1998-12-21 2001-06-28 Method for kidney disease treatment by drug intervention

Publications (1)

Publication Number Publication Date
US20040106155A1 true US20040106155A1 (en) 2004-06-03

Family

ID=25645955

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/893,346 Abandoned US20020012906A1 (en) 1998-12-21 2001-06-28 Method for kidney disease treatment by drug intervention
US09/892,797 Expired - Fee Related US6589748B2 (en) 1998-12-21 2001-06-28 Method for kidney disease detection and treatment
US10/721,351 Abandoned US20040106155A1 (en) 1998-12-21 2003-11-26 Method for kidney disease treatment by drug intervention

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/893,346 Abandoned US20020012906A1 (en) 1998-12-21 2001-06-28 Method for kidney disease treatment by drug intervention
US09/892,797 Expired - Fee Related US6589748B2 (en) 1998-12-21 2001-06-28 Method for kidney disease detection and treatment

Country Status (13)

Country Link
US (3) US20020012906A1 (en)
EP (1) EP1141728B1 (en)
JP (2) JP4160263B2 (en)
CN (1) CN1331801A (en)
AT (1) ATE376187T1 (en)
AU (1) AU1674400A (en)
BR (1) BR9916407A (en)
CA (1) CA2356174A1 (en)
DE (1) DE69937368T2 (en)
DK (1) DK1141728T3 (en)
IL (1) IL143892A0 (en)
MX (1) MXPA01006404A (en)
WO (1) WO2000037944A1 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050112123A1 (en) * 2003-10-06 2005-05-26 Vaughan Michael R. Methods of treating proteinuria
US20060062783A1 (en) * 2003-08-08 2006-03-23 Lorin Roskos Antibodies against parathyroid hormone
US20110174062A1 (en) * 2008-08-29 2011-07-21 Joseph Anderberg Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110195429A1 (en) * 2008-08-28 2011-08-11 Astute Medical Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110201038A1 (en) * 2008-10-21 2011-08-18 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110207161A1 (en) * 2008-10-21 2011-08-25 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110229915A1 (en) * 2008-11-22 2011-09-22 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20120264629A1 (en) * 2009-08-07 2012-10-18 Joseph Anderberg Methods and compositions for diagnosis and prognosis of renal injury and renal failure
KR101215759B1 (en) 2010-11-01 2012-12-26 서울대학교산학협력단 Uses of Compositions Comprising Orosomucoid
CN102854325A (en) * 2011-07-01 2013-01-02 复旦大学 Method for detecting affinity indicators of reversible binding of insulin and serum protein and application thereof
US8993250B2 (en) 2008-11-10 2015-03-31 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9029093B2 (en) 2010-02-26 2015-05-12 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9229010B2 (en) 2009-02-06 2016-01-05 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9360488B2 (en) 2013-01-17 2016-06-07 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10324093B2 (en) 2009-11-07 2019-06-18 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10823742B2 (en) 2010-06-23 2020-11-03 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10830773B2 (en) 2009-12-20 2020-11-10 Astute Medical, Inc. Methods for prognosis of future acute renal injury and acute renal failure
US10928403B2 (en) 2010-06-23 2021-02-23 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10935548B2 (en) 2011-12-08 2021-03-02 Astute Medical, Inc. Methods for diagnosis and prognosis of renal injury and renal failure using insulin-like growth factor-binding protein 7 and metalloproteinase inhibitor 2
US11243217B2 (en) 2016-06-06 2022-02-08 Astute Medical, Inc. Management of acute kidney injury using insulin-like growth factor-binding protein 7 and tissue inhibitor of metalloproteinase 2
US11454635B2 (en) 2010-02-05 2022-09-27 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPP784398A0 (en) 1998-12-21 1999-01-21 Monash University Kidney disease detection and treatment
BR9916407A (en) 1998-12-21 2001-09-25 Univ Monash Detection and treatment of kidney disease
US7250294B2 (en) * 2000-05-17 2007-07-31 Geron Corporation Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
DE60234759D1 (en) * 2001-03-28 2010-01-28 Heska Corp IDENTIFICATION PROCEDURE FOR AN EARLY KIDNEY IN ANIMALS
US20030003588A1 (en) * 2001-06-28 2003-01-02 Comper Wayne D. Method for kidney disease detection by protein profiling
JP2006518251A (en) * 2003-02-13 2006-08-10 ベクトン・ディキンソン・アンド・カンパニー Apparatus for removing components during blood collection and use thereof
US7482128B2 (en) 2003-03-27 2009-01-27 Heska Corporation Anti-feline albumin antibodies
US20060286602A1 (en) * 2004-05-10 2006-12-21 Harald Mischak Method and markers for the diagnosis of renal diseases
JP5785355B2 (en) * 2004-11-24 2015-09-30 ヒルズ・ペット・ニュートリシャン・インコーポレーテッド Methods for improving liver clearance of xenobiotic substances in animals
US20090176864A1 (en) * 2004-11-24 2009-07-09 Hill's Pet Nutrition, Inc. Methods For Improving Hepatic and Immune Function In An Animal
US8647660B2 (en) 2004-12-29 2014-02-11 Hill's Pet Nutrition, Inc. Combination of limited nutrients and enhanced dietary antioxidants to impart improved kidney health
US8252742B2 (en) 2004-12-30 2012-08-28 Hill's Pet Nutrition, Inc. Methods for enhancing the quality of life of a senior animal
EP1830629A4 (en) * 2004-12-30 2008-08-27 Hills Pet Nutrition Inc Methods for enhancing the quality of life of a senior animal
US20060205818A1 (en) * 2005-03-08 2006-09-14 Burzynski Stanislaw R Method for the treatment of von Hippel-Lindau (VHL) disease with phenylacetyl-derivatives
CN101198259B (en) * 2005-04-19 2014-05-28 希尔氏宠物营养品公司 Methods and compositions for the prevention and treatment of kidney disease
RU2398447C2 (en) * 2005-04-29 2010-09-10 Хилл'С Пет Ньютришн, Инк. Methods for prolongation of cats' lives
EP1885203B1 (en) * 2005-05-20 2018-01-03 Hill's Pet Nutrition, Inc. Methods for promoting health or wellness in adult animals
US9149062B2 (en) 2005-06-29 2015-10-06 Hill's Pet Nutrition, Inc. Methods and compositions for the prevention and treatment of kidney disease
BRPI0615211A2 (en) * 2005-08-17 2013-01-15 Hills Pet Nutrition Inc method, method for preventing or treating renal disease or improving renal function, kit, and means for communicating information on or instructions for mixing and administering one or more of the compositions, composition components, food compositions, food ingredients and renal medicines and for the use of renal diagnostic devices
BRPI0808703A2 (en) * 2007-03-07 2014-09-02 Mosaiques Diagnostics & Therap Process for Standardization of Component Concentration for Analysis in a Urine Sample
EP1972940A1 (en) * 2007-03-14 2008-09-24 mosaiques diagnostics and therapeutics AG Method and marker for diagnosing kidney disease
EP2153234A2 (en) * 2007-05-11 2010-02-17 The Institutes for Pharmaceutical Discovery, LLC Methods for early diagnosis of kidney disease
AU2008309605A1 (en) * 2007-10-09 2009-04-16 Mosaiques Diagnostics And Therapeutics Ag Polypeptide marker for the diagnosis of prostate cancer
EP2051078A1 (en) * 2007-10-19 2009-04-22 mosaiques diagnostics and therapeutics AG Method and marker for diagnosing diabetes mellitus
EP2255203A2 (en) * 2008-03-19 2010-12-01 Mosaiques Diagnostics And Therapeutics AG Method and marker for diagnosis of tubular kidney damage and illnesses
SI2313778T1 (en) 2008-07-18 2015-06-30 Boston Medical Center Corporation Diagnostics for membranous nephropathy
US20110214990A1 (en) * 2008-09-17 2011-09-08 Mosaiques Diagnostics And Therapeutics Ag Kidney cell carcinoma
US8481324B2 (en) 2008-12-04 2013-07-09 Technion Research And Development Foundation Ltd. Apparatus and methods for diagnosing renal disorders
EP2391653B8 (en) * 2009-01-28 2015-09-30 Industrial Technology Research Institute Biomarkers associated with nephropathy
JP5775874B2 (en) * 2009-08-28 2015-09-09 アスチュート メディカル,インコーポレイテッド Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EA201290195A1 (en) * 2009-11-07 2012-12-28 Астьют Медикал, Инк. METHODS AND COMPOSITIONS FOR DIAGNOSIS AND PREDICTION OF KIDNEY DAMAGE AND RENAL FAILURE
CA2793959C (en) 2010-03-25 2019-06-04 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
US8930222B2 (en) * 2010-08-05 2015-01-06 Abbott Laboratories Method and system for managing patient healthcare
DE102010038014B4 (en) 2010-10-06 2021-10-07 Numares Ag Use of specific substances as markers to determine the risk of kidney rejection
AU2012267786B2 (en) 2011-06-10 2017-08-03 Oregon Health & Science University CMV glycoproteins and recombinant vectors
AU2012216792A1 (en) 2011-09-12 2013-03-28 International Aids Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing HIV-1 proteins by broadly neutralizing antibodies
US9402894B2 (en) 2011-10-27 2016-08-02 International Aids Vaccine Initiative Viral particles derived from an enveloped virus
US20130115640A1 (en) * 2011-11-03 2013-05-09 James A. Tumlin ACTH for Treatment of Kidney Disease
US9689826B2 (en) 2012-03-11 2017-06-27 Technion Research And Development Foundation Ltd. Detection of chronic kidney disease and disease progression
EP2679596B1 (en) 2012-06-27 2017-04-12 International Aids Vaccine Initiative HIV-1 env glycoprotein variant
JP2016188761A (en) * 2013-08-23 2016-11-04 国立研究開発法人国立国際医療研究センター Method and kit for detecting onset of diabetic nephropathy or onset risk
EP2848937A1 (en) 2013-09-05 2015-03-18 International Aids Vaccine Initiative Methods of identifying novel HIV-1 immunogens
CN104007258B (en) * 2013-09-30 2016-07-06 中国医学科学院基础医学研究所 The protein marker of focal segmental glomerulosclerosis
US10058604B2 (en) 2013-10-07 2018-08-28 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
WO2015084939A1 (en) 2013-12-03 2015-06-11 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
CN104849466A (en) * 2014-02-14 2015-08-19 张曼 Application of urine vitronectin in diagnosis and treatment of type 2 diabetes mellitus combined early renal injury
WO2016012864A2 (en) * 2014-07-23 2016-01-28 University Of British Columbia Biomarkers for anderson-fabry disease
US10174292B2 (en) 2015-03-20 2019-01-08 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
EP3072901A1 (en) 2015-03-23 2016-09-28 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
WO2016164854A1 (en) 2015-04-09 2016-10-13 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
CA3072427C (en) 2017-08-08 2023-12-05 Fresenius Medical Care Holdings, Inc. Systems and methods for treating and estimating progression of chronic kidney disease
CN107561175A (en) * 2017-08-10 2018-01-09 武汉大学 A kind of evaluation method of glomerular sclerosis rat model
RU2697722C1 (en) * 2018-12-06 2019-08-19 Ирина Сергеевна Шатохина Diagnostic technique of activity of inflammatory process in latent flow of chronic pyelonephritis
CN110632229A (en) * 2019-09-24 2019-12-31 华兰生物工程重庆有限公司 Method for detecting protein content of human immunoglobulin (pH4) infused in static environment by high performance liquid chromatograph
CN111714517A (en) * 2020-04-14 2020-09-29 南开大学 Application of lysosome in field of medicine preparation
RU2768464C1 (en) * 2021-04-27 2022-03-24 Федеральное государственное бюджетное образовательное учреждение высшего образования "Рязанский государственный медицинский университет имени академика И.П. Павлова" Министерства здравоохранения Российской Федерации Method for diagnosing membranous nephropathy as one of forms of chronic glomerulonephritis

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5246835A (en) * 1991-05-24 1993-09-21 Wakamoto Pharmaceutical Co., Ltd. Method of diagnosing renal diseases
US5534431A (en) * 1994-02-28 1996-07-09 The Board Of Regents Of The University Of Nebraska Hybridomas and monoclonal antibodies specific for unique determinants of nephropathy-related immunoglobulin G and complexes thereof
US5908925A (en) * 1996-06-27 1999-06-01 Exocell, Inc. Genetically engineered immunoglobulins with specificity for glycated albumin

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4994165A (en) * 1989-02-16 1991-02-19 Cornell Research Foundation, Inc. Liquid junction coupling for capillary zone electrophoresis/ion spray spectrometry
JPH0682446A (en) * 1992-08-31 1994-03-22 Wakamoto Pharmaceut Co Ltd Diagnostic method for kidney disease
DE4336498A1 (en) * 1993-10-26 1995-04-27 Bender & Co Gmbh Method for diagnosing renal and vascular diseases
US5800998A (en) * 1996-11-12 1998-09-01 Millennium Pharmaceuticals, Inc. Assays for diagnosing type II diabetes in a subject
BR9916407A (en) 1998-12-21 2001-09-25 Univ Monash Detection and treatment of kidney disease

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5246835A (en) * 1991-05-24 1993-09-21 Wakamoto Pharmaceutical Co., Ltd. Method of diagnosing renal diseases
US5534431A (en) * 1994-02-28 1996-07-09 The Board Of Regents Of The University Of Nebraska Hybridomas and monoclonal antibodies specific for unique determinants of nephropathy-related immunoglobulin G and complexes thereof
US5654158A (en) * 1994-02-28 1997-08-05 Board Of Regents Of The University Of Nebraska Methods for detection of nephropathy-related immunoglobulin G using monoclonal antibodies specific for nephropathy-related immunoglobulin G
US5908925A (en) * 1996-06-27 1999-06-01 Exocell, Inc. Genetically engineered immunoglobulins with specificity for glycated albumin

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060062783A1 (en) * 2003-08-08 2006-03-23 Lorin Roskos Antibodies against parathyroid hormone
US20050112123A1 (en) * 2003-10-06 2005-05-26 Vaughan Michael R. Methods of treating proteinuria
US11150250B2 (en) 2008-08-28 2021-10-19 Astute Medical, Inc. Methods for diagnosing acute kidney injury or renal failure
US20110195429A1 (en) * 2008-08-28 2011-08-11 Astute Medical Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110174062A1 (en) * 2008-08-29 2011-07-21 Joseph Anderberg Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9057735B2 (en) 2008-08-29 2015-06-16 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110201038A1 (en) * 2008-10-21 2011-08-18 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110207161A1 (en) * 2008-10-21 2011-08-25 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US11754566B2 (en) 2008-10-21 2023-09-12 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US8778615B2 (en) 2008-10-21 2014-07-15 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10823733B2 (en) 2008-10-21 2020-11-03 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US8993250B2 (en) 2008-11-10 2015-03-31 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20110229915A1 (en) * 2008-11-22 2011-09-22 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9229010B2 (en) 2009-02-06 2016-01-05 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20120264629A1 (en) * 2009-08-07 2012-10-18 Joseph Anderberg Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US8871459B2 (en) * 2009-08-07 2014-10-28 Astute Medical, Inc. Method for evaluating renal status by determining beta-2-glycoprotein 1
US10324093B2 (en) 2009-11-07 2019-06-18 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US11262363B2 (en) 2009-12-20 2022-03-01 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10830773B2 (en) 2009-12-20 2020-11-10 Astute Medical, Inc. Methods for prognosis of future acute renal injury and acute renal failure
US11454635B2 (en) 2010-02-05 2022-09-27 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9029093B2 (en) 2010-02-26 2015-05-12 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10823742B2 (en) 2010-06-23 2020-11-03 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10928403B2 (en) 2010-06-23 2021-02-23 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US11761967B2 (en) 2010-06-23 2023-09-19 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
KR101215759B1 (en) 2010-11-01 2012-12-26 서울대학교산학협력단 Uses of Compositions Comprising Orosomucoid
CN102854325A (en) * 2011-07-01 2013-01-02 复旦大学 Method for detecting affinity indicators of reversible binding of insulin and serum protein and application thereof
US10935548B2 (en) 2011-12-08 2021-03-02 Astute Medical, Inc. Methods for diagnosis and prognosis of renal injury and renal failure using insulin-like growth factor-binding protein 7 and metalloproteinase inhibitor 2
US11099194B2 (en) 2013-01-17 2021-08-24 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9696322B2 (en) 2013-01-17 2017-07-04 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9360488B2 (en) 2013-01-17 2016-06-07 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US11243217B2 (en) 2016-06-06 2022-02-08 Astute Medical, Inc. Management of acute kidney injury using insulin-like growth factor-binding protein 7 and tissue inhibitor of metalloproteinase 2

Also Published As

Publication number Publication date
US20020012906A1 (en) 2002-01-31
AU1674400A (en) 2000-07-12
CA2356174A1 (en) 2000-06-29
BR9916407A (en) 2001-09-25
JP2002533680A (en) 2002-10-08
JP4160263B2 (en) 2008-10-01
US20020022236A1 (en) 2002-02-21
DE69937368T2 (en) 2008-07-17
US6589748B2 (en) 2003-07-08
JP2006038877A (en) 2006-02-09
EP1141728A4 (en) 2005-01-12
MXPA01006404A (en) 2003-06-06
CN1331801A (en) 2002-01-16
EP1141728A1 (en) 2001-10-10
DE69937368D1 (en) 2007-11-29
JP4388515B2 (en) 2009-12-24
ATE376187T1 (en) 2007-11-15
DK1141728T3 (en) 2008-02-04
WO2000037944A1 (en) 2000-06-29
EP1141728B1 (en) 2007-10-17
IL143892A0 (en) 2002-04-21

Similar Documents

Publication Publication Date Title
US6589748B2 (en) Method for kidney disease detection and treatment
US6447989B1 (en) Kidney disease detection and treatment
US20030003588A1 (en) Method for kidney disease detection by protein profiling
EP2310861B1 (en) Healthy kidney biomarkers
EP1628136A1 (en) Method of diagnosis of disease using copeptin
EP1352238A2 (en) Method for detection of vitamin d metabolites
CA2008360A1 (en) Myocardial infarction immunoassay
US20040029175A1 (en) Method for kidney disease detection
US7977072B2 (en) Sandwich immunoassay for identifying partial proANP peptides
JPH06502912A (en) Analyte variant analysis
Arendt et al. Increased endothelin plasma concentrations in patients with coronary artery disease or hyperlipoproteinemia without coronary events
EP1169647B1 (en) Method for analyzing the amount of intraabdominal adipose tissue
Ohta et al. Measurement of intact rat osteocalcin in osteoblast (ROS17/2.8) cells and in ovariectomized rats with a sandwich enzyme immunoassay
Snitkoff et al. Development of an Immunoassay for Monitoring the Levels of Ciprofloxacin Patient Samples
JP3998245B2 (en) Method and kit for measuring oxidized apolipoprotein AI and oxidized lipoprotein containing the same
JP3499875B2 (en) Antibody reagent for detecting dissecting aortic aneurysm and use thereof
ZA200309593B (en) Method for kidney disease detection by protein profiling.
US5593842A (en) Method of measuring thymopoietin proteins in plasma and serum including acidification of the plasma and serum
de Cos et al. Parent cyclosporine in whole blood by monoclonal fluorescence polarization immunoassay for Axsym and monoclonal enzyme-multiplied immunoassay for Cobas-Fara
EP0692715A1 (en) Method of assaying for fructose-1,6-biphosphatase and diagnostic test therefrom
JPH09318627A (en) Screening method for metabolic error of osseous tissue in renal failure or basedow&#39;s disease
JP2007279066A (en) Method and kit for measuring appolipoprotein oxide ai and lipoprotein oxide containing it
JPH0436657A (en) Method for measuring swine elastase in human serum
JPH0833398B2 (en) Method and kit for measuring human tissue plasminogen activator / human plasminogen activator inhibitor complex

Legal Events

Date Code Title Description
AS Assignment

Owner name: NORTHSHORE ASSET MANAGEMENT, LLC, ILLINOIS

Free format text: SECURITY AGREEMENT;ASSIGNOR:AUSAM BIOTECHNOLOGIES, INC.;REEL/FRAME:015074/0827

Effective date: 20031201

AS Assignment

Owner name: TRIDENT GROWTH FUND, LP, TEXAS

Free format text: SECURITY AGREEMENT;ASSIGNOR:AUSAM BIOTECHNOLOGIES, INC.;REEL/FRAME:016182/0255

Effective date: 20050421

STCB Information on status: application discontinuation

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