WO2017040488A1 - Prediction of kidney disease, severity and related treatment approaches - Google Patents

Prediction of kidney disease, severity and related treatment approaches Download PDF

Info

Publication number
WO2017040488A1
WO2017040488A1 PCT/US2016/049421 US2016049421W WO2017040488A1 WO 2017040488 A1 WO2017040488 A1 WO 2017040488A1 US 2016049421 W US2016049421 W US 2016049421W WO 2017040488 A1 WO2017040488 A1 WO 2017040488A1
Authority
WO
WIPO (PCT)
Prior art keywords
supar
subject
egfr
ckd
level
Prior art date
Application number
PCT/US2016/049421
Other languages
French (fr)
Inventor
Jochen Reiser
Sanja Sever
Arshed A. Quyyumi
Original Assignee
Rush University Medical Center
Massachusetts General Hospital
Emory University School Of Medicine
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rush University Medical Center, Massachusetts General Hospital, Emory University School Of Medicine filed Critical Rush University Medical Center
Publication of WO2017040488A1 publication Critical patent/WO2017040488A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/972Plasminogen activators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/34Genitourinary disorders
    • G01N2800/347Renal failures; Glomerular diseases; Tubulointerstitial diseases, e.g. nephritic syndrome, glomerulonephritis; Renovascular diseases, e.g. renal artery occlusion, nephropathy
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/50Determining the risk of developing a disease

Definitions

  • CKD Chronic kidney disease
  • CVD cardiovascular disease
  • eGFR glomerular filtration rate
  • Soluble urokinase-type plasminogen activator receptor is the circulating form of a glycosyl-phosphatidylinositol (GPI)-anchored three-domain membrane protein expressed on a variety of cells including immunologically active cells, endothelial cells and podocytes. 7"9 Both forms are directly involved in the regulation of cell adhesion and migration through binding of integrins. 7
  • SuPAR is produced by cleavage of membrane-bound uPAR, and is readily detected in plasma, serum, urine, and other bodily fluids. 10"12 As a biomarker of immune system activation and inflammation, elevated suPAR levels are
  • suPAR has been implicated in the pathogenesis of kidney disease, specifically focal and segmental glomerulosclerosis (FSGS) and diabetic nephropathy, through interference with podocyte migration and
  • a method of treating a subject at risk of developing a chronic kidney disease includes obtaining a biological sample from the subject where the subject has an estimated glomerular filtration rate (eGFR) of >60 mL/min/1.73m 2 and assaying a soluble urokinase-type
  • eGFR estimated glomerular filtration rate
  • the method also includes determining the suPAR level in the sample relative to a control sample, identifying the subject having an elevated suPAR level relative to the control and stratifying the subject having the elevated suPAR level to a treatment group for treating CKD.
  • a method of identifying a subject at risk of developing chronic kidney disease includes obtaining a biological sample from the subject where the subject has an estimated glomerular filtration rate (eGFR) of >60 mL/min/1 .73m 2 and assaying a level of soluble urokinase-type plasminogen activator receptor (suPAR) in the sample.
  • the method also includes identifying the subject having a suPAR level of about 3000 pg/ml or greater as being at risk for developing CKD.
  • Figure 1 illustrates suPAR levels and decline in eGFR
  • Figures 2A and 2B illustrate suPAR-related decline in eGFR stratified by relevant subgroups.
  • Figure 3 illustrates suPAR levels and incident CKD.
  • the P-value was the test result of log- rank test to compare the survival distributions among the four suPAR quartile groups. The log-rank statistic is approximately a ⁇ 2 with 3 df for large samples.
  • Figure 4 is a flow diagram showing eGFR measurements.
  • Figures 5A-5D illustrate the relationship between suPAR and eGFR at baseline. Bar graphs depicting (A) mean suPAR levels and (B) percentage of subjects with baseline suPAR level >3040 pg/mL stratified according to eGFR categories. Panels C and D are scatterplots showing the relationship between baseline suPAR levels and eGFR stratified according to eGFR lower (C) or higher (D) than 90 mL/min/1.73m2.
  • Figures 6A-6B illustrate the relationship between suPAR and
  • FIGS 7A-7B illustrate the Kaplan Meier curves with eGFR ⁇ 45 mL/min and eGFR ⁇ 30 mL/min as outcomes.
  • the shaded areas represent the 95% confidence intervals.
  • Figure 8 illustrates suPAR levels and decline in eGFR in the WIHS subset.
  • Embodiments of the present invention relate to methods of treating a subject at risk of developing a chronic kidney disease and methods of identifying subjects at risk of developing chronic kidney disease.
  • the methods include determining a suPAR level in a subject having an eGFR of >60 mL/min/1.73m 2 .
  • chronic kidney disease is defined as having an eGFR of ⁇ 60 mL/min/1.73m 2 .
  • Treating means an alleviation of symptoms associated with a disorder or disease, or halt or delay of progression to a chronic disease or worsening of the symptoms associated with disease progression, or prevention or prophylaxis of the disease or disorder.
  • successful treatment may include halt or delay of progression related to a chronic kidney disease.
  • a therapeutically effective amount of a compound is a quantity sufficient to delay, diminish or alleviate at least one symptom associated with the conditions being treated.
  • “Therapeutically effective amount” refers to the quantity of a component which is sufficient to yield a desired therapeutic response without undue adverse side effects (such as toxicity, irritation, or allergic response) commensurate with a reasonable benefit/risk ratio when used in the manner of this invention.
  • Identifying the subjects may include obtaining a biological sample and assaying a suPAR level in the subject.
  • the biological sample may be isolated from the subject by collecting a blood sample, urine sample or other tissue sample from the subject. In a blood sample, plasma or serum may be isolated using any technique known to one skilled in the art.
  • the subject may be stratified to a treatment group if the subject has an elevated suPAR level.
  • the subject may have an eGFR of >60 mL/min/1.73m 2 and an elevated suPAR level and be stratified to a treatment group.
  • subjects may be stratified to a treatment group for optimal blood pressure treatment.
  • the subjects in the blood pressure treatment group may receive treatment in the form of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers (ARBs), diuretics, beta-blockers, calcium channel blockers, alpha-blockers, alpha-agonists, renin inhibitors, combination
  • ACE angiotensin-converting enzyme
  • ARBs angiotensin II receptor blockers
  • beta-blockers calcium channel blockers
  • alpha-blockers alpha-agonists
  • renin inhibitors combination
  • the subjects may be stratified to a clinical trial group.
  • the treatment may include administration to of an agent to the subject at risk of developing a CKD.
  • the agent may be an antibody, aptamer, antisense oligonucleotide, a natural agent, a synthetic agent or combinations thereof.
  • the agent is a chemical compound, natural or synthetic, in particular an organic or inorganic molecule of plant, bacterial, viral, animal, eukaryotic, synthetic or semisynthetic origin, capable of reducing, delaying or inhibiting progression to a chronic kidney disease in a subject having an elevated suPAR level.
  • the treatment may include an oral administration of a compound.
  • the dose of the compound administered to the subject may be in the range from about 500 mg to 2000 mg per day for patients.
  • the dose of the compound to be administered alone or in combination therapy warm-blooded animals, for example humans is preferably from approximately 0.01 mg/kg to approximately 1000 mg/kg, more preferably from approximately 1 mg/kg to approximately 100 mg/kg, per day, divided preferably into 1 to 3 single doses which may, for example, be of the same size.
  • Usually children receive half of the adult dose, and thus the
  • preferential dose range for the inhibitor in children is 0.5 mg/kg to approximately 500 mg/kg, per day, divided preferably into 1 to 3 single doses that may be of the same size.
  • a compound can be administered alone or in combination with another autophagy activators, possible combination therapy taking the form of fixed combinations or the administration of a compound and another inhibitor being staggered or given independently of one another.
  • Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above.
  • Other possible treatments are therapy to maintain the subject's status after symptom amelioration, or even preventive therapy, for example in subjects at risk.
  • Effective amounts of the compounds described herein generally include any amount sufficient to detectably ameliorate one or more symptoms of a neurodegenerative disorder, or by detecting an inhibition or alleviation of symptoms of a kidney disease or disorder.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the specific dose level for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy.
  • the therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.
  • a chronic kidney disease is reduced or prevented in a subject such as a human or lower mammal by stratifying the subject to a treatment group, wherein the treatment may include administering to the subject an amount of an agent, in such amounts and for such time as is necessary to achieve the desired result.
  • the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
  • compositions for administration of the active agent in the method of the invention may be prepared by means known in the art for the preparation of compositions (such as in the art of veterinary and pharmaceutical compositions) including blending, grinding, homogenising, suspending, dissolving, emulsifying, dispersing and where appropriate, mixing of the active agent, together with selected excipients, diluents, carriers and adjuvants.
  • the composition may be in the form of tablets, lozenges, pills, troches, capsules, elixirs, powders, including lyophilised powders, solutions, granules, suspensions, emulsions, syrups and tinctures.
  • Slow-release, or delayed-release, forms may also be prepared, for example in the form of coated particles, multi-layer tablets or microgranules.
  • Solid forms for oral administration may contain binders acceptable in human and veterinary pharmaceutical practice, sweeteners, disintegrating agents, diluents, flavourings, coating agents, preservatives, lubricants and/or time delay agents.
  • Suitable binders include gum acacia, gelatine, corn starch, gum tragacanth, sodium alginate, carboxymethylcellulose or polyethylene glycol.
  • Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine.
  • Suitable disintegrating agents include corn starch, methyl cellulose,
  • Suitable diluents include lactose, sorbitol, mannitol, dextrose, kaolin, cellulose, calcium carbonate, calcium silicate or dicalcium phosphate.
  • Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring.
  • Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten.
  • Suitable preservatives include sodium benzoate, vitamin E, alpha- tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
  • Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc.
  • Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • Suitable liquid carriers include water, oils such as olive oil, peanut oil, sesame oil, sunflower oil, safflower oil, arachis oil, coconut oil, liquid paraffin, ethylene glycol, propylene glycol, polyethylene glycol, ethanol, propanol, isopropanol, glycerol, fatty alcohols, triglycerides or mixtures thereof.
  • Suspensions for oral administration may further include dispersing agents and/or suspending agents.
  • Suitable suspending agents include sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, poly- vinyl-pyrrolidone, sodium alginate or acetyl alcohol.
  • Suitable dispersing agents include lecithin, polyoxyethylene esters of fatty acids such as stearic acid, polyoxyethylene sorbitol mono- or di-oleate, -stearate or -laurate,
  • the emulsions for oral administration may further include one or more emulsifying agents.
  • Suitable emulsifying agents include dispersing agents as exemplified above or natural gums such as guar gum, gum acacia or gum tragacanth.
  • suPAR was then measured using a different assay (human uPAR QuantikineR ELISA kit (R&D, Minneapolis MN) in a separate replication cohort of 347 women (mean age 40, 62% African American, 31 % HIV+, Table 8) with follow-up eGFR (median 26 measurements over at least 10 years). Over five years, subjects with
  • suPAR ⁇ 3000 pg/mL had a 3.0% decline in eGFR (95% CI [1.6%, 4.4%]) compared to only a 0.7% (95% CI [0.6%, 2.0%]) decline in those with
  • Cardiovascular Biobank cohort (3% versus 20% decline over 5 years,
  • SuPAR- mediated kidney injury may be due to several molecular mechanisms.
  • CD40 auto-antibodies have been implicated in modifying the effect of suPAR in FSGS, 37 whereas levels of acid sphingomyelinase-like
  • suPAR may interact with other molecules to induce podocyte dysfunction and mediated progression to CKD in a broad range of conditions.
  • suPAR acts as a marker of underlying low-grade inflammatory processes leading to disease. That said, in our cohort, hs-CRP, a well-accepted marker of chronic inflammation was predictive of cardiovascular events (similarly to suPAR), but not of decline in kidney function, suggesting that role of suPAR in CKD goes beyond inflammatory processes.
  • SuPAR could thus satisfy the need for a long-sought biomarker for the prediction of CKD.
  • Early identification and management of CKD is highly cost-effective and can reduce the risk of progression of CKD and CVD by up to 50%.
  • proteinuria is considered the most sensitive marker for CKD progression in clinical practice, especially when combined with decline in eGFR.
  • diagnostic accuracy and sensitivity of these parameters for detection of CKD in the population While multiple biomarkers are currently being explored as predictors of kidney disease progression and associated risk of accelerated CVD, none have been demonstrated to predict the incidence of CKD in patients with normal eGFR.
  • Our results suggest that suPAR meets critical requirements for a CKD biomarker.
  • suPAR levels are relatively stable in plasma, 33 and elevated levels are predictive of a heightened risk of developing CKD before there is any decline in eGFR.
  • it adds prognostic value in all patients and in the subgroups of patients who have diabetes or hypertension, the two most prevalent diseases associated with CKD in the U.S.
  • it is predictive of CKD in Caucasians and African Americans, despite the marked differences in likelihood of CKD in these two racial groups.
  • measurement of suPAR may allow for more accurate stratification of patients early in their disease course enabling rational targeting of preventive health resources and enrollment into potential trials of novel renoprotective therapies.
  • suPAR may be a common mediator of both CKD and CVD, conditions that often co-exist and are associated with poor prognosis and outcome.
  • Elevated plasma suPAR levels are associated with incident CKD and a more rapid decline in eGFR in subjects with normal baseline kidney function. Measurement of plasma suPAR level may be a practical and cost-effective tool for identification of adults at higher risk of developing CKD early in the course of disease.
  • Study subjects were recruited from the Emory Cardiovascular Biobank, a prospective registry of patients undergoing cardiac catheterization at three Emory Healthcare sites in Atlanta, GA between 2003 and 2009. 17 Subjects with congenital heart disease, severe valvular heart disease, severe anemia, recent blood transfusion, myocarditis, or history of active inflammatory disease and cancer were excluded. Subjects aged 20 to 90 years were interviewed to collect information on demographic characteristics, medical history, medication use, and behavioral habits. Coronary artery disease was defined as the presence of an obstructive lesion (>50%) on coronary angiogram. Cardiovascular and kidney risk-factor prevalence was determined by physician diagnosis and treatment for hypertension, hyperlipidemia, and diabetes. Medical records were reviewed to confirm self-reported medical history. The study was approved by the Institutional Review Board at Emory University (Atlanta, GA). All subjects provided written informed consent at the time of enrollment.
  • SuPAR and baseline parameters of kidney function Independent determinants of baseline suPAR and eGFR were determined using linear regression after adjustment for age, gender, race (African American vs. others), body mass index (BMI), history of smoking, diabetes, proteinuria (>1 + vs.
  • renin- angiotensin system RAS
  • SuPAR and change in eGFR at follow-up The association between baseline suPAR levels and change in eGFR over time was investigated in 2292 subjects with follow-up eGFR measurements using linear mixed effects modeling with random subject-specific intercept and random time effect, by regressing log(eGFR) on log(suPAR), follow-up time (years since baseline), log(suPAR) * time, baseline eGFR, in addition to the aforementioned covariates. Three-way interaction terms were incorporated into the model to determine whether eGFR, race, and diabetes modified the association between suPAR and change in eGFR. Estimated decline in eGFR in each subgroup was derived accordingly.
  • SuPAR and incident CKD In 1335 participants with eGFR > 60 mL/min/1 .73 m 2 at baseline, we compared the progression to clinical CKD (eGFR ⁇ 60 mL/min/1 .73 m 2 ) among the suPAR quartile groups using the log-rank test and a Cox proportional hazard model adjusting for the aforementioned
  • the Women's Interagency HIV Study is a longitudinal study of HIV infected and uninfected at-risk women that enrolled 2054 HIV-infected women and 569 uninfected women at six sites, Chicago, San Francisco Bay Area, Brooklyn and Bronx/Manhattan, New York, Washington, DC and Los Angeles from October 1994 through November 1995. From October 2001 through September 2002, an additional 737 HIV-infected women and 406 uninfected women were enrolled. Informed consent was obtained from all participants in accordance with the US Department of Health and Human Services (DHHS) guidelines and the
  • Fuhrman B The urokinase system in the pathogenesis of atherosclerosis. Atherosclerosis 2012;222:8-14.
  • plasminogen activator receptor level is an independent predictor of the presence and severity of coronary artery disease and of future adverse events. J Am Heart Assoc 2014;3:e001 1 18. 18. Backes Y, van der Sluijs KF, Mackie DP, et al. Usefulness of suPAR as a biological marker in patients with systemic inflammation or infection: a systematic review. Intensive Care Med 2012;38:1418-28.
  • de Bock CE Wang Y. Clinical significance of urokinase-type plasminogen activator receptor (uPAR) expression in cancer. Med Res Rev 2004;24:13-39.
  • phosphodiesterase 3b expression levels determine podocyte injury phenotypes in glomerular disease. Journal of the American Society of Nephrology : JASN 2015;26:133-47.
  • Kidney international a review.
  • Soluble urokinase receptor is a biomarker of cardiovascular disease in chronic kidney disease. Kidney
  • Renin-angiotensin system antagonists n 2046 (63%) 998 (62%) 1048 (64%) 0.09
  • Coronary artery disease is defined as the presence of a >50% obstructive lesion in any of the major vessels on coronary angiogram.
  • P value denotes differences between suPAR >3040 and ⁇ 3040 pg/mL.
  • CI confidence interval
  • eGFR estimated glomerular filtration rate.
  • HR hazard ratio
  • hs-CRP high sensitivity c-reactive protein
  • RAS renin- angiotensin system
  • suPAR soluble urokinase-type plasminogen activator receptor.
  • all variables were dichotomous (e.g., presence vs. abscense).
  • the effect of suPAR is reported as a continuous variable, dichotomized by median (3040 pg/mL) as well as by quartiles. Subjects with a suPAR level in the third (Q3) and fourth (Q4) quartiles had a 2.0- and 3.13-fold increased hazard of incident CKD compared to those in the lowest quartile (Ql).
  • Hyperlipidemia -6.4 (-9.1,-3.6) O.001 -2.1 (-5.0,0.9) 0.17
  • Coronary Artery Disease 1.2 (-1.9,4.4) 0.45 2.9 (-0.3,6.3) 0.08
  • O.001 hsCRP high sensitivity C-reactive protein
  • eGFR estimated glomerular filtration rate
  • RAS renin-angiotensin system
  • suPAR soluble urokinase plasminogen-type activator receptor.
  • eGFR estimated glomerular filtration rate.
  • Model 0 includes baseline eGFR, age, gender, race (African American vs. others), body mass index (BMI), history of smoking, diabetes, proteinuria (>1+ vs. negative or trace protein on dipstick), hypertension, hyperlipidemia, history of myocardial infarction, hsCRP, and use of renin-angiontensin system (RAS) inhibitors.
  • Model 1 includes all variables in Model 0 plus suPAR as a continuous variable.
  • Model 2 include all variables in Model 0 plus suPAR quartile groups.
  • CI confidence interval
  • NRI net reclassification index. The change in C- statistic as well as the NRI shows the addition of suPAR to the model has improved predictive accuracy of the model.
  • Adjusted R-square, R-square, and increase in R-square of the linear regression models are shown with log(eGFR) as the response variable using baseline data. Adjusted R-square and R-square values are reported for the model with the variable on the same row and all the variables above tha row. Increase in R-square is the change in R-square for adding the variable to the model, compared to the model using all variables above that row. Note that changing the order of the variable will affect the change in R-square. Note that the increase in R 2 with the addition of suPAR to the model is by far the largest compared to all other variables.
  • RAS Antagonists 0.77 (0.53, 1.1 1) 0.16 0.93 (0.64,1.35) 0.70 hsCRP, per unit increase 0.98 (0.95, 1.01) 0.22 1.00 (0.98,1.01) 0.50 suPAR
  • CI confidence interval
  • eGFR estimated glomerular filtration rate.
  • HR hazard ratio
  • hsCRP high sensitivity c-reactive protein
  • RAS renin-angiotensin system
  • suPAR soluble urokinase plasminogen activator receptor. Note that in both validation cohorts, suPAR was independently predictive of incident CKD, with both 3 and 4* having significantly higher hazard ratios compared to the 1 st quartile
  • suPAR pg/mL 2079 (1740, 2753) 4310 (3616, 5529) ⁇ 0.001
  • HIV viral load per 100%

Landscapes

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

Abstract

Methods of treatment for subjects at risk of developing a chronic kidney disease and methods of identifying subjects at risk of developing a chronic kidney disease are provided.

Description

PREDICTION OF KIDNEY DISEASE, SEVERITY AND RELATED
TREATMENT APPROACHES
RELATED APPLICATIONS
[0001] This application claims the benefit of US Provisional Application No. 62/212,370, filed August 31 , 2015 which is incorporated by reference herein in their entirety.
FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This invention was made with Government support under Grant No. RO1 DK101350, awarded by National Institutes of Health. The Government has certain rights in the invention.
BACKGROUND
1. Technical Field
[0003] Methods of treating subjects at risk of developing a chronic kidney disease and methods of identifying subjects at risk of developing a chronic kidney disease are provided.
2. Background
[0004] Chronic kidney disease (CKD) and progressive loss of kidney function constitute a major public health problem affecting 1 1 % of the US population.1 The three most common diseases associated with CKD are diabetes mellitus, hypertension, and glomerulonephritis.2 Patients with CKD are at high risk of cardiovascular disease (CVD) and mortality.3 It is thus important to identify patients at high risk for CKD and treat underlying disease processes that drive kidney injury.4 In clinical practice, methods of screening for kidney disease are limited to measurement of proteinuria, and estimation of glomerular filtration rate (eGFR). Proteinuria and decline in eGFR are relative insensitive indices of early injury and have limited utility in mass screening for CKD.4 Hence, more sensitive biomarkers are required to identify patients at risk earlier in the disease process, which is essential to design and study interventions aimed at preventing the progression to CKD. [0005] Soluble urokinase-type plasminogen activator receptor (suPAR) is the circulating form of a glycosyl-phosphatidylinositol (GPI)-anchored three-domain membrane protein expressed on a variety of cells including immunologically active cells, endothelial cells and podocytes.7"9 Both forms are directly involved in the regulation of cell adhesion and migration through binding of integrins.7
SuPAR is produced by cleavage of membrane-bound uPAR, and is readily detected in plasma, serum, urine, and other bodily fluids.10"12 As a biomarker of immune system activation and inflammation, elevated suPAR levels are
associated with poor outcomes in the general population including those with CVD,13"17 infectious conditions,18 cancer,19 and diabetes.20,21
[0006] More recently, suPAR has been implicated in the pathogenesis of kidney disease, specifically focal and segmental glomerulosclerosis (FSGS) and diabetic nephropathy, through interference with podocyte migration and
apoptosis.8'20'22'23 These findings suggest a broader role for suPAR as a mediator of kidney injury beyond being a biomarker for a specific glomerular disease entity. Therefore, in a large prospective cohort of patients with CVD, the hypothesis that plasma suPAR levels predict risk of developing new-onset CKD was tested.
BRIEF SUMMARY
[0007] A method of treating a subject at risk of developing a chronic kidney disease (CKD) is provided. The method includes obtaining a biological sample from the subject where the subject has an estimated glomerular filtration rate (eGFR) of >60 mL/min/1.73m2 and assaying a soluble urokinase-type
plasminogen activator receptor (suPAR) level in the sample. The method also includes determining the suPAR level in the sample relative to a control sample, identifying the subject having an elevated suPAR level relative to the control and stratifying the subject having the elevated suPAR level to a treatment group for treating CKD.
[0008] A method of identifying a subject at risk of developing chronic kidney disease (CKD) is provided. The method includes obtaining a biological sample from the subject where the subject has an estimated glomerular filtration rate (eGFR) of >60 mL/min/1 .73m2 and assaying a level of soluble urokinase-type plasminogen activator receptor (suPAR) in the sample. The method also includes identifying the subject having a suPAR level of about 3000 pg/ml or greater as being at risk for developing CKD.
BRIEF DESCRIPTION OF THE DRAWINGS
[0009] Figure 1 illustrates suPAR levels and decline in eGFR
[0010] Percent change in estimated glomerular filtration rate (eGFR) over time stratified by suPAR quartiles in 2292 subjects. Q4 and Q3 had significantly higher rates of decline compared to Q1 and Q2 (P<0.001 , P=0.0002 respectively). There were no significant differences in rate of decline between Q1 and Q2 (P=0.91 ).
The statistical significance for the difference between Q3 and Q4 was marginal
(P=0.08).
[0011] Figures 2A and 2B illustrate suPAR-related decline in eGFR stratified by relevant subgroups.
[0012] Figure 2(A) SuPAR-related percent change in eGFR per year during follow-up stratified by diabetes status, African American race, and baseline of eGFR <60 or >60 mL/min/1 .73 m2. P-values reflect the results of the interaction analysis. Figure 2(B) SuPAR-related percent decline in eGFR stratified by baseline eGFR. Note that subjects with eGFR within the normal range (90-120) had the highest suPAR-related percent eGFR decline.
[0013] Figure 3 illustrates suPAR levels and incident CKD.
[0014] Kaplan-Meier curves demonstrating cumulative incidence of CKD, defined as eGFR<60 mL/min/1 .73 rri2, for subjects with baseline eGFR>60 mL/min/1 .73 rri2 (N=1331 ) stratified by suPAR level quartiles. The shaded areas represent the 95% confidence intervals. The P-value was the test result of log- rank test to compare the survival distributions among the four suPAR quartile groups. The log-rank statistic is approximately a χ2 with 3 df for large samples.
[0015] Figure 4 is a flow diagram showing eGFR measurements. [0016] Figures 5A-5D illustrate the relationship between suPAR and eGFR at baseline. Bar graphs depicting (A) mean suPAR levels and (B) percentage of subjects with baseline suPAR level >3040 pg/mL stratified according to eGFR categories. Panels C and D are scatterplots showing the relationship between baseline suPAR levels and eGFR stratified according to eGFR lower (C) or higher (D) than 90 mL/min/1.73m2. The correlation between suPAR and eGFR was strongest in those with eGFR <90 mL/min/1.73 m2 (r=-0.47, P<0.001 ), and weaker for those with eGFR≥90 mL/min/1.73 m2 (r=-0.07, P=0.041 ) with a P- value for the interaction of <0.001.
[0017] Figures 6A-6B illustrate the relationship between suPAR and
proteinuria at baseline. Bar graphs depicting (A) mean suPAR levels stratified by semi-quantitative proteinuria grade and (B) percentage of subjects with baseline suPAR level >3040 pg/mL stratified according to proteinuria grade. Note the positive correlation between suPAR and proteinuria (r=0.22, P<0.001 ). Nearly half of the subjects without proteinuria had suPAR levels above 3040 pg/mL.
[0018] Figures 7A-7B illustrate the Kaplan Meier curves with eGFR<45 mL/min and eGFR<30 mL/min as outcomes. Kaplan-Meier curves demonstrating cumulative incidence of CKD, defined as (A) eGFR<45 mL/min/1.73 m2, or (B) eGFR<30 mL/min/1.73 m2, for subjects with baseline eGFR≥60 mL/min/1.73 m2 (N=1331 ) stratified by suPAR level quartiles. The shaded areas represent the 95% confidence intervals.
[0019] Figure 8 illustrates suPAR levels and decline in eGFR in the WIHS subset. Percent change in estimated glomerular filtration rate (eGFR) over time stratified by median plasma suPAR (3000 pg/mL) in 347 subjects. Over five years, subjects with suPAR≥3000 pg/mL (N=175) had a 3.0% decline in eGFR (95% CI [1.6%, 4.4%]), compared to only a 0.7% (95% CI [0.6%, 2.0%]) decline in those with suPAR<3000 (N=172).
DETAILED DESCRIPTION
[0020] The embodiments disclosed below are not intended to be exhaustive or to limit the scope of the disclosure to the precise form in the following description. Rather, the embodiments are chosen and described as examples so that others skilled in the art may utilize its teachings.
[0021] Embodiments of the present invention relate to methods of treating a subject at risk of developing a chronic kidney disease and methods of identifying subjects at risk of developing chronic kidney disease. In some embodiments, the methods include determining a suPAR level in a subject having an eGFR of >60 mL/min/1.73m2.
[0022] As used herein, chronic kidney disease (CKD) is defined as having an eGFR of <60 mL/min/1.73m2.
[0023] "Treating", "treat", or "treatment" within the context of the instant invention, means an alleviation of symptoms associated with a disorder or disease, or halt or delay of progression to a chronic disease or worsening of the symptoms associated with disease progression, or prevention or prophylaxis of the disease or disorder. In some embodiments, successful treatment may include halt or delay of progression related to a chronic kidney disease. Likewise, a therapeutically effective amount of a compound is a quantity sufficient to delay, diminish or alleviate at least one symptom associated with the conditions being treated. "Therapeutically effective amount" refers to the quantity of a component which is sufficient to yield a desired therapeutic response without undue adverse side effects (such as toxicity, irritation, or allergic response) commensurate with a reasonable benefit/risk ratio when used in the manner of this invention.
[0024] Methods of treatment of a subject at risk for developing a CKD are provided. Identifying the subjects may include obtaining a biological sample and assaying a suPAR level in the subject. In some embodiments, the biological sample may be isolated from the subject by collecting a blood sample, urine sample or other tissue sample from the subject. In a blood sample, plasma or serum may be isolated using any technique known to one skilled in the art. In some embodiments, the subject may be stratified to a treatment group if the subject has an elevated suPAR level. In some embodiments, the subject may have an eGFR of >60 mL/min/1.73m2 and an elevated suPAR level and be stratified to a treatment group. In some embodiment, subjects may be stratified to a treatment group for optimal blood pressure treatment. In some embodiments, the subjects in the blood pressure treatment group may receive treatment in the form of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers (ARBs), diuretics, beta-blockers, calcium channel blockers, alpha-blockers, alpha-agonists, renin inhibitors, combination
medications and other therapies for blood pressure. In some embodiments, the subjects may be stratified to a clinical trial group.
[0025] In some embodiments, the treatment may include administration to of an agent to the subject at risk of developing a CKD. In some embodiments, the agent may be an antibody, aptamer, antisense oligonucleotide, a natural agent, a synthetic agent or combinations thereof. In some embodiments, the agent is a chemical compound, natural or synthetic, in particular an organic or inorganic molecule of plant, bacterial, viral, animal, eukaryotic, synthetic or semisynthetic origin, capable of reducing, delaying or inhibiting progression to a chronic kidney disease in a subject having an elevated suPAR level.
[0026] In some embodiments, the treatment may include an oral administration of a compound. In some embodiments, the dose of the compound administered to the subject may be in the range from about 500 mg to 2000 mg per day for patients. In some embodiments, the dose of the compound to be administered alone or in combination therapy warm-blooded animals, for example humans, is preferably from approximately 0.01 mg/kg to approximately 1000 mg/kg, more preferably from approximately 1 mg/kg to approximately 100 mg/kg, per day, divided preferably into 1 to 3 single doses which may, for example, be of the same size. Usually children receive half of the adult dose, and thus the
preferential dose range for the inhibitor in children is 0.5 mg/kg to approximately 500 mg/kg, per day, divided preferably into 1 to 3 single doses that may be of the same size.
[0027] A compound can be administered alone or in combination with another autophagy activators, possible combination therapy taking the form of fixed combinations or the administration of a compound and another inhibitor being staggered or given independently of one another. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the subject's status after symptom amelioration, or even preventive therapy, for example in subjects at risk.
[0028] Effective amounts of the compounds described herein generally include any amount sufficient to detectably ameliorate one or more symptoms of a neurodegenerative disorder, or by detecting an inhibition or alleviation of symptoms of a kidney disease or disorder. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
It will be understood, however, that the specific dose level for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination, and the severity of the particular disease undergoing therapy. The therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.
[0029] According to the methods of treatment of the present invention, a chronic kidney disease is reduced or prevented in a subject such as a human or lower mammal by stratifying the subject to a treatment group, wherein the treatment may include administering to the subject an amount of an agent, in such amounts and for such time as is necessary to achieve the desired result.
[0030] It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
[0031] Compositions for administration of the active agent in the method of the invention may be prepared by means known in the art for the preparation of compositions (such as in the art of veterinary and pharmaceutical compositions) including blending, grinding, homogenising, suspending, dissolving, emulsifying, dispersing and where appropriate, mixing of the active agent, together with selected excipients, diluents, carriers and adjuvants.
[0032] For oral administration, the composition may be in the form of tablets, lozenges, pills, troches, capsules, elixirs, powders, including lyophilised powders, solutions, granules, suspensions, emulsions, syrups and tinctures. Slow-release, or delayed-release, forms may also be prepared, for example in the form of coated particles, multi-layer tablets or microgranules.
[0033] Solid forms for oral administration may contain binders acceptable in human and veterinary pharmaceutical practice, sweeteners, disintegrating agents, diluents, flavourings, coating agents, preservatives, lubricants and/or time delay agents. Suitable binders include gum acacia, gelatine, corn starch, gum tragacanth, sodium alginate, carboxymethylcellulose or polyethylene glycol.
Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine. Suitable disintegrating agents include corn starch, methyl cellulose,
polyvinylpyrrolidone, guar gum, xanthan gum, bentonite, alginic acid or agar. Suitable diluents include lactose, sorbitol, mannitol, dextrose, kaolin, cellulose, calcium carbonate, calcium silicate or dicalcium phosphate. Suitable flavouring agents include peppermint oil, oil of wintergreen, cherry, orange or raspberry flavouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zein, shellac or gluten. Suitable preservatives include sodium benzoate, vitamin E, alpha- tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleate, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate. [0034] Liquid forms for oral administration may contain, in addition to the above agents, a liquid carrier. Suitable liquid carriers include water, oils such as olive oil, peanut oil, sesame oil, sunflower oil, safflower oil, arachis oil, coconut oil, liquid paraffin, ethylene glycol, propylene glycol, polyethylene glycol, ethanol, propanol, isopropanol, glycerol, fatty alcohols, triglycerides or mixtures thereof.
[0035] Suspensions for oral administration may further include dispersing agents and/or suspending agents. Suitable suspending agents include sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, poly- vinyl-pyrrolidone, sodium alginate or acetyl alcohol. Suitable dispersing agents include lecithin, polyoxyethylene esters of fatty acids such as stearic acid, polyoxyethylene sorbitol mono- or di-oleate, -stearate or -laurate,
polyoxyethylene sorbitan mono- or di-oleate, -stearate or -laurate and the like.
[0036] The emulsions for oral administration may further include one or more emulsifying agents. Suitable emulsifying agents include dispersing agents as exemplified above or natural gums such as guar gum, gum acacia or gum tragacanth.
[0037] Examples
[0038] Emory Cardiovascular Biobank cohort characteristics at baseline
[0039] Subject demographic and clinical characteristics in the total cohort and dichotomized according to median suPAR level (3040 pg/mL (2373, 4019 [IQR]) are shown in Table 1. Elevated plasma suPAR level were independently associated with gender, a history of smoking, hypertension, hyperlipidemia, diabetes mellitus, myocardial infarction, hs-CRP levels, proteinuria and eGFR (Table 3).
[0040] Relationship between, eGFR, proteinuria, and suPAR at baseline.
[0041] After multivariable regression analysis adjusting for the aforementioned covariates, a lower eGFR at baseline was independently associated with increasing age, male gender, African American race, higher BMI, hypertension, smoking, proteinuria, lack of RAS inhibitors, and suPAR levels (Table S3, Figure
5). The correlation between suPAR and eGFR varied depending on baseline eGFR (P-value<0.001 for interaction), and was weak in subjects with eGFR >90 mL/min/1.73 m2 (r=-0.07, P=0.041 , Figure 5 Panels C and D). Note that more than 30% of subjects with eGFR≥90/min/1.73 m2 had suPAR>3040 pg/mL (Figure 5, Panel B). In addition, we found a positive correlation between proteinuria and suPAR levels (r=0.22, P<0.001 ) that was independent of the aforementioned covariates including eGFR (Table 3, Figure 6). Of note, over half of the subjects without proteinuria (52%) had suPAR levels above 3040 pg/mL (Figure 6, Panel B)
[0042] Relationship between suPAR levels and change in eGFR during follow- up.
[0043] The relationship between the plasma suPAR level at baseline and the change in eGFR in 2292 (62%) subjects with follow-up eGFR measurements was determined. The decline in eGFR was greater in subjects with higher suPAR levels (Table 4, Figure 1 ). Subjects in the two higher quartiles of suPAR levels
(>3040 pg/mL) had a significantly greater decline in eGFR compared to the lower two quartiles (<3040 pg/mL) after adjusting for age, gender, race, the use of RAS inhibitors, and serum hs-CRP (Figure 1A). Thus, over 5-years, the decline in eGFR in those in the lower two quartiles was 7.3% (95% CI [5.0%, 9.5%]) whereas it was 14.5% (95% CI [10.7%, 18.2%]) and 20.4% (95% CI [12.9%,
27.3%]) in those in the third and fourth highest quartiles, respectively (Figure 1 ).
[0044] Sensitivity analyses were performed to test whether race, presence of diabetes, proteinuria, baseline eGFR or baseline hs-CRP values influenced the relationship between the suPAR level and change in eGFR. SuPAR remained an independent predictor of eGFR decline regardless of race or presence of diabetes (Figure 2A). However, the interaction with baseline eGFR was
significant (P<0.001 ): there was no relationship between suPAR levels and eGFR decline in subjects with baseline eGFR<60 mL/min/1.73 m2, whereas the suPAR level was predictive of eGFR change in those with eGFR>60 mL/min/1.73 m2 at baseline (Figure 2A). Importantly, subjects with normal eGFR>90 mL/min/1.73 m2
(n=921 ) at baseline had the largest suPAR-related eGFR decline (P<0.001 for interaction, Figure 2B). We did not find a significant interaction with baseline proteinuria (P=0.29) nor baseline serum hs-CRP levels (P=0.89) [0045] Relationship between plasma suPAR levels and incident CKD
[0046] In 1335 participants with eGFR≥60 mL/min/1.73 m2 at baseline, we determined whether baseline suPAR level predicted progression to clinical CKD (defined as eGFR<60 mL/min/1.73 m2). A total of 320 (24%) subjects developed CKD during follow-up. Independent predictors of incident CKD were age, baseline eGFR, hypertension, and plasma suPAR level (Table 2). Higher baseline suPAR level was associated with a significantly greater incidence of CKD (P<0.001 ) (Figure 3). Subjects with a suPAR level in the third and fourth quartiles had a 2.0- and 3.13-fold increased hazard of incident CKD compared to those in the lowest quartile (Table 2). Thus, at 1 year and 5 years, 7% and 41 % of subjects with suPAR>3040 ng/mL (third and fourth quartiles) developed CKD respectively, compared to 1 % and 12% of those with suPAR<3040 ng/mL (1 st and second quartiles) at the respective follow-up times. Even after excluding subjects with potential acute kidney injury (N=208), defined as a 50% decline in eGFR compared to the prior eGFR measurement regardless of time we found both the 3rd (OR 2.20 95%CI [1.33-3.06) and 4th highest quartiles (OR 2.93 95%CI [1.86-4.62]) of suPAR remained predictive of incident CKD compared to the lowest quartile, suggesting that suPAR is associated with a progressive decline in kidney function.
[0047] Risk Discrimination: The incremental value of adding suPAR to a model with traditional risk factors in predicting incident CKD was tested. The C-statistic for incident CKD at both 1 and 5 years of follow-up increased with the addition of suPAR quartile groups (W=0.08, 95% CI [0.01 , 0.15] and W=0.03, 95% CI [0.01 ,
0.06]), respectively (Table 5). The continuous NRI metric shows significant reclassification of subject risk by addition of suPAR to the model at both 1 (0.40,
95%CI [0.15, 0.53]) and 5 years (0.34, 95%CI [0.19, 0.41]) (Table 5). Addition of suPAR to the model with conventional risk factors was associated with a change in R2 of 0.33 that was larger than all other variables (Table 6). Validation analyses: We repeated the longitudinal analyses in (1 ) two randomly selected subsets of the Emory Cardiovascular Biobank cohort and (2) in a subset of subjects enrolled in the WIHS cohort. In both randomly selected subsets of the Emory Cardiovascular Biobank, elevated baseline suPAR level remained an independent predictor of eGFR decline during follow-up and of incident CKD, findings consistent with the primary analysis (Tables 4, 5). suPAR was then measured using a different assay (human uPAR QuantikineR ELISA kit (R&D, Minneapolis MN) in a separate replication cohort of 347 women (mean age 40, 62% African American, 31 % HIV+, Table 8) with follow-up eGFR (median 26 measurements over at least 10 years). Over five years, subjects with
suPAR≥3000 pg/mL (N=175) had a 3.0% decline in eGFR (95% CI [1.6%, 4.4%]) compared to only a 0.7% (95% CI [0.6%, 2.0%]) decline in those with
suPAR<3000 (ISM 72) (Table 9, Figure 8).
[0048] DISCUSSION
[0049] In this prospective cohort of adults with CVD, an association between elevated plasma suPAR levels and both the decline in eGFR and the
development of new-onset CKD has been identified. This relationship was observed in patients with normal baseline kidney function, and was independent of conventional kidney and cardiovascular disease risk factors, including baseline eGFR, age, race, diabetes, hypertension, and hs-CRP levels. Moreover, the lack of association between hs-CRP and eGFR decline or incident CKD suggests that the effect of suPAR on kidney dysfunction is unlikely to be the result of acute inflammatory processes. Importantly, suPAR significantly improved discrimination of future risk of CKD over a standard clinical model as evidenced by significant improvement in the C-statistic and NRI. This risk re-classification is greater than well-established biomarkers such as CRP and BNP that are used to predict cardiovascular events and heart failure, respectively.30 We also show that suPAR remains associated with renal function decline in younger subjects with a significantly lower CVD risk factor burden, suggesting the effect of suPAR is truly independent of traditional CVD/CKD risk factors. The estimate in the WIHS subset was unsurprisingly lower than the one observed in the Emory
Cardiovascular Biobank cohort (3% versus 20% decline over 5 years,
respectively), likely due to the older age and greater burden of CVD/CKD risk factors in the Biobank cohort. [0050] High suPAR levels have typically been attributed to a state of inflammation or decreased renal clearance.18'38 While we found a negative correlation between plasma suPAR and the baseline eGFR levels, this
association was weak at baseline eGFR>90 mL/min/1.73 m2, with a substantial proportion (>30%) of subjects with normal kidney function having suPAR levels >3000 pg/mL in the absence of sepsis or cancer. This suggests that elevated suPAR levels are unlikely to simply result from a decrease in renal clearance, and may be related to the underlying pathogenic mechanisms initiating kidney disease.
[0051] Evidence of suPAR's pathogenic role in kidney disease has emerged mainly from studies of FSGS, where suPAR was found to activate ανβ3 integrin on podocytes, leading to effacement of foot processes and proteinuria.22
Elevated suPAR levels have also been associated with diabetic nephropathy and with progression of lupus nephritis.29'21 In animal models of diabetic kidney disease, blockade of β3 integrin using a monoclonal antibody was
protective.21'35'36 In addition to activation of ανβ3 integrin on podocytes, SuPAR- mediated kidney injury may be due to several molecular mechanisms. For example, CD40 auto-antibodies have been implicated in modifying the effect of suPAR in FSGS,37 whereas levels of acid sphingomyelinase-like
phosphodiesterase 3b have been implicated in modulating the effect of suPAR in diabetic nephropathy.20 Thus, suPAR may interact with other molecules to induce podocyte dysfunction and mediated progression to CKD in a broad range of conditions. Alternatively, given the stability of the suPAR protein both in vivo and in vitro,33 it is possible that suPAR acts as a marker of underlying low-grade inflammatory processes leading to disease. That said, in our cohort, hs-CRP, a well-accepted marker of chronic inflammation was predictive of cardiovascular events (similarly to suPAR), but not of decline in kidney function, suggesting that role of suPAR in CKD goes beyond inflammatory processes. SuPAR could thus satisfy the need for a long-sought biomarker for the prediction of CKD. Early identification and management of CKD is highly cost-effective and can reduce the risk of progression of CKD and CVD by up to 50%.31 Currently, proteinuria is considered the most sensitive marker for CKD progression in clinical practice, especially when combined with decline in eGFR. However, there are significant shortfalls in the diagnostic accuracy and sensitivity of these parameters for detection of CKD in the population. While multiple biomarkers are currently being explored as predictors of kidney disease progression and associated risk of accelerated CVD, none have been demonstrated to predict the incidence of CKD in patients with normal eGFR.32 Our results suggest that suPAR meets critical requirements for a CKD biomarker. First, suPAR levels are relatively stable in plasma,33 and elevated levels are predictive of a heightened risk of developing CKD before there is any decline in eGFR. Second, it adds prognostic value in all patients and in the subgroups of patients who have diabetes or hypertension, the two most prevalent diseases associated with CKD in the U.S. Third, it is predictive of CKD in Caucasians and African Americans, despite the marked differences in likelihood of CKD in these two racial groups. Finally, measurement of suPAR may allow for more accurate stratification of patients early in their disease course enabling rational targeting of preventive health resources and enrollment into potential trials of novel renoprotective therapies. Importantly, we and others have shown that elevated suPAR levels are associated with increased risk of adverse cardiovascular events in subjects with or without mild to moderate CKD.17'34 Thus, suPAR may be a common mediator of both CKD and CVD, conditions that often co-exist and are associated with poor prognosis and outcome.
[0052] Elevated plasma suPAR levels are associated with incident CKD and a more rapid decline in eGFR in subjects with normal baseline kidney function. Measurement of plasma suPAR level may be a practical and cost-effective tool for identification of adults at higher risk of developing CKD early in the course of disease.
[0053] METHODS
[0054] Study population.
[0055] Study subjects were recruited from the Emory Cardiovascular Biobank, a prospective registry of patients undergoing cardiac catheterization at three Emory Healthcare sites in Atlanta, GA between 2003 and 2009.17 Subjects with congenital heart disease, severe valvular heart disease, severe anemia, recent blood transfusion, myocarditis, or history of active inflammatory disease and cancer were excluded. Subjects aged 20 to 90 years were interviewed to collect information on demographic characteristics, medical history, medication use, and behavioral habits. Coronary artery disease was defined as the presence of an obstructive lesion (>50%) on coronary angiogram. Cardiovascular and kidney risk-factor prevalence was determined by physician diagnosis and treatment for hypertension, hyperlipidemia, and diabetes. Medical records were reviewed to confirm self-reported medical history. The study was approved by the Institutional Review Board at Emory University (Atlanta, GA). All subjects provided written informed consent at the time of enrollment.
[0056] Study design.
[0057] The relationship between baseline suPAR levels and kidney function (eGFR and semi-quantitative assessment of proteinuria) was examined in 3683 subjects. To investigate the relationship between suPAR levels and change in eGFR during follow-up, 2292 (62%) subjects had at least one additional measure of eGFR (median number of measurements=7) during a median follow-up period of 1337 days. Finally, the relationship between suPAR levels and the
development of incident CKD, defined as eGFR<60 mL/min/1.73m2 beyond 30 days, was determined in 1335 subjects with a baseline eGFR>60 mL/min/1.73rri2 (Figure S1 ).24
[0058] Sample collection and measurement of suPAR and hs-CRP.
[0059] Fasting arterial blood samples were collected and serum and plasma stored at -80°C for a mean duration of 4.9 years. Serum high sensitivity CRP (hs- CRP) concentrations were determined using a particle-enhanced
immunoturbidimetry assay with a lower detection limit of 0.03 mg/L (FirstMark, Division of GenWay Biotech Inc, San Diego, CA).25 Plasma levels of suPAR were measured by Virogates (suPARnostic kit; Copenhagen, Denmark) with a lower detection limit of 100 pg/mL and intra- and inter-assay variation of 2.75% and
9.17%, respectively as determined by the assay manufacturer. [0060] Parameters of kidney function.
[0061] Serum creatinine measurements at enrollment and all subsequent values acquired during routine follow-up clinic visits or hospitalizations within the Emory Healthcare system were collected. eGFR was calculated using the CKD- EPI equation.26 Semi-quantitative random urine protein excretion by dipstick testing was available for 1477 subjects at the time of enrollment. Subjects with subsequent dipstick tests showing trace or no proteinuria (N=815) were assumed to have trace or no proteinuria at enrollment, for a total of N=2292.
[0062] Statistical analysis.
[0063] Continuous variables are presented as means (standard deviation) or as median (interquartile range), and categorical variables as proportions (%). Independent sample t-tests and chi-square tests were used to compare continuous and categorical variables, respectively. Proteinuria was dichotomized as "no proteinuria" (n=2188) which included negative or trace, and "proteinuria" (n=104) that included grades 1 + and above. Estimated GFR values >120 mL/min/1 .73 m2 (<1 % of measurements) were set at 120 mL/min/1 .73 m2. Both suPAR and eGFR were log transformed prior to analyses.
[0064] SuPAR and baseline parameters of kidney function: Independent determinants of baseline suPAR and eGFR were determined using linear regression after adjustment for age, gender, race (African American vs. others), body mass index (BMI), history of smoking, diabetes, proteinuria (>1 + vs.
negative or trace protein on dipstick), hypertension, hyperlipidemia, coronary artery disease, history of myocardial infarction, hs-CRP, and use of renin- angiotensin system (RAS) inhibitors.
[0065] SuPAR and change in eGFR at follow-up: The association between baseline suPAR levels and change in eGFR over time was investigated in 2292 subjects with follow-up eGFR measurements using linear mixed effects modeling with random subject-specific intercept and random time effect, by regressing log(eGFR) on log(suPAR), follow-up time (years since baseline), log(suPAR) * time, baseline eGFR, in addition to the aforementioned covariates. Three-way interaction terms were incorporated into the model to determine whether eGFR, race, and diabetes modified the association between suPAR and change in eGFR. Estimated decline in eGFR in each subgroup was derived accordingly.
[0066] SuPAR and incident CKD: In 1335 participants with eGFR > 60 mL/min/1 .73 m2 at baseline, we compared the progression to clinical CKD (eGFR < 60 mL/min/1 .73 m2) among the suPAR quartile groups using the log-rank test and a Cox proportional hazard model adjusting for the aforementioned
covariates. In addition, the C-statistic and category free net reclassification improvement (NRI) for risk discrimination were calculated.27
[0067] To validate our results, the analysis was repeated (1 ) after randomly dividing the entire study sample into two groups, and (2) in a subset of 347 subjects enrolled as part of the Women's Interagency HIV Study (WIHS). The Women's Interagency HIV Study (WIHS) is a longitudinal study of HIV infected and uninfected at-risk women that enrolled 2054 HIV-infected women and 569 uninfected women at six sites, Chicago, San Francisco Bay Area, Brooklyn and Bronx/Manhattan, New York, Washington, DC and Los Angeles from October 1994 through November 1995. From October 2001 through September 2002, an additional 737 HIV-infected women and 406 uninfected women were enrolled. Informed consent was obtained from all participants in accordance with the US Department of Health and Human Services (DHHS) guidelines and the
institutional review boards of participating institutions. Women are seen semiannually for interview, physical exam and collection of blood and genital specimens. The cohort was designed to reflect the demographics of the HIV epidemic among US women. Details of cohort recruitment, retention and demographics are published elsewhere. (1 ,2)
[0068] For the purpose of replicating the findings of the Emory Cardiovascular
Biobank cohort, we measured plasma suPAR in 374 randomly selected WIHS participants with eGFR>30 mL/min (N=135 or 36% with HIV) who had follow-up eGFR measurements available (median 26 measurements over at least 10 years) and determined whether suPAR levels were predictive of eGFR decline as described in the manuscript. A cutoff of 3000 pg/mL (median) for plasma suPAR was used to dichotomize the cohort subset into high and low suPAR. Plasma suPAR was measured in a blinded fashion using the human uPAR Quantikine® ELISA kit (R&D, Minneapolis, MN) following the manufacturer's instruction. The minimum detectable dose of suPAR is typically less than 33 pg/mL. The intra- and inter-assay variation is less than 5% respectively. 28,29 Two-tailed P values≤ 0.05 were considered statistical significant. All analyses were performed using SAS 9.3 (Cary, NC, USA).
[0069] The above Figures and disclosure are intended to be illustrative and not exhaustive. This description will suggest many variations and alternatives to one of ordinary skill in the art. All such variations and alternatives are intended to be encompassed within the scope of the attached claims. Those familiar with the art may recognize other equivalents to the specific embodiments described herein which equivalents are also intended to be encompassed by the attached claims.
REFERENCES
1. Coresh J, Selvin E, Stevens LA, et al. Prevalence of chronic kidney disease in the United States. Jama 2007;298:2038-47.
2. Collins AJ, Foley RN, Chavers B, et al. US Renal Data System 2013 Annual Data Report. Am J Kidney Dis 2014;63:A7.
3. Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, et al. Chronic kidney disease and cardiovascular risk: epidemiology, mechanisms, and prevention. Lancet 2013;382:339-52.
4. James MT, Hemmelgarn BR, Tonelli M. Early recognition and prevention of chronic kidney disease. Lancet 2010;375:1296-309.
5. Levey AS, Cattran D, Friedman A, et al. Proteinuria as a surrogate outcome in CKD: report of a scientific workshop sponsored by the National Kidney
Foundation and the US Food and Drug Administration. Am J Kidney Dis
2009;54:205-26.
6. Hallan SI, Ritz E, Lydersen S, Romundstad S, Kvenild K, Orth SR. Combining GFR and albuminuria to classify CKD improves prediction of ESRD. Journal of the American Society of Nephrology : JASN 2009;20:1069-77. 7. Thuno M, Macho B, Eugen-Olsen J. suPAR: the molecular crystal ball.
Disease markers 2009;27:157-72.
8. Wei C, Moller CC, Altintas MM, et al. Modification of kidney barrier function by the urokinase receptor. Nature medicine 2008;14:55-63.
9. Huai Q, Mazar AP, Kuo A, et al. Structure of human urokinase plasminogen activator in complex with its receptor. Science 2006;31 1 :656-9.
10. De Witte H, Sweep F, Brunner N, et al. Complexes between urokinase-type plasminogen activator and its receptor in blood as determined by enzyme-linked immunosorbent assay. International journal of cancer Journal international du cancer 1998;77:236-42.
1 1. Sier CF, Sidenius N, Mariani A, et al. Presence of urokinase-type
plasminogen activator receptor in urine of cancer patients and its possible clinical relevance. Laboratory investigation; a journal of technical methods and pathology 1999;79:717-22.
12. Gustafsson A, Ajeti V, Ljunggren L. Detection of suPAR in the Saliva of Healthy Young Adults: Comparison with Plasma Levels. Biomarker insights 201 1 ;6:119-25.
13. Borne Y, Persson M, Melander O, Smith JG, Engstrom G. Increased plasma level of soluble urokinase plasminogen activator receptor is associated with incidence of heart failure but not atrial fibrillation. Eur J Heart Fail 2014;16:377- 83.
14. Eugen-Olsen J, Andersen O, Linneberg A, et al. Circulating soluble urokinase plasminogen activator receptor predicts cancer, cardiovascular disease, diabetes and mortality in the general population. Journal of internal medicine
2010;268:296-308.
15. Fuhrman B. The urokinase system in the pathogenesis of atherosclerosis. Atherosclerosis 2012;222:8-14.
16. Lyngbaek S, Marott JL, Sehestedt T, et al. Cardiovascular risk prediction in the general population with use of suPAR, CRP, and Framingham Risk Score. Int J Cardiol 2013;167:2904-1 1.
17. Eapen DJ, Manocha P, Ghasemzedah N, et al. Soluble urokinase
plasminogen activator receptor level is an independent predictor of the presence and severity of coronary artery disease and of future adverse events. J Am Heart Assoc 2014;3:e001 1 18. 18. Backes Y, van der Sluijs KF, Mackie DP, et al. Usefulness of suPAR as a biological marker in patients with systemic inflammation or infection: a systematic review. Intensive Care Med 2012;38:1418-28.
19. de Bock CE, Wang Y. Clinical significance of urokinase-type plasminogen activator receptor (uPAR) expression in cancer. Med Res Rev 2004;24:13-39.
20. Yoo TH, Pedigo CE, Guzman J, et al. Sphingomyelinase-like
phosphodiesterase 3b expression levels determine podocyte injury phenotypes in glomerular disease. Journal of the American Society of Nephrology : JASN 2015;26:133-47.
21. Theilade S, Lyngbaek S, Hansen TW, et al. Soluble urokinase plasminogen activator receptor levels are elevated and associated with complications in patients with type 1 diabetes. Journal of internal medicine 2014.
22. Wei C, El Hindi S, Li J, et al. Circulating urokinase receptor as a cause of focal segmental glomerulosclerosis. Nature medicine 201 1 ;17:952-60.
23. Wei C, Trachtman H, Li J, et al. Circulating suPAR in two cohorts of primary FSGS. Journal of the American Society of Nephrology : JASN 2012;23:2051 -9.
24. National Kidney F. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis
2002;39:S1 -266.
25. Jeppesen J, Hansen TW, Olsen MH, et al. C-reactive protein, insulin resistance and risk of cardiovascular disease: a population-based study. Eur J Cardiovasc Prev Rehabil 2008;15:594-8.
26. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med 2009;150:604-12.
27. Uno H, Cai T, Pencina MJ, D'Agostino RB, Wei LJ. On the C-statistics for evaluating overall adequacy of risk prediction procedures with censored survival data. Stat Med 201 1 ;30:1 105-17.
28. Barkan SE, Melnick SL, Preston-Martin S, et al. The Women's Interagency HIV Study. WIHS Collaborative Study Group. Epidemiology 1998;9:1 17-25.
29. Hessol NA, Schneider M, Greenblatt RM, et al. Retention of women enrolled in a prospective study of human immunodeficiency virus infection: impact of race, unstable housing, and use of human immunodeficiency virus therapy. Am J Epidemiol 2001 ;154:563-73. 30. Wang TJ. Assessing the role of circulating, genetic, and imaging biomarkers in cardiovascular risk prediction. Circulation 201 1 ;123:551 -65.
31. Johnson DW. Evidence-based guide to slowing the progression of early renal insufficiency. Intern Med J 2004;34:50-7.
32. Fassett RG, Venuthurupalli SK, Gobe GC, Coombes JS, Cooper MA, Hoy WE. Biomarkers in chronic kidney disease: a review. Kidney international
201 1 ;80:806-21.
33. Andersen O, Eugen-Olsen J, Kofoed K, Iversen J, Haugaard SB. Soluble urokinase plasminogen activator receptor is a marker of dysmetabolism in HIV- infected patients receiving highly active antiretroviral therapy. J Med Virol
2008;80:209-16.
34. Meijers B, Poesen R, Claes K, et al. Soluble urokinase receptor is a biomarker of cardiovascular disease in chronic kidney disease. Kidney
international 2015;87:210-6.
35. Maile LA, Gollahon K, Wai C, Dunbar P, Busby W, Clemmons D. Blocking alphaVbeta3 integrin ligand occupancy inhibits the progression of albuminuria in diabetic rats. Journal of diabetes research 2014;2014:421827.
36. Maile LA, Busby WH, Gollahon KA, et al. Blocking ligand occupancy of the alphaVbeta3 integrin inhibits the development of nephropathy in diabetic pigs. Endocrinology 2014;155:4665-75.
37. Delville M, Sigdel TK, Wei C, et al. A circulating antibody panel for
pretransplant prediction of FSGS recurrence after kidney transplantation. Science translational medicine 2014;6:256ra136.
38. Spinale JM, Mariani LH, Kapoor S, et al. A reassessment of soluble
urokinase-type plasminogen activator receptor in glomerular disease. Kidney international 2015;87:564-74.
39. Fox CS, Larson MG, Leip EP, Culleton B, Wilson PWF, Levy D. Predictors of new-onset kidney disease in a community-based population. Jama
2004;291 :844-50. Table 1. Demographics and clinical characteristics of the Emory Cardiovascular Biobank cohort
Variables Entire Cohort suPAR<3040 pg/mL suPAR>3040 pg/mL P Value
(N=3683) (N=1839) (N=1844)
Age, years 63 (12) 60 (1 1) 65 (12) O.001 Male, n (%) 2404 (65%) 1324 (72%) 1108 (59%) O.001
African American, n (%) 654 (18%) 334 (18%) 320 (17%) 0.55
2
Body mass index, kg/m 30 (6) 29 (6) 30 (7) 0.07 Clinical Characteristics
History of smoking, n (%) 2063 (58%) 968 (55%) 1095 (61%) O.001
Hypertension, n (%) 2601 (72%) 1220 (67%) 1381 (76%) O.001
Diabetes mellitus, n (%) 1 168 (32%) 423 (23%) 745 (41%) O.001
Hy erlipidemia, n (%) 2494 (69%) 1220 (67%) 1274 (70%) 0.06
History of myocardial infarction, n (%) 1296 (36%) 568 (32%) 728 (40%) O.001
Coronary artery disease, n (%) 2279 (66%) 1092 (63%) 1 187 (69%) O.001
Renin-angiotensin system antagonists, n 2046 (63%) 998 (62%) 1048 (64%) 0.09
Urine dipstick protein >1+, n (%) 137 (9%) 32 (5%) 105 (13%) O.001 eGFR, mL/min/1.73m2 73 (23) 81 (18) 64 (24) O.001 hs-CRP, ng/mL 6.85 (12.86) 5.23 (10.83) 8.46 (14.42) O.001 suPAR, pg/mL 3493 (1938) 2330 (452) 4654 (2145) O.001
Values are mean (SD), or n (%) as noted. Coronary artery disease is defined as the presence of a >50% obstructive lesion in any of the major vessels on coronary angiogram. P value denotes differences between suPAR >3040 and <3040 pg/mL.
Table 2. Predictors of incident chronic kidney disease
HR 95% CI P Value
Baseline eGFR, per mL/min/1.73 m2 increase 0.96 (0.95,0.97) <0.001
Age, per year increase 1.01 (1.00,1.02) 0.05
Gender (female) 0.94 (0.72,1.23) 0.66
African American 1.23 (0.90,1.70) 0.20
Body mass index 1.01 (0.99,1.03) 0.39
History of smoking 1.16 (0.91,1.47) 0.24
Hypertension 1.34 (1.00,1.78) 0.05
Diabetes 1.18 (0.91,1.52) 0.22
Proteinuria 1.55 (0.86,2.81) 0.15
Hyperlipidemia 1.03 (0.78,1.35) 0.86
History of myocardial infarction 0.96 (0.76,1.23) 0.76
RAS antagonists 0.84 (0.65,1.09) 0.18
hs-CRP, > 2.8 vs. < 2.8 ng/niL 1.08 (0.84,1.38) 0.55
suPAR
Per quartile increase (1236 pg/mL) 1.40 (1.26,1.55) <0.001
> 3040 vs. < 3040 pg/mL 1.97 (1.53,2.54) <0.001
2nd vs. 1st quartile 1.18 (0.81,1.73) 0.39
3rd vs. 1st quartile 2.00 (1.38,2.89) <0.001
4th vs. 1st quartile 3.13 (2.11,4.65) <0.001
4th vs. 3rd quartile 1.51 (1.11,2.06) 0.01
CI: confidence interval, eGFR: estimated glomerular filtration rate. HR: hazard ratio, hs-CRP: high sensitivity c-reactive protein; RAS: renin- angiotensin system; suPAR: soluble urokinase-type plasminogen activator receptor. Except for baseline eGFR, age, hs- CRP, and suPAR, all variables were dichotomous (e.g., presence vs. abscense). The effect of suPAR is reported as a continuous variable, dichotomized by median (3040 pg/mL) as well as by quartiles. Subjects with a suPAR level in the third (Q3) and fourth (Q4) quartiles had a 2.0- and 3.13-fold increased hazard of incident CKD compared to those in the lowest quartile (Ql).
Table 3. Independent predictors of baseline suPAR and eGFR suPAR eGFR
Variables Change (%) 95 % CI P Value Change (%) 95 % CI P Value
Age, per year increase 0.1 (0.0,0.2) 0.19 -0.6 (-0.8,-0.5) O.001
Female gender 14.8 (11.7,18.0) O.001 5.1 (2.1,8.2) O.01
African American 2.1 (-1.5,5.8) 0.25 -5.2 (-8.6,-1.7) O.01
Body Mass Index, per unit
0.2 (0.0,0.4) 0.03 0.3 (0.1,0.5) 0.01 increase
History of Smoking 6.2 (3.5,9.0) <0.001 5.3 (2.5,8.2) <0.001
Hypertension 3.3 (0.2,6.4) 0.04 -5.6 (-8.5,-2.6) O.001
Diabetes Mellitus 14.5 (11.3,17.8) O.001 0.4 (-2.6,3.4) 0.82
Proteinuria 20.5 (12.4,29.2) O.001 -20.8 (-26.3,-15.0) O.001
Hyperlipidemia -6.4 (-9.1,-3.6) O.001 -2.1 (-5.0,0.9) 0.17
History of Myocardial Infarction 4.4 (1.4,7.4) 0.004 -1.7 (-4.6,1.2) 0.24
Coronary Artery Disease 1.2 (-1.9,4.4) 0.45 2.9 (-0.3,6.3) 0.08
RAS Antagonists -1.1 (-5.0,0.9) 0.42 3.2 (0.3,6.1) 0.03 hsCRP, per unit increase 0.4 (0.3,0.5) O.001 0.0 (-0.1,0.1) 0.88 eGFR, per unit increase -0.8 (-0.9,-0.8) <0.001 — — — suPAR, per 100% higher value — — — -14.4 (-15.1 ,-13.6) O.001 hsCRP: high sensitivity C-reactive protein; eGFR: estimated glomerular filtration rate; RAS: renin-angiotensin system; suPAR: soluble urokinase plasminogen-type activator receptor. Given both suPAR and eGFR were both log-transformed for the purpose of this analysis, estimated changes are expressed in %; i.e. female gender was independently associated with a 14.8% (95% CI [1 1.7,18.0]) increase in suPAR levels.
Table 4. Percent change in eGFR per year associated with different baseline suPAR levels for (1) all study participants and (2) validation cohorts
Overall (N=2292) Validation Cohort 1 (N= =1160) Validation Cohort 2 (N= =1132)
Figure imgf000027_0001
eGFR; estimated glomerular filtration rate. AeGFR (%): percent change in eGFR per year. Note the incrementally larger absolute decrease in eGFR associated with higher baseline suPAR values in the overall as well as the validation cohorts.
Table 5. Risk prediction metrics for suPAR for the outcome of incident CKD
Model C-statistic (95% CI) AC-statistic (95% CI) Continuous NRI (95% CI)
One Year ofFollow-Up
Model 0 0.75 (0.64, 0.85)
Model 1 (+suPAR pg/mL) 0.80 (0.71, 0.89) 0.05 (0.01, 0.10) 0.43 (0.18, 0.58)
Model 2 (+suPAR quartiles) 0.82 (0.76, 0.89) 0.08 (0.01, 0.15) 0.40 (0.15, 0.53)
Five Years of Follow-Up
Model 0 0.71 (0.67, 0.76)
Model 1 (+suPAR pg/mL) 0.74 (0.70, 0.78) 0.03 (0.01, 0.06) 0.29 (0.10, 0.38)
Model 2 (+suPAR quartiles) 0.75 (0.70, 0.79) 0.03 (0.01, 0.06) 0.34 (0.19, 0.41)
Model 0 includes baseline eGFR, age, gender, race (African American vs. others), body mass index (BMI), history of smoking, diabetes, proteinuria (>1+ vs. negative or trace protein on dipstick), hypertension, hyperlipidemia, history of myocardial infarction, hsCRP, and use of renin-angiontensin system (RAS) inhibitors. Model 1 includes all variables in Model 0 plus suPAR as a continuous variable. Model 2 include all variables in Model 0 plus suPAR quartile groups. CI = confidence interval, NRI = net reclassification index. The change in C- statistic as well as the NRI shows the addition of suPAR to the model has improved predictive accuracy of the model.
Table 6. R2 change with addition of each variable to the prediction model
Variable Adjusted R2 R2 Increase in R2
BMI 0.0022 0.0025 0.0025
Age 0.0733 0.0739 0.0714
Female Gender 0.0753 0.0761 0.0022
History Smoking 0.0795 0.0806 0.0045
African American 0.0821 0.0835 0.0029
Hypertension 0.0924 0.0941 0.0106
Diabetes Mellitus 0.1023 0.1041 0.0100
Hyperlipidemia 0.1020 0.1042 0.0001
Proteinuria 0.1314 0.1338 0.0296
Myocardial Infarction 0.1331 0.1357 0.0019
Coronary Artery Disease 0.1327 0.1358 0.0001
RAS Antagonists 0.1333 0.1368 0.0010
hsCRP 0.1371 0.1409 0.0041
Log(suPAR) 0.3289 0.3322 0.1913
Adjusted R-square, R-square, and increase in R-square of the linear regression models are shown with log(eGFR) as the response variable using baseline data. Adjusted R-square and R-square values are reported for the model with the variable on the same row and all the variables above tha row. Increase in R-square is the change in R-square for adding the variable to the model, compared to the model using all variables above that row. Note that changing the order of the variable will affect the change in R-square. Note that the increase in R2 with the addition of suPAR to the model is by far the largest compared to all other variables.
Table 7. Predictors of incident CKD and associated hazard ratios in two validation cohorts
Validation Cohort 1 (N = 661) Validation Cohort 2 (N = 674)
Effect HR 95% CI P Value HR 95% CI P Value
Baseline eGFR, per unit increase 0.96 (0.94,0.97) <0.001 0.97 (0.95,0.98) <0.001
Age, per year increase 1.01 (0.99, 1.03) 0.36 1.01 (1.00,1.03) 0.09
Gender (female) 0.88 (0.59, 1.31) 0.53 0.97 (0.67,1.40) 0.85
African American 0.97 (0.59, 1.61) 0.91 1.46 (0.94,2.27) 0.09
Body Mass Index 1.01 (0.98, 1.05) 0.50 1.01 (0.98,1.03) 0.64
History of Smoking 1.43 (1.00,2.06) 0.05 0.97 (0.69,1.36) 0.84
Hypertension 1.76 (1.14,2.71) 0.01 1.05 (0.71,1.55) 0.80
Diabetes 1.10 (0.75, 1.61) 0.63 1.29 (0.90,1.84) 0.16
Proteinuria 2.79 (1.06,7.31) 0.04 1.00 (0.46,2.20) 0.99
Hyperlipidemia 0.92 (0.62, 1.35) 0.66 1.12 (0.76,1.65) 0.56
History of Myocardial Infarction 1.01 (0.71, 1.43) 0.97 0.88 (0.62,1.26) 0.49
RAS Antagonists 0.77 (0.53, 1.1 1) 0.16 0.93 (0.64,1.35) 0.70 hsCRP, per unit increase 0.98 (0.95, 1.01) 0.22 1.00 (0.98,1.01) 0.50 suPAR
2nd vs. 1st Quartile 0.98 (0.57,1.70) 0.95 1.39 (0.81,2.40) 0.24
3rd vs. 1st Quartile 1.77 (1.07,2.93) 0.03 2.26 (1.31,3.90) <0.001
4th vs. 1st Quartile 2.76 (1.55,4.91) <0.001 3.90 (2.23,6.82) O.001
CI: confidence interval, eGFR: estimated glomerular filtration rate. HR: hazard ratio, hsCRP: high sensitivity c-reactive protein; RAS: renin-angiotensin system; suPAR: soluble urokinase plasminogen activator receptor. Note that in both validation cohorts, suPAR was independently predictive of incident CKD, with both 3 and 4* having significantly higher hazard ratios compared to the 1st quartile
Table 8. Demographics and clinical characteristics of the WIHS subset
Entire Cohort suPAR <3000 pg/mL suPAR > 3000 pg/mL P Value
Variables
(N -374) (N -172) (N -175)
Age, years 39.5 (8.2) 39.0 (8.1) 40.0 (8.3) 0.25
African American, n (%) 214 (57%) 110 (64%) 104 (59%) 0.39
Body mass index, kg/m2 28.9 (8.0) 29.5 (7.7) 28.3 (8.2) 0.18
Clinical Characteristics
HIV positive, n (%) 135 (36%) 24 (14%) 111 (63%) <0.001
History of smoking, n (%) 278 (74%) 141 (82%) 137 (78%) 0.33
Hypertension, n (%) 94 (25%) 39 (23%) 55 (31%) 0.07
Diabetes mellitus, n (%) 24 (6%) 10 (6%) 14 (8%) 0.42
HIV viral load, copies/mL of
0 (0, 80) 0 (0, 0) 80 (0, 610) <0.001 blood
eGFR, niL/min/1.73 m2 94.2 (20.0) 97.7 (19.2) 90.8 (20.1) 0.001
3023 (2085,
suPAR, pg/mL 2079 (1740, 2753) 4310 (3616, 5529) <0.001
4334)
Values are mean (SD), n (%), or median (first quartile, third quartile) as noted. P value denotes statistical significance for differences between suPAR >3000 and <3000 pg/mL using appropriate tests.
Table 9. Predictors of eGFR decline in the WIHS subset
95% Confidence
Effect Estimated AeGFR P Value
Limits
Baseline eGFR, per 1 unit
0.3% 0.2% 0.4% O.001 increase
Baseline Age, per 1 year
-0.2% -0.4% -0.1% 0.002 increase
BMI, per 1 unit increase 0.0% -0.2% 0.1% 0.76
African American race 1.3% -0.9% 3.5% 0.26
Hypertension -1.5% -4.0% 1.0% 0.24
Diabetes mellitus -1.9% -6.0% 2.4% 0.38
History of smoking 0.3% -2.3% 2.9% 0.83
HIV viral load, per 100%
-0.1% -0.3% 0.2% 0.53 increase
Follow-up Time, year -0.1% -0.4% 0.1% 0.29 suPAR: >3000 pg/mL vs.
0.4% -1.9% 2.9% 0.72 <3000 pg/mL
Follow-up Time x suPAR
-0.5% -0.8% -0.1% 0.02 >3000 pg/mL
Results of the linear mixed model with eGFR at follow-up visits as the outcome variable and binary suPAR (>3000 vs. <3000 pg/mL) at baseline as the independent variable along with the covariates listed.

Claims

1. A method of treating a subject at risk of developing a chronic kidney disease (CKD), the method comprising: obtaining a biological sample from the subject, the subject having an estimated glomerular filtration rate (eGFR) of >60 mL/min/1.73m2; assaying a soluble urokinase-type plasminogen activator receptor (suPAR) level in the sample; determining the suPAR level in the sample relative to a control sample; identifying the subject having an elevated suPAR level relative to the control; and stratifying the subject having the elevated suPAR level to a treatment group for treating CKD.
2. The method according to claim 1 , comprising stratifying the subject to the treatment group when the suPAR level is about 3000 pg/ml or greater.
3. The method according to claim 1 , comprising stratifying the subject to the treatment group when the suPAR level is about 3040 pg/ml or greater.
4. The method according to claim 1 , comprising stratifying the subject to the treatment group when the suPAR level is about 4020 pg/ml or greater.
5. The method according to any one of claims 1 -4, comprising treating the subject with a therapy to treat CKD or to decrease the suPAR level in the subject.
6. The method according to any one of claims 1 -5, comprising stratifying the subject having the elevated suPAR level to the treatment group for subjects having an eGFR of >90 mL/min/1.73m2.
7. The method according to any one of claims 1 -6, wherein the biological sample comprises a blood sample.
8. The method according to claim 7, wherein the blood sample comprises a plasma sample.
9. The method according to any one of claims 1 -8, comprising stratifying the subject having the elevated suPAR level to the treatment group in the absence of proteinuria.
10. The method according to any one of claims 1 -9, wherein the subject has diabetes or hypertension.
1 1. The method according to any one of claims 1 -9, comprising obtaining a second biological sample from the subject after a treatment has been given and determining the suPAR level in the second sample.
12. A method of identifying a subject at risk of developing chronic kidney disease (CKD), the method comprising: obtaining a biological sample from the subject, the subject having an estimated glomerular filtration rate (eGFR) of >60 mL/min/1.73m2; assaying a level of soluble urokinase-type plasminogen activator receptor (suPAR) in the sample; identifying the subject having a suPAR level of about 3000 pg/ml or greater as being at risk for developing CKD.
13. The method according to claim 12, comprising identifying the subject as being at risk for developing CKD when the suPAR level is about 3040 pg/ml or greater.
14. The method according to claim 12 comprising identifying the subject as being at risk for developing CKD when the suPAR level is about 4020 pg/ml or greater.
15. The method according to any one of claims 12-14, comprising stratifying the subject having the elevated suPAR level to the treatment group for subjects having an eGFR of >90 mL/min/1.73m2.
16. The method according to any one of claims 12-15, wherein the biological sample comprises a blood sample.
17. The method according to claim 16, wherein the blood sample comprises a plasma sample.
PCT/US2016/049421 2015-08-31 2016-08-30 Prediction of kidney disease, severity and related treatment approaches WO2017040488A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562212370P 2015-08-31 2015-08-31
US62/212,370 2015-08-31

Publications (1)

Publication Number Publication Date
WO2017040488A1 true WO2017040488A1 (en) 2017-03-09

Family

ID=58188325

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/049421 WO2017040488A1 (en) 2015-08-31 2016-08-30 Prediction of kidney disease, severity and related treatment approaches

Country Status (1)

Country Link
WO (1) WO2017040488A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020146745A1 (en) * 2019-01-11 2020-07-16 Quadrus Medical Technologies, Inc. Systems and methods for assessing and evaluating renal health diagnosis, staging, and therapy recommendation
WO2020206129A1 (en) * 2019-04-02 2020-10-08 The Regents Of The University Of Michigan Use of soluble urokinase plasminogen activator receptor levels in the management of patients with cardiovascular disease
US11026625B2 (en) 2017-08-08 2021-06-08 Fresenius Medical Care Holdings, Inc. Systems and methods for treating and estimating progression of chronic kidney disease
WO2021204902A1 (en) * 2020-04-08 2021-10-14 Virogates A/S Sars-cov-2 infection risk assessment method
WO2023081817A1 (en) * 2021-11-04 2023-05-11 The Regents Of The University Of Michigan Methods to target the soluble urokinase plasminogen activator receptor pathway for the prevention and treatment of atherosclerosis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140083945A1 (en) * 2011-05-09 2014-03-27 The University Of Miami Reducing Soluble Urokinase Receptor in the Circulation
US20140302065A1 (en) * 2011-10-31 2014-10-09 The University Of Miami Soluble urokinase receptor (supar) in diabetic kidney disease

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140083945A1 (en) * 2011-05-09 2014-03-27 The University Of Miami Reducing Soluble Urokinase Receptor in the Circulation
US20140302065A1 (en) * 2011-10-31 2014-10-09 The University Of Miami Soluble urokinase receptor (supar) in diabetic kidney disease

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HAYEK, SS ET AL.: "Soluble Urokinase Receptor and Chronic Kidney Disease.", NEW ENGLAND JOURNAL OF MEDICINE., vol. 373, no. 20, 5 November 2015 (2015-11-05), pages 1916 - 1925, XP055318995 *
MALLAPPALLIL, M ET AL.: "Chronic kidney disease in the elderly: evaluation and management.", CLINICAL PRACTICE, vol. 11, no. 5, 2014, pages 525 - 535, XP055372630 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11026625B2 (en) 2017-08-08 2021-06-08 Fresenius Medical Care Holdings, Inc. Systems and methods for treating and estimating progression of chronic kidney disease
WO2020146745A1 (en) * 2019-01-11 2020-07-16 Quadrus Medical Technologies, Inc. Systems and methods for assessing and evaluating renal health diagnosis, staging, and therapy recommendation
WO2020206129A1 (en) * 2019-04-02 2020-10-08 The Regents Of The University Of Michigan Use of soluble urokinase plasminogen activator receptor levels in the management of patients with cardiovascular disease
WO2021204902A1 (en) * 2020-04-08 2021-10-14 Virogates A/S Sars-cov-2 infection risk assessment method
WO2023081817A1 (en) * 2021-11-04 2023-05-11 The Regents Of The University Of Michigan Methods to target the soluble urokinase plasminogen activator receptor pathway for the prevention and treatment of atherosclerosis

Similar Documents

Publication Publication Date Title
JP7068256B2 (en) Methods and Compositions for Diagnosis and Prognosis of Renal Injury and Renal Failure
Otaki et al. Association of plasma xanthine oxidoreductase activity with severity and clinical outcome in patients with chronic heart failure
Hodges et al. suPAR: a new biomarker for cardiovascular disease?
Sonmez et al. Novel fibro-inflammation markers in assessing left atrial remodeling in non-valvular atrial fibrillation
WO2017040488A1 (en) Prediction of kidney disease, severity and related treatment approaches
Xing et al. Clinical utility of biomarkers of endothelial activation in sepsis-a systematic review
Delanaye et al. Clinical and biological determinants of sclerostin plasma concentration in hemodialysis patients
US11243217B2 (en) Management of acute kidney injury using insulin-like growth factor-binding protein 7 and tissue inhibitor of metalloproteinase 2
EP3004873B1 (en) Methods and compositions for diagnosis and prognosis of renal injury and renal failure
Fragoso et al. Insulin resistance as a predictor of cardiovascular morbidity and end-stage renal disease
Hayek et al. Cardiovascular disease biomarkers and suPAR in predicting decline in renal function: a prospective cohort study
McManus et al. Guidelines for chronic kidney disease: Defining, staging, and managing in primary care
Coiro et al. Elevated serum uric acid concentration at discharge confers additive prognostic value in elderly patients with acute heart failure
Wang et al. Comparison between the effects of hydrochlorothiazide and indapamide on the kidney in hypertensive patients inadequately controlled with losartan
Vianello et al. β2-Microglobulin and TIMP1 are linked together in cardiorenal remodeling and failure
Mihai et al. Inflammation-related patterns in the clinical staging and severity assessment of chronic kidney disease
Banai et al. Neutrophil gelatinase-associated lipocalin (NGAL) for the prediction of acute kidney injury in chronic kidney disease patients treated with primary percutaneous coronary intervention
Ishiwata et al. Clinical and prognostic values of urinary alpha1-microglobulin as a tubular marker in acute heart failure
Duan et al. Detection and evaluation of renal biomarkers in a swine model of acute myocardial infarction and reperfusion
Roehm et al. Novel Biomarkers of Kidney Disease in Advanced Heart Failure: Beyond GFR and Proteinuria
Murakoshi et al. Circulating tumor necrosis factor-related biomarkers predict kidney function decline in Japanese patients with diabetes: An observational cohort study
Åkerblom et al. Outcome and causes of renal deterioration evaluated by serial cystatin C measurements in acute coronary syndrome patients—results from the PLATelet inhibition and patient Outcomes (PLATO) study
Funayama et al. Renal tubulointerstitial damage is associated with short-term cardiovascular events in patients with myocardial infarction
EP2882767B1 (en) Evaluating renal injury using hyaluronic acid
Khalil et al. The early detection and prevention of contrast induced nephropathy post coronary intervention in catheterization unit

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16842798

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16842798

Country of ref document: EP

Kind code of ref document: A1