User login
Exercise-induced proteinuria?
• Rely on a spot urine microalbumin-to-creatinine or protein-to-creatinine ratio to accurately assess proteinuria. B
• Repeat testing if routine urinalysis detects proteinuria—especially if the patient reports having exercised in the previous 24 hours. B
Strength of recommendation (SOR)
A Good-quality patient-oriented evidence
B Inconsistent or limited-quality patient-oriented evidence
C Consensus, usual practice, opinion, disease-oriented evidence, case series
CASE As part of a routine physical examination, urinalysis reveals that a patient new to your practice is excreting an excessive level of protein. The patient is physically fit and shared during the history taking that he is an avid runner. The physical examination and other laboratory values were unremarkable. How concerned should you be about the finding of proteinuria?
Exercise-induced proteinuria is generally benign and a function of the intensity—rather than the duration—of exercise.1 It occurs most often among athletes participating in such sports as running, swimming, rowing, football, or boxing.2 It’s also transient—lasting 24 to 48 hours.1 Recognizing exercise-induced proteinuria is fairly straightforward—once you know what to look for.
But first, a word about the processes at work.
Diverse processes that work alone—or together
The normal range of protein excretion in healthy individuals is 150 to 200 mg of protein per day, of which albumin constitutes 10 to 20 mg.3 Individuals with proteinuria persistently higher than this level need further evaluation.
Diverse processes leading to proteinuria—working alone or concomitantly—occur at the level of the nephron.3
Glomerular proteinuria results from increased filtration of macromolecules such as albumin across the glomerular capillary barrier. This type of proteinuria can occur with different glomerulopathies, upright posture, or exercise.4
Researchers have not identified the mechanisms leading to postexercise proteinuria, but there are several theories. (For more on this, see “Why does exercise increase protein excretion?”.)
Tubular proteinuria is due to a deranged tubular apparatus with an intact glomerulus. This results in the escape of β2-microglobulin and immunoglobulin light chains from proximal tubular reabsorption. It is often missed on dipstick testing, which detects only albumin. This type of proteinuria is usually seen in tubulointerstitial diseases or in patients with idiopathic nephrotic syndrome.5
Overflow proteinuria occurs when small molecular light chains escape the glomerular filtration barrier and overwhelm the tubular reabsorptive capacity. This type of proteinuria can be seen in multiple myeloma, and is detectable by protein-to-creatinine ratio or urine protein electrophoresis.
The root cause of exercise-induced proteinuria is unclear, but the renin-angiotensin system (RAS) and prostaglandins are thought to play a major role. The plasma concentration of angiotensin II increases during exercise, leading to filtration of protein through the glomerular membrane.30 And angiotensin-converting enzyme (ACE) inhibitors have been shown to significantly diminish exercise-induced proteinuria, thus supporting this theory.31,32
Also, strenuous exercise increases sympathetic nervous system activity as well as blood levels of catecholamines, thereby increasing permeability of the glomerular capillary membrane.33 Furthermore, lactate increases with strenuous exercise and causes conformational changes in serum proteins that, when coupled with glomerular barrier changes, can lead to increased permeability and protein excretion.
The surest means of detecting proteinuria
Albumin excretion >300 mg/d is called macroalbuminuria, overt proteinuria, or dipstick-positive proteinuria. Albumin persistently excreted in the urine between 30 and 300 mg/d is referred to as microalbuminuria.
Because microalbuminuria is not detectable by a standard urine dipstick test, some providers routinely screen for protein with the microalbumin-to-creatinine ratio. A first-voided morning urine specimen is recommended, but random urine samples are an acceptable alternative.6 The microalbumin-to-creatinine ratio is recommended as a screen for early diabetic nephropathy and other kidney diseases. And a positive test result may also suggest increased risk of cardiovascular disease.6 Microalbuminuria is defined as persistent albumin excretion between 30 and 300 mg/d.7
When exercise is a factor, here’s what to look for
As noted earlier, exercise-induced proteinuria is a function of the intensity of the exercise. Moderate and strenuous (vigorous) exercise are the 2 types of exercise that come into play when discussing proteinuria. Differentiating them is not precise, but is often defined by maximal oxygen consumption (vigorous=60% of VO2max; moderate <60% VO2max); metabolic equivalents (vigorous=6 METS; moderate <6 METS); walking/running speeds (various); and heart rate reserve (vigorous=60% HRR; moderate <60% HRR).8
Moderate exercise produces glomerular proteinuria, with an increase in macromolecular (albumin) filtration across the glomerular barrier. Strenuous exercise increases glomerular filtration of low-molecular-weight proteins (β2-microglobulin), which overwhelm the reabsorbing capacity of the tubular apparatus, causing temporary dysfunction and tubular proteinuria.9 Thus, the pathophysiology is mixed, with a major contribution from glomerular proteinuria.10
Strenuous exercise can cause protein excretion to exceed 1.5 mg/min.11 However, it seldom rises beyond 1 to 2 g/d,4 and this increase usually reverts to normal physiologic levels within 24 to 48 hours after exercise.12
Exercise-induced proteinuria is biphasic.13 Increased protein excretion occurs 30 minutes after exercise and is related to changes in intraglomerular hemodynamics and the resulting saturation of the renal tubules. Around 24 hours after exercise, oxidative stress on the glomeruli causes another slight elevation in albumin excretion without changes in β2-microglobulin, thereby indicating glomerular proteinuria exclusively.
Even the pros aren’t exempt. Exercise-induced proteinuria does not decrease with regular physical training. This was demonstrated in a study of 10 well-trained professional cyclists for whom strenuous exercise increased overnight protein excretion of both tubular and glomerular origin despite ongoing regular physical training.14
Creatine supplements do not increase proteinuria. A study of creatine supplementation in animal models noted no changes in 24-hour proteinuria or albumin excretion in both normal and two-thirds-nephrectomized animals.15 Another study compared creatine use with nonuse in athletes who had been training regularly and strenuously (12- 18 h/wk) for 5 to 10 years. They were evaluated for 10 months to 5 years. The groups exhibited equivalent urine excretion rates for albumin and creatinine, with no deleterious effect on kidney function.16
What happens when chronic disease is factored into the exercise equation?
Patients with a 2- to 20-year history of insulin-dependent diabetes without chronic kidney disease (CKD) who exhibited normal albumin excretion at baseline were more likely to develop proteinuria after exercise than healthy controls.17,18 The postulated cause was undetected glomerular changes due to diabetes. An exercise-provocation test may one day be useful in predicting future development of nephropathy, but further studies are needed.19-21
Exercise increases proteinuria immediately in individuals with metabolic disorders like obesity, through a mechanism different from diabetes mellitus. Proteinuria in the obese population is thought to be glomerular in origin, as opposed to both tubular and glomerular proteinuria in diabetic nephropathy.22,23
In CKD, low-intensity exercise long term does not promote proteinuria or lead to rapid progression of CKD. In one study, obese patients (body mass index >30 kg/m2) with diabetes and CKD stage II to IV who exercised 3 times weekly (aerobic training for 6 weeks, followed by 18 weeks of supervised home exercise) increased their stamina and exhibited slight, statistically insignificant decreases in resting systolic blood pressure and 24-hour proteinuria.24 A 12-week low-intensity aquatic exercise program for 26 patients with mild to moderate CKD decreased blood pressure and proteinuria and slightly improved glomerular filtration rate (GFR).25 These results for proteinuria and GFR were shown previously in rats with subtotal nephrectomy.26
Elevated urinary albumin excretion with exercise is significantly higher in patients with acromegaly when compared with normal healthy subjects. The underlying pathology is thought to occur at the glomerular filtration barrier with intact tubular function. Somatostatin analog treatment for acromegaly leads to reductions in postexercise albuminuria.27,28
How to manage suspected exercise-induced proteinuria
When interpreting the meaning of proteinuria detected on routine urinalysis, keep in mind the temporal relevance between exercise and urine collection. If urine is found to have been collected within 24 hours of intense exercise, repeat testing in the absence of prior exercise on at least one other occasion to differentiate between transient and persistent proteinuria. In confirming transient proteinuria after exercise, reassure the patient that it is a benign condition. This holds true as well for routine microalbumin-to-creatinine urine testing in patients with diabetes who exercise. If the result of a repeat test is high, turn your attention to another possible cause of proteinuria, such as diabetic nephropathy.
Screening for proteinuria during sports preparticipation examinations is not recommended because the diagnostic utility is low.29 Researchers performed urine dipstick testing for protein, blood, and glucose in preparticipation assessments of 701 students.29 They detected proteinuria in 40 students and glucosuria in one. Follow-up testing with first-voided morning urine specimens and glucose tolerance testing was normal in all students.
CORRESPONDENCE Fahad Saeed, MD, 313 Brook Hollow, Hanover, NH 03755; [email protected]
1. Poortmans JR. Exercise and renal function. Sports Med. 1984;1:125-153.
2. Gebke KB. Genitourinary system. In: McKeag DB, Moeller JL, eds. ACSM’s Primary Care Sports Medicine. 2nd ed. Philadelphia, Pa: Lippincott Williams & Wilkins; 2007;234.-
3. Venkat KK. Proteinuria and microalbuminuria in adults: significance, evaluation, and treatment. South Med J. 2004;97:969-979.
4. Rose BD. Pathophysiology of Renal Disease. 2nd ed. New York, NY: McGraw-Hill; 1987;11-16.
5. Sesso R, Santos AP, Nishida SK, et al. Prediction of steroid responsiveness in the idiopathic nephrotic syndrome using urinary retinol-binding protein and beta-2-microglobulin. Ann Intern Med. 1992;116:905-909.
6. Levey AS, Coresh J, Balk E, et al. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139:137-147.
7. Family Practice Notebook Urine protein to creatinine ratio. Available at: http://www.fpnotebook.com/urology/lab/urnprtntcrtnrt.htm. Accessed August 9, 2011.
8. Swain DP, Franklin BA. Comparison of cardioprotective benefits of vigorous versus moderate intensity aerobic exercise. Am J Cardiol. 2006;97:141-147.
9. Poortmans JR, Labilloy D. The influence of work intensity on postexercise proteinuria. Eur J Appl Physiol Occup Physiol. 1988;57:260-263.
10. Estivi P, Urbino R, Tetta C, et al. Urinary protein excretion induced by exercise: effect of a mountain agonistic footrace in healthy subjects. Renal function and mountain footrace. J Sports Med Phys Fitness. 1992;32:196-200.
11. Poortmans JR, Brauman H, Staroukine M, et al. Indirect evidence of glomerular/tubular mixed-type postexercise proteinuria in healthy humans. Am J Physiol. 1988;254:F277-F283.
12. Heathcote KL, Wilson MP, Quest DW, et al. Prevalence and duration of exercise induced albuminuria in healthy people. Clin Invest Med. 2009;32:E261-E265.
13. Sentürk UK, Kuru O, Koçer G, et al. Biphasic pattern of exercise-induced proteinuria in sedentary and trained men. Nephron Physiol. 2007;105:22-32.
14. Clerico A, Giammattei C, Cecchini L, et al. Exercise-induced proteinuria in well-trained athletes. Clin Chem. 1990;36:562-564.
15. Taes YE, Delanghe JR, Wuyts B, et al. Creatine supplementation does not affect kidney function in an animal model with pre-existing renal failure. Nephrol Dial Transplant. 2003;18:258-264.
16. Poortmans JR, Francaux M. Long-term oral creatine supplementation does not impair renal function in healthy athletes. Med Sci Sports Exerc. 1999;31:1108-1110.
17. Mogensen CE, Vittinghus E, Sølling K. Abnormal albumin excretion after two provocative renal tests in diabetes: physical exercise and lysine injection. Kidney Int. 1979;16:385-393.
18. Vittinghus E, Mogensen CE. Albumin excretion during physical exercise in diabetes. Studies on the effect of insulin treatment and of the renal haemodynamic response. Acta Endocrinol Suppl (Copenh). 1981;242:61-62.
19. Watts GF, Williams I, Morris RW, et al. An acceptable exercise test to study microalbuminuria in type 1 diabetes. Diabet Med. 1989;6:787-792.
20. Pan X, Wang P, Hu N, et al. A physiologically based pharmacokinetic model characterizing mechanism-based inhibition of CYP1A2 for predicting theophylline/antofloxacin interaction in both rats and humans. Drug Metab Pharmacokinet. 2011;26:387-398.
21. O’Brien SF, Watts GF, Powrie JK, et al. Exercise testing as a long-term predictor of the development of microalbuminuria in normoalbuminuric IDDM patients. Diabetes Care. 1995;18:1602-1605.
22. Hidaka S, Kaneko O, Shirai M, et al. Do obesity and non-insulin dependent diabetes mellitus aggravate exercise-induced microproteinuria? Clin Chim Acta. 1998;275:115-126.
23. Hidaka S, Kakuta S, Okada H, et al. Exercise-induced proteinuria in diseases with metabolic disorders. Contrib Nephrol. 1990;83:136-143.
24. Leehey DJ, Moinuddin I, Bast JP, et al. Aerobic exercise in obese diabetic patients with chronic kidney disease: a randomized and controlled pilot study. Cardiovasc Diabetol. 2009;8:62.-
25. Pechter U, Ots M, Mesikepp S, et al. Beneficial effects of water-based exercise in patients with chronic kidney disease. Int J Rehabil Res. 2003;26:153-156.
26. Heifets M, Davis TA, Tegtmeyer E, et al. Exercise training ameliorates progressive renal disease in rats with subtotal nephrectomy. Kidney Int. 1987;32:815-820.
27. Manelli F, Bossoni S, Burattin A, et al. Exercise-induced microalbuminuria in patients with active acromegaly: acute effects of slow-release lanreotide, a long-acting somatostatin analog. Metabolism. 2000;49:634-639.
28. Hoogenberg K, Sluiter WJ, Dullaart RP. Effect of growth hormone and insulin-like growth factor I on urinary albumin excretion: studies in acromegaly and growth hormone deficiency. Acta Endocrinol (Copenh). 1993;129:151-157.
29. Goldberg B, Saraniti A, Witman P, et al. Pre-participation sports assessment—an objective evaluation. Pediatrics. 1980;66:736-745.
30. Garrett WE, Kirkendall DT, Squire DL. eds. Principles and Practice of Primary Care Sports Medicine. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001;299-310.
31. Cosenzi A, Carraro M, Sacerdote A, et al. Involvement of the renin angiotensin system in the pathogenesis of postexercise proteinuria. Scand J Urol Nephrol. 1993;27:301-304.
32. Székács B, Vajo Z, Dachman W. Effect of ACE inhibition by benazepril, enalapril and captopril on chronic and post exercise proteinuria. Acta Physiol Hung. 1996;84:361-367.
33. Poortmans JR, Haggenmacher C, Vanderstraeten J. Postexercise proteinuria in humans and its adrenergic component. J Sports Med Phys Fitness. 2001;41:95-100.
• Rely on a spot urine microalbumin-to-creatinine or protein-to-creatinine ratio to accurately assess proteinuria. B
• Repeat testing if routine urinalysis detects proteinuria—especially if the patient reports having exercised in the previous 24 hours. B
Strength of recommendation (SOR)
A Good-quality patient-oriented evidence
B Inconsistent or limited-quality patient-oriented evidence
C Consensus, usual practice, opinion, disease-oriented evidence, case series
CASE As part of a routine physical examination, urinalysis reveals that a patient new to your practice is excreting an excessive level of protein. The patient is physically fit and shared during the history taking that he is an avid runner. The physical examination and other laboratory values were unremarkable. How concerned should you be about the finding of proteinuria?
Exercise-induced proteinuria is generally benign and a function of the intensity—rather than the duration—of exercise.1 It occurs most often among athletes participating in such sports as running, swimming, rowing, football, or boxing.2 It’s also transient—lasting 24 to 48 hours.1 Recognizing exercise-induced proteinuria is fairly straightforward—once you know what to look for.
But first, a word about the processes at work.
Diverse processes that work alone—or together
The normal range of protein excretion in healthy individuals is 150 to 200 mg of protein per day, of which albumin constitutes 10 to 20 mg.3 Individuals with proteinuria persistently higher than this level need further evaluation.
Diverse processes leading to proteinuria—working alone or concomitantly—occur at the level of the nephron.3
Glomerular proteinuria results from increased filtration of macromolecules such as albumin across the glomerular capillary barrier. This type of proteinuria can occur with different glomerulopathies, upright posture, or exercise.4
Researchers have not identified the mechanisms leading to postexercise proteinuria, but there are several theories. (For more on this, see “Why does exercise increase protein excretion?”.)
Tubular proteinuria is due to a deranged tubular apparatus with an intact glomerulus. This results in the escape of β2-microglobulin and immunoglobulin light chains from proximal tubular reabsorption. It is often missed on dipstick testing, which detects only albumin. This type of proteinuria is usually seen in tubulointerstitial diseases or in patients with idiopathic nephrotic syndrome.5
Overflow proteinuria occurs when small molecular light chains escape the glomerular filtration barrier and overwhelm the tubular reabsorptive capacity. This type of proteinuria can be seen in multiple myeloma, and is detectable by protein-to-creatinine ratio or urine protein electrophoresis.
The root cause of exercise-induced proteinuria is unclear, but the renin-angiotensin system (RAS) and prostaglandins are thought to play a major role. The plasma concentration of angiotensin II increases during exercise, leading to filtration of protein through the glomerular membrane.30 And angiotensin-converting enzyme (ACE) inhibitors have been shown to significantly diminish exercise-induced proteinuria, thus supporting this theory.31,32
Also, strenuous exercise increases sympathetic nervous system activity as well as blood levels of catecholamines, thereby increasing permeability of the glomerular capillary membrane.33 Furthermore, lactate increases with strenuous exercise and causes conformational changes in serum proteins that, when coupled with glomerular barrier changes, can lead to increased permeability and protein excretion.
The surest means of detecting proteinuria
Albumin excretion >300 mg/d is called macroalbuminuria, overt proteinuria, or dipstick-positive proteinuria. Albumin persistently excreted in the urine between 30 and 300 mg/d is referred to as microalbuminuria.
Because microalbuminuria is not detectable by a standard urine dipstick test, some providers routinely screen for protein with the microalbumin-to-creatinine ratio. A first-voided morning urine specimen is recommended, but random urine samples are an acceptable alternative.6 The microalbumin-to-creatinine ratio is recommended as a screen for early diabetic nephropathy and other kidney diseases. And a positive test result may also suggest increased risk of cardiovascular disease.6 Microalbuminuria is defined as persistent albumin excretion between 30 and 300 mg/d.7
When exercise is a factor, here’s what to look for
As noted earlier, exercise-induced proteinuria is a function of the intensity of the exercise. Moderate and strenuous (vigorous) exercise are the 2 types of exercise that come into play when discussing proteinuria. Differentiating them is not precise, but is often defined by maximal oxygen consumption (vigorous=60% of VO2max; moderate <60% VO2max); metabolic equivalents (vigorous=6 METS; moderate <6 METS); walking/running speeds (various); and heart rate reserve (vigorous=60% HRR; moderate <60% HRR).8
Moderate exercise produces glomerular proteinuria, with an increase in macromolecular (albumin) filtration across the glomerular barrier. Strenuous exercise increases glomerular filtration of low-molecular-weight proteins (β2-microglobulin), which overwhelm the reabsorbing capacity of the tubular apparatus, causing temporary dysfunction and tubular proteinuria.9 Thus, the pathophysiology is mixed, with a major contribution from glomerular proteinuria.10
Strenuous exercise can cause protein excretion to exceed 1.5 mg/min.11 However, it seldom rises beyond 1 to 2 g/d,4 and this increase usually reverts to normal physiologic levels within 24 to 48 hours after exercise.12
Exercise-induced proteinuria is biphasic.13 Increased protein excretion occurs 30 minutes after exercise and is related to changes in intraglomerular hemodynamics and the resulting saturation of the renal tubules. Around 24 hours after exercise, oxidative stress on the glomeruli causes another slight elevation in albumin excretion without changes in β2-microglobulin, thereby indicating glomerular proteinuria exclusively.
Even the pros aren’t exempt. Exercise-induced proteinuria does not decrease with regular physical training. This was demonstrated in a study of 10 well-trained professional cyclists for whom strenuous exercise increased overnight protein excretion of both tubular and glomerular origin despite ongoing regular physical training.14
Creatine supplements do not increase proteinuria. A study of creatine supplementation in animal models noted no changes in 24-hour proteinuria or albumin excretion in both normal and two-thirds-nephrectomized animals.15 Another study compared creatine use with nonuse in athletes who had been training regularly and strenuously (12- 18 h/wk) for 5 to 10 years. They were evaluated for 10 months to 5 years. The groups exhibited equivalent urine excretion rates for albumin and creatinine, with no deleterious effect on kidney function.16
What happens when chronic disease is factored into the exercise equation?
Patients with a 2- to 20-year history of insulin-dependent diabetes without chronic kidney disease (CKD) who exhibited normal albumin excretion at baseline were more likely to develop proteinuria after exercise than healthy controls.17,18 The postulated cause was undetected glomerular changes due to diabetes. An exercise-provocation test may one day be useful in predicting future development of nephropathy, but further studies are needed.19-21
Exercise increases proteinuria immediately in individuals with metabolic disorders like obesity, through a mechanism different from diabetes mellitus. Proteinuria in the obese population is thought to be glomerular in origin, as opposed to both tubular and glomerular proteinuria in diabetic nephropathy.22,23
In CKD, low-intensity exercise long term does not promote proteinuria or lead to rapid progression of CKD. In one study, obese patients (body mass index >30 kg/m2) with diabetes and CKD stage II to IV who exercised 3 times weekly (aerobic training for 6 weeks, followed by 18 weeks of supervised home exercise) increased their stamina and exhibited slight, statistically insignificant decreases in resting systolic blood pressure and 24-hour proteinuria.24 A 12-week low-intensity aquatic exercise program for 26 patients with mild to moderate CKD decreased blood pressure and proteinuria and slightly improved glomerular filtration rate (GFR).25 These results for proteinuria and GFR were shown previously in rats with subtotal nephrectomy.26
Elevated urinary albumin excretion with exercise is significantly higher in patients with acromegaly when compared with normal healthy subjects. The underlying pathology is thought to occur at the glomerular filtration barrier with intact tubular function. Somatostatin analog treatment for acromegaly leads to reductions in postexercise albuminuria.27,28
How to manage suspected exercise-induced proteinuria
When interpreting the meaning of proteinuria detected on routine urinalysis, keep in mind the temporal relevance between exercise and urine collection. If urine is found to have been collected within 24 hours of intense exercise, repeat testing in the absence of prior exercise on at least one other occasion to differentiate between transient and persistent proteinuria. In confirming transient proteinuria after exercise, reassure the patient that it is a benign condition. This holds true as well for routine microalbumin-to-creatinine urine testing in patients with diabetes who exercise. If the result of a repeat test is high, turn your attention to another possible cause of proteinuria, such as diabetic nephropathy.
Screening for proteinuria during sports preparticipation examinations is not recommended because the diagnostic utility is low.29 Researchers performed urine dipstick testing for protein, blood, and glucose in preparticipation assessments of 701 students.29 They detected proteinuria in 40 students and glucosuria in one. Follow-up testing with first-voided morning urine specimens and glucose tolerance testing was normal in all students.
CORRESPONDENCE Fahad Saeed, MD, 313 Brook Hollow, Hanover, NH 03755; [email protected]
• Rely on a spot urine microalbumin-to-creatinine or protein-to-creatinine ratio to accurately assess proteinuria. B
• Repeat testing if routine urinalysis detects proteinuria—especially if the patient reports having exercised in the previous 24 hours. B
Strength of recommendation (SOR)
A Good-quality patient-oriented evidence
B Inconsistent or limited-quality patient-oriented evidence
C Consensus, usual practice, opinion, disease-oriented evidence, case series
CASE As part of a routine physical examination, urinalysis reveals that a patient new to your practice is excreting an excessive level of protein. The patient is physically fit and shared during the history taking that he is an avid runner. The physical examination and other laboratory values were unremarkable. How concerned should you be about the finding of proteinuria?
Exercise-induced proteinuria is generally benign and a function of the intensity—rather than the duration—of exercise.1 It occurs most often among athletes participating in such sports as running, swimming, rowing, football, or boxing.2 It’s also transient—lasting 24 to 48 hours.1 Recognizing exercise-induced proteinuria is fairly straightforward—once you know what to look for.
But first, a word about the processes at work.
Diverse processes that work alone—or together
The normal range of protein excretion in healthy individuals is 150 to 200 mg of protein per day, of which albumin constitutes 10 to 20 mg.3 Individuals with proteinuria persistently higher than this level need further evaluation.
Diverse processes leading to proteinuria—working alone or concomitantly—occur at the level of the nephron.3
Glomerular proteinuria results from increased filtration of macromolecules such as albumin across the glomerular capillary barrier. This type of proteinuria can occur with different glomerulopathies, upright posture, or exercise.4
Researchers have not identified the mechanisms leading to postexercise proteinuria, but there are several theories. (For more on this, see “Why does exercise increase protein excretion?”.)
Tubular proteinuria is due to a deranged tubular apparatus with an intact glomerulus. This results in the escape of β2-microglobulin and immunoglobulin light chains from proximal tubular reabsorption. It is often missed on dipstick testing, which detects only albumin. This type of proteinuria is usually seen in tubulointerstitial diseases or in patients with idiopathic nephrotic syndrome.5
Overflow proteinuria occurs when small molecular light chains escape the glomerular filtration barrier and overwhelm the tubular reabsorptive capacity. This type of proteinuria can be seen in multiple myeloma, and is detectable by protein-to-creatinine ratio or urine protein electrophoresis.
The root cause of exercise-induced proteinuria is unclear, but the renin-angiotensin system (RAS) and prostaglandins are thought to play a major role. The plasma concentration of angiotensin II increases during exercise, leading to filtration of protein through the glomerular membrane.30 And angiotensin-converting enzyme (ACE) inhibitors have been shown to significantly diminish exercise-induced proteinuria, thus supporting this theory.31,32
Also, strenuous exercise increases sympathetic nervous system activity as well as blood levels of catecholamines, thereby increasing permeability of the glomerular capillary membrane.33 Furthermore, lactate increases with strenuous exercise and causes conformational changes in serum proteins that, when coupled with glomerular barrier changes, can lead to increased permeability and protein excretion.
The surest means of detecting proteinuria
Albumin excretion >300 mg/d is called macroalbuminuria, overt proteinuria, or dipstick-positive proteinuria. Albumin persistently excreted in the urine between 30 and 300 mg/d is referred to as microalbuminuria.
Because microalbuminuria is not detectable by a standard urine dipstick test, some providers routinely screen for protein with the microalbumin-to-creatinine ratio. A first-voided morning urine specimen is recommended, but random urine samples are an acceptable alternative.6 The microalbumin-to-creatinine ratio is recommended as a screen for early diabetic nephropathy and other kidney diseases. And a positive test result may also suggest increased risk of cardiovascular disease.6 Microalbuminuria is defined as persistent albumin excretion between 30 and 300 mg/d.7
When exercise is a factor, here’s what to look for
As noted earlier, exercise-induced proteinuria is a function of the intensity of the exercise. Moderate and strenuous (vigorous) exercise are the 2 types of exercise that come into play when discussing proteinuria. Differentiating them is not precise, but is often defined by maximal oxygen consumption (vigorous=60% of VO2max; moderate <60% VO2max); metabolic equivalents (vigorous=6 METS; moderate <6 METS); walking/running speeds (various); and heart rate reserve (vigorous=60% HRR; moderate <60% HRR).8
Moderate exercise produces glomerular proteinuria, with an increase in macromolecular (albumin) filtration across the glomerular barrier. Strenuous exercise increases glomerular filtration of low-molecular-weight proteins (β2-microglobulin), which overwhelm the reabsorbing capacity of the tubular apparatus, causing temporary dysfunction and tubular proteinuria.9 Thus, the pathophysiology is mixed, with a major contribution from glomerular proteinuria.10
Strenuous exercise can cause protein excretion to exceed 1.5 mg/min.11 However, it seldom rises beyond 1 to 2 g/d,4 and this increase usually reverts to normal physiologic levels within 24 to 48 hours after exercise.12
Exercise-induced proteinuria is biphasic.13 Increased protein excretion occurs 30 minutes after exercise and is related to changes in intraglomerular hemodynamics and the resulting saturation of the renal tubules. Around 24 hours after exercise, oxidative stress on the glomeruli causes another slight elevation in albumin excretion without changes in β2-microglobulin, thereby indicating glomerular proteinuria exclusively.
Even the pros aren’t exempt. Exercise-induced proteinuria does not decrease with regular physical training. This was demonstrated in a study of 10 well-trained professional cyclists for whom strenuous exercise increased overnight protein excretion of both tubular and glomerular origin despite ongoing regular physical training.14
Creatine supplements do not increase proteinuria. A study of creatine supplementation in animal models noted no changes in 24-hour proteinuria or albumin excretion in both normal and two-thirds-nephrectomized animals.15 Another study compared creatine use with nonuse in athletes who had been training regularly and strenuously (12- 18 h/wk) for 5 to 10 years. They were evaluated for 10 months to 5 years. The groups exhibited equivalent urine excretion rates for albumin and creatinine, with no deleterious effect on kidney function.16
What happens when chronic disease is factored into the exercise equation?
Patients with a 2- to 20-year history of insulin-dependent diabetes without chronic kidney disease (CKD) who exhibited normal albumin excretion at baseline were more likely to develop proteinuria after exercise than healthy controls.17,18 The postulated cause was undetected glomerular changes due to diabetes. An exercise-provocation test may one day be useful in predicting future development of nephropathy, but further studies are needed.19-21
Exercise increases proteinuria immediately in individuals with metabolic disorders like obesity, through a mechanism different from diabetes mellitus. Proteinuria in the obese population is thought to be glomerular in origin, as opposed to both tubular and glomerular proteinuria in diabetic nephropathy.22,23
In CKD, low-intensity exercise long term does not promote proteinuria or lead to rapid progression of CKD. In one study, obese patients (body mass index >30 kg/m2) with diabetes and CKD stage II to IV who exercised 3 times weekly (aerobic training for 6 weeks, followed by 18 weeks of supervised home exercise) increased their stamina and exhibited slight, statistically insignificant decreases in resting systolic blood pressure and 24-hour proteinuria.24 A 12-week low-intensity aquatic exercise program for 26 patients with mild to moderate CKD decreased blood pressure and proteinuria and slightly improved glomerular filtration rate (GFR).25 These results for proteinuria and GFR were shown previously in rats with subtotal nephrectomy.26
Elevated urinary albumin excretion with exercise is significantly higher in patients with acromegaly when compared with normal healthy subjects. The underlying pathology is thought to occur at the glomerular filtration barrier with intact tubular function. Somatostatin analog treatment for acromegaly leads to reductions in postexercise albuminuria.27,28
How to manage suspected exercise-induced proteinuria
When interpreting the meaning of proteinuria detected on routine urinalysis, keep in mind the temporal relevance between exercise and urine collection. If urine is found to have been collected within 24 hours of intense exercise, repeat testing in the absence of prior exercise on at least one other occasion to differentiate between transient and persistent proteinuria. In confirming transient proteinuria after exercise, reassure the patient that it is a benign condition. This holds true as well for routine microalbumin-to-creatinine urine testing in patients with diabetes who exercise. If the result of a repeat test is high, turn your attention to another possible cause of proteinuria, such as diabetic nephropathy.
Screening for proteinuria during sports preparticipation examinations is not recommended because the diagnostic utility is low.29 Researchers performed urine dipstick testing for protein, blood, and glucose in preparticipation assessments of 701 students.29 They detected proteinuria in 40 students and glucosuria in one. Follow-up testing with first-voided morning urine specimens and glucose tolerance testing was normal in all students.
CORRESPONDENCE Fahad Saeed, MD, 313 Brook Hollow, Hanover, NH 03755; [email protected]
1. Poortmans JR. Exercise and renal function. Sports Med. 1984;1:125-153.
2. Gebke KB. Genitourinary system. In: McKeag DB, Moeller JL, eds. ACSM’s Primary Care Sports Medicine. 2nd ed. Philadelphia, Pa: Lippincott Williams & Wilkins; 2007;234.-
3. Venkat KK. Proteinuria and microalbuminuria in adults: significance, evaluation, and treatment. South Med J. 2004;97:969-979.
4. Rose BD. Pathophysiology of Renal Disease. 2nd ed. New York, NY: McGraw-Hill; 1987;11-16.
5. Sesso R, Santos AP, Nishida SK, et al. Prediction of steroid responsiveness in the idiopathic nephrotic syndrome using urinary retinol-binding protein and beta-2-microglobulin. Ann Intern Med. 1992;116:905-909.
6. Levey AS, Coresh J, Balk E, et al. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139:137-147.
7. Family Practice Notebook Urine protein to creatinine ratio. Available at: http://www.fpnotebook.com/urology/lab/urnprtntcrtnrt.htm. Accessed August 9, 2011.
8. Swain DP, Franklin BA. Comparison of cardioprotective benefits of vigorous versus moderate intensity aerobic exercise. Am J Cardiol. 2006;97:141-147.
9. Poortmans JR, Labilloy D. The influence of work intensity on postexercise proteinuria. Eur J Appl Physiol Occup Physiol. 1988;57:260-263.
10. Estivi P, Urbino R, Tetta C, et al. Urinary protein excretion induced by exercise: effect of a mountain agonistic footrace in healthy subjects. Renal function and mountain footrace. J Sports Med Phys Fitness. 1992;32:196-200.
11. Poortmans JR, Brauman H, Staroukine M, et al. Indirect evidence of glomerular/tubular mixed-type postexercise proteinuria in healthy humans. Am J Physiol. 1988;254:F277-F283.
12. Heathcote KL, Wilson MP, Quest DW, et al. Prevalence and duration of exercise induced albuminuria in healthy people. Clin Invest Med. 2009;32:E261-E265.
13. Sentürk UK, Kuru O, Koçer G, et al. Biphasic pattern of exercise-induced proteinuria in sedentary and trained men. Nephron Physiol. 2007;105:22-32.
14. Clerico A, Giammattei C, Cecchini L, et al. Exercise-induced proteinuria in well-trained athletes. Clin Chem. 1990;36:562-564.
15. Taes YE, Delanghe JR, Wuyts B, et al. Creatine supplementation does not affect kidney function in an animal model with pre-existing renal failure. Nephrol Dial Transplant. 2003;18:258-264.
16. Poortmans JR, Francaux M. Long-term oral creatine supplementation does not impair renal function in healthy athletes. Med Sci Sports Exerc. 1999;31:1108-1110.
17. Mogensen CE, Vittinghus E, Sølling K. Abnormal albumin excretion after two provocative renal tests in diabetes: physical exercise and lysine injection. Kidney Int. 1979;16:385-393.
18. Vittinghus E, Mogensen CE. Albumin excretion during physical exercise in diabetes. Studies on the effect of insulin treatment and of the renal haemodynamic response. Acta Endocrinol Suppl (Copenh). 1981;242:61-62.
19. Watts GF, Williams I, Morris RW, et al. An acceptable exercise test to study microalbuminuria in type 1 diabetes. Diabet Med. 1989;6:787-792.
20. Pan X, Wang P, Hu N, et al. A physiologically based pharmacokinetic model characterizing mechanism-based inhibition of CYP1A2 for predicting theophylline/antofloxacin interaction in both rats and humans. Drug Metab Pharmacokinet. 2011;26:387-398.
21. O’Brien SF, Watts GF, Powrie JK, et al. Exercise testing as a long-term predictor of the development of microalbuminuria in normoalbuminuric IDDM patients. Diabetes Care. 1995;18:1602-1605.
22. Hidaka S, Kaneko O, Shirai M, et al. Do obesity and non-insulin dependent diabetes mellitus aggravate exercise-induced microproteinuria? Clin Chim Acta. 1998;275:115-126.
23. Hidaka S, Kakuta S, Okada H, et al. Exercise-induced proteinuria in diseases with metabolic disorders. Contrib Nephrol. 1990;83:136-143.
24. Leehey DJ, Moinuddin I, Bast JP, et al. Aerobic exercise in obese diabetic patients with chronic kidney disease: a randomized and controlled pilot study. Cardiovasc Diabetol. 2009;8:62.-
25. Pechter U, Ots M, Mesikepp S, et al. Beneficial effects of water-based exercise in patients with chronic kidney disease. Int J Rehabil Res. 2003;26:153-156.
26. Heifets M, Davis TA, Tegtmeyer E, et al. Exercise training ameliorates progressive renal disease in rats with subtotal nephrectomy. Kidney Int. 1987;32:815-820.
27. Manelli F, Bossoni S, Burattin A, et al. Exercise-induced microalbuminuria in patients with active acromegaly: acute effects of slow-release lanreotide, a long-acting somatostatin analog. Metabolism. 2000;49:634-639.
28. Hoogenberg K, Sluiter WJ, Dullaart RP. Effect of growth hormone and insulin-like growth factor I on urinary albumin excretion: studies in acromegaly and growth hormone deficiency. Acta Endocrinol (Copenh). 1993;129:151-157.
29. Goldberg B, Saraniti A, Witman P, et al. Pre-participation sports assessment—an objective evaluation. Pediatrics. 1980;66:736-745.
30. Garrett WE, Kirkendall DT, Squire DL. eds. Principles and Practice of Primary Care Sports Medicine. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001;299-310.
31. Cosenzi A, Carraro M, Sacerdote A, et al. Involvement of the renin angiotensin system in the pathogenesis of postexercise proteinuria. Scand J Urol Nephrol. 1993;27:301-304.
32. Székács B, Vajo Z, Dachman W. Effect of ACE inhibition by benazepril, enalapril and captopril on chronic and post exercise proteinuria. Acta Physiol Hung. 1996;84:361-367.
33. Poortmans JR, Haggenmacher C, Vanderstraeten J. Postexercise proteinuria in humans and its adrenergic component. J Sports Med Phys Fitness. 2001;41:95-100.
1. Poortmans JR. Exercise and renal function. Sports Med. 1984;1:125-153.
2. Gebke KB. Genitourinary system. In: McKeag DB, Moeller JL, eds. ACSM’s Primary Care Sports Medicine. 2nd ed. Philadelphia, Pa: Lippincott Williams & Wilkins; 2007;234.-
3. Venkat KK. Proteinuria and microalbuminuria in adults: significance, evaluation, and treatment. South Med J. 2004;97:969-979.
4. Rose BD. Pathophysiology of Renal Disease. 2nd ed. New York, NY: McGraw-Hill; 1987;11-16.
5. Sesso R, Santos AP, Nishida SK, et al. Prediction of steroid responsiveness in the idiopathic nephrotic syndrome using urinary retinol-binding protein and beta-2-microglobulin. Ann Intern Med. 1992;116:905-909.
6. Levey AS, Coresh J, Balk E, et al. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139:137-147.
7. Family Practice Notebook Urine protein to creatinine ratio. Available at: http://www.fpnotebook.com/urology/lab/urnprtntcrtnrt.htm. Accessed August 9, 2011.
8. Swain DP, Franklin BA. Comparison of cardioprotective benefits of vigorous versus moderate intensity aerobic exercise. Am J Cardiol. 2006;97:141-147.
9. Poortmans JR, Labilloy D. The influence of work intensity on postexercise proteinuria. Eur J Appl Physiol Occup Physiol. 1988;57:260-263.
10. Estivi P, Urbino R, Tetta C, et al. Urinary protein excretion induced by exercise: effect of a mountain agonistic footrace in healthy subjects. Renal function and mountain footrace. J Sports Med Phys Fitness. 1992;32:196-200.
11. Poortmans JR, Brauman H, Staroukine M, et al. Indirect evidence of glomerular/tubular mixed-type postexercise proteinuria in healthy humans. Am J Physiol. 1988;254:F277-F283.
12. Heathcote KL, Wilson MP, Quest DW, et al. Prevalence and duration of exercise induced albuminuria in healthy people. Clin Invest Med. 2009;32:E261-E265.
13. Sentürk UK, Kuru O, Koçer G, et al. Biphasic pattern of exercise-induced proteinuria in sedentary and trained men. Nephron Physiol. 2007;105:22-32.
14. Clerico A, Giammattei C, Cecchini L, et al. Exercise-induced proteinuria in well-trained athletes. Clin Chem. 1990;36:562-564.
15. Taes YE, Delanghe JR, Wuyts B, et al. Creatine supplementation does not affect kidney function in an animal model with pre-existing renal failure. Nephrol Dial Transplant. 2003;18:258-264.
16. Poortmans JR, Francaux M. Long-term oral creatine supplementation does not impair renal function in healthy athletes. Med Sci Sports Exerc. 1999;31:1108-1110.
17. Mogensen CE, Vittinghus E, Sølling K. Abnormal albumin excretion after two provocative renal tests in diabetes: physical exercise and lysine injection. Kidney Int. 1979;16:385-393.
18. Vittinghus E, Mogensen CE. Albumin excretion during physical exercise in diabetes. Studies on the effect of insulin treatment and of the renal haemodynamic response. Acta Endocrinol Suppl (Copenh). 1981;242:61-62.
19. Watts GF, Williams I, Morris RW, et al. An acceptable exercise test to study microalbuminuria in type 1 diabetes. Diabet Med. 1989;6:787-792.
20. Pan X, Wang P, Hu N, et al. A physiologically based pharmacokinetic model characterizing mechanism-based inhibition of CYP1A2 for predicting theophylline/antofloxacin interaction in both rats and humans. Drug Metab Pharmacokinet. 2011;26:387-398.
21. O’Brien SF, Watts GF, Powrie JK, et al. Exercise testing as a long-term predictor of the development of microalbuminuria in normoalbuminuric IDDM patients. Diabetes Care. 1995;18:1602-1605.
22. Hidaka S, Kaneko O, Shirai M, et al. Do obesity and non-insulin dependent diabetes mellitus aggravate exercise-induced microproteinuria? Clin Chim Acta. 1998;275:115-126.
23. Hidaka S, Kakuta S, Okada H, et al. Exercise-induced proteinuria in diseases with metabolic disorders. Contrib Nephrol. 1990;83:136-143.
24. Leehey DJ, Moinuddin I, Bast JP, et al. Aerobic exercise in obese diabetic patients with chronic kidney disease: a randomized and controlled pilot study. Cardiovasc Diabetol. 2009;8:62.-
25. Pechter U, Ots M, Mesikepp S, et al. Beneficial effects of water-based exercise in patients with chronic kidney disease. Int J Rehabil Res. 2003;26:153-156.
26. Heifets M, Davis TA, Tegtmeyer E, et al. Exercise training ameliorates progressive renal disease in rats with subtotal nephrectomy. Kidney Int. 1987;32:815-820.
27. Manelli F, Bossoni S, Burattin A, et al. Exercise-induced microalbuminuria in patients with active acromegaly: acute effects of slow-release lanreotide, a long-acting somatostatin analog. Metabolism. 2000;49:634-639.
28. Hoogenberg K, Sluiter WJ, Dullaart RP. Effect of growth hormone and insulin-like growth factor I on urinary albumin excretion: studies in acromegaly and growth hormone deficiency. Acta Endocrinol (Copenh). 1993;129:151-157.
29. Goldberg B, Saraniti A, Witman P, et al. Pre-participation sports assessment—an objective evaluation. Pediatrics. 1980;66:736-745.
30. Garrett WE, Kirkendall DT, Squire DL. eds. Principles and Practice of Primary Care Sports Medicine. Philadelphia, Pa: Lippincott Williams & Wilkins; 2001;299-310.
31. Cosenzi A, Carraro M, Sacerdote A, et al. Involvement of the renin angiotensin system in the pathogenesis of postexercise proteinuria. Scand J Urol Nephrol. 1993;27:301-304.
32. Székács B, Vajo Z, Dachman W. Effect of ACE inhibition by benazepril, enalapril and captopril on chronic and post exercise proteinuria. Acta Physiol Hung. 1996;84:361-367.
33. Poortmans JR, Haggenmacher C, Vanderstraeten J. Postexercise proteinuria in humans and its adrenergic component. J Sports Med Phys Fitness. 2001;41:95-100.
Urinary Dysfunction Stands Out on Retigabine's Safety Profile
BALTIMORE – The safety profile for retigabine is generally consistent with that of other antiepileptic drugs, but data from studies in its evaluation process show that it also carries a novel, small increased risk of urinary voiding dysfunction.
At the annual meeting of the American Epilepsy Society Dr. Neil Brickel of GlaxoSmithKline’s clinical safety and pharmacovigilance group in Middlesex, England, reported on the safety of the novel drug in three phase III, randomized, double-blind, placebo-controlled trials, four phase II studies, and six long-term open-label extension studies, totalling 1,365 patients.
Retigabine is the international nonproprietary name of the drug, but it was approved in the United States in June 2011 under the adopted name ezogabine as an adjunctive therapy in adults with partial-onset seizures. It is a first-in-class potassium channel opener manufactured by GlaxoSmithKline and Valeant Pharmaceuticals International, and sold under the brand name Trobalt in Europe and Potiga in the United States.
In all of the studies that Dr. Brickel analyzed, retigabine was evaluated as a second, add-on agent. As of Oct. 2, 2009, 1,217 of the 1,365 patients had been on treatment at least 1 month, 801 for at least 6 months, and 586 for 1 year or longer.
As expected with any antiepileptic drug (AED), the most frequent adverse events involved the central nervous system. The most common in the three pivotal controlled trials were dizziness (23% of retigabine-treated patients vs. 9% of placebo-treated patients) and somnolence (reported by 22% vs. 12%, respectively). The only CNS adverse events that were not dose related were headache (reported by 15% of the retigabine group vs. 16% of the placebo group) and fatigue (by 15% and 6%, respectively). Other adverse events occurring in more than 10% of subjects were confusional state, tremor, and abnormal coordination. For the most part, patients reported these adverse events early on, during the titration phases of the trials, Dr. Brickel said.
Adverse events led to discontinuation in 437 patients (32%) of the total phase II/III study population. Again, these were primarily CNS and dose related. Discontinuations for any event occurred in 11% of the placebo group. Among those taking retigabine, discontinuations occurred in 17% of those assigned to 600 mg, 25% of those assigned to 900 mg, and 31% of those assigned to 1,200 mg. The most common reason for discontinuation was dizziness.
"Two-thirds of patients were able to continue in the study, even with the fixed titration regimen and at the 1,200-mg dose," Dr. Brickel noted.
Serious adverse events (SAEs) were infrequent, and also typically CNS related. In the three pivotal controlled trials, SAEs occurred in 6% of the placebo group and 9% of the retigabine patients. Six of the patients taking retigabine developed a psychotic disorder (five with 1,200 mg, one with 900 mg), compared with none of the placebo patients. Convulsions occurred in 1.2% of the placebo group and in 1.5% of the retigabine patients, across all doses. This finding was not unexpected, considering the underlying epilepsy, he said.
No other SAE was reported in more than two patients in any group. Deaths occurred in three placebo patients and two patients in the retigabine group. One in each group was a sudden death.
In the three pivotal trials, urinary and renal-related adverse events occurred in 17% of retigabine patients, compared with 13% of the placebo-treated patients. In the entire phase II/III study population, 26% of the 1,365 patients reported a urinary adverse event.
However, events related to urinary voiding dysfunction, which have a plausible biological relation to retigabine’s pharmacology, occurred in less than 4% of retigabine-treated patients in the phase III trials alone and in all phase II and III trial populations combined. Urinary hesitation was the most common of these in the entire phase II and III study group, occurring in 3.1%, followed by urinary retention in 1.9%. Catheterization was required in four patients taking retigabine and in one patient on placebo.
"These all occurred in a smaller number of patients and provide a better guide as to how many patents are potentially at risk. It is also important to note that these events were for the most part ‘mild’ in intensity and the patients were able to continue treatment," Dr. Brickel said in an interview.
Dr. Brickel also said that GSK is "communicating these risks to potential prescribers and encouraging them to advise patients of the symptoms relating to difficulty in passing urine that could range from discomfort on bladder emptying, hesitancy, poor flow through to a complete inability to void (acute urinary retention). GSK is keen to proactively manage this."
The study was funded by GSK and Valeant Pharmaceuticals International, and all of the investigators are employees of either company.
BALTIMORE – The safety profile for retigabine is generally consistent with that of other antiepileptic drugs, but data from studies in its evaluation process show that it also carries a novel, small increased risk of urinary voiding dysfunction.
At the annual meeting of the American Epilepsy Society Dr. Neil Brickel of GlaxoSmithKline’s clinical safety and pharmacovigilance group in Middlesex, England, reported on the safety of the novel drug in three phase III, randomized, double-blind, placebo-controlled trials, four phase II studies, and six long-term open-label extension studies, totalling 1,365 patients.
Retigabine is the international nonproprietary name of the drug, but it was approved in the United States in June 2011 under the adopted name ezogabine as an adjunctive therapy in adults with partial-onset seizures. It is a first-in-class potassium channel opener manufactured by GlaxoSmithKline and Valeant Pharmaceuticals International, and sold under the brand name Trobalt in Europe and Potiga in the United States.
In all of the studies that Dr. Brickel analyzed, retigabine was evaluated as a second, add-on agent. As of Oct. 2, 2009, 1,217 of the 1,365 patients had been on treatment at least 1 month, 801 for at least 6 months, and 586 for 1 year or longer.
As expected with any antiepileptic drug (AED), the most frequent adverse events involved the central nervous system. The most common in the three pivotal controlled trials were dizziness (23% of retigabine-treated patients vs. 9% of placebo-treated patients) and somnolence (reported by 22% vs. 12%, respectively). The only CNS adverse events that were not dose related were headache (reported by 15% of the retigabine group vs. 16% of the placebo group) and fatigue (by 15% and 6%, respectively). Other adverse events occurring in more than 10% of subjects were confusional state, tremor, and abnormal coordination. For the most part, patients reported these adverse events early on, during the titration phases of the trials, Dr. Brickel said.
Adverse events led to discontinuation in 437 patients (32%) of the total phase II/III study population. Again, these were primarily CNS and dose related. Discontinuations for any event occurred in 11% of the placebo group. Among those taking retigabine, discontinuations occurred in 17% of those assigned to 600 mg, 25% of those assigned to 900 mg, and 31% of those assigned to 1,200 mg. The most common reason for discontinuation was dizziness.
"Two-thirds of patients were able to continue in the study, even with the fixed titration regimen and at the 1,200-mg dose," Dr. Brickel noted.
Serious adverse events (SAEs) were infrequent, and also typically CNS related. In the three pivotal controlled trials, SAEs occurred in 6% of the placebo group and 9% of the retigabine patients. Six of the patients taking retigabine developed a psychotic disorder (five with 1,200 mg, one with 900 mg), compared with none of the placebo patients. Convulsions occurred in 1.2% of the placebo group and in 1.5% of the retigabine patients, across all doses. This finding was not unexpected, considering the underlying epilepsy, he said.
No other SAE was reported in more than two patients in any group. Deaths occurred in three placebo patients and two patients in the retigabine group. One in each group was a sudden death.
In the three pivotal trials, urinary and renal-related adverse events occurred in 17% of retigabine patients, compared with 13% of the placebo-treated patients. In the entire phase II/III study population, 26% of the 1,365 patients reported a urinary adverse event.
However, events related to urinary voiding dysfunction, which have a plausible biological relation to retigabine’s pharmacology, occurred in less than 4% of retigabine-treated patients in the phase III trials alone and in all phase II and III trial populations combined. Urinary hesitation was the most common of these in the entire phase II and III study group, occurring in 3.1%, followed by urinary retention in 1.9%. Catheterization was required in four patients taking retigabine and in one patient on placebo.
"These all occurred in a smaller number of patients and provide a better guide as to how many patents are potentially at risk. It is also important to note that these events were for the most part ‘mild’ in intensity and the patients were able to continue treatment," Dr. Brickel said in an interview.
Dr. Brickel also said that GSK is "communicating these risks to potential prescribers and encouraging them to advise patients of the symptoms relating to difficulty in passing urine that could range from discomfort on bladder emptying, hesitancy, poor flow through to a complete inability to void (acute urinary retention). GSK is keen to proactively manage this."
The study was funded by GSK and Valeant Pharmaceuticals International, and all of the investigators are employees of either company.
BALTIMORE – The safety profile for retigabine is generally consistent with that of other antiepileptic drugs, but data from studies in its evaluation process show that it also carries a novel, small increased risk of urinary voiding dysfunction.
At the annual meeting of the American Epilepsy Society Dr. Neil Brickel of GlaxoSmithKline’s clinical safety and pharmacovigilance group in Middlesex, England, reported on the safety of the novel drug in three phase III, randomized, double-blind, placebo-controlled trials, four phase II studies, and six long-term open-label extension studies, totalling 1,365 patients.
Retigabine is the international nonproprietary name of the drug, but it was approved in the United States in June 2011 under the adopted name ezogabine as an adjunctive therapy in adults with partial-onset seizures. It is a first-in-class potassium channel opener manufactured by GlaxoSmithKline and Valeant Pharmaceuticals International, and sold under the brand name Trobalt in Europe and Potiga in the United States.
In all of the studies that Dr. Brickel analyzed, retigabine was evaluated as a second, add-on agent. As of Oct. 2, 2009, 1,217 of the 1,365 patients had been on treatment at least 1 month, 801 for at least 6 months, and 586 for 1 year or longer.
As expected with any antiepileptic drug (AED), the most frequent adverse events involved the central nervous system. The most common in the three pivotal controlled trials were dizziness (23% of retigabine-treated patients vs. 9% of placebo-treated patients) and somnolence (reported by 22% vs. 12%, respectively). The only CNS adverse events that were not dose related were headache (reported by 15% of the retigabine group vs. 16% of the placebo group) and fatigue (by 15% and 6%, respectively). Other adverse events occurring in more than 10% of subjects were confusional state, tremor, and abnormal coordination. For the most part, patients reported these adverse events early on, during the titration phases of the trials, Dr. Brickel said.
Adverse events led to discontinuation in 437 patients (32%) of the total phase II/III study population. Again, these were primarily CNS and dose related. Discontinuations for any event occurred in 11% of the placebo group. Among those taking retigabine, discontinuations occurred in 17% of those assigned to 600 mg, 25% of those assigned to 900 mg, and 31% of those assigned to 1,200 mg. The most common reason for discontinuation was dizziness.
"Two-thirds of patients were able to continue in the study, even with the fixed titration regimen and at the 1,200-mg dose," Dr. Brickel noted.
Serious adverse events (SAEs) were infrequent, and also typically CNS related. In the three pivotal controlled trials, SAEs occurred in 6% of the placebo group and 9% of the retigabine patients. Six of the patients taking retigabine developed a psychotic disorder (five with 1,200 mg, one with 900 mg), compared with none of the placebo patients. Convulsions occurred in 1.2% of the placebo group and in 1.5% of the retigabine patients, across all doses. This finding was not unexpected, considering the underlying epilepsy, he said.
No other SAE was reported in more than two patients in any group. Deaths occurred in three placebo patients and two patients in the retigabine group. One in each group was a sudden death.
In the three pivotal trials, urinary and renal-related adverse events occurred in 17% of retigabine patients, compared with 13% of the placebo-treated patients. In the entire phase II/III study population, 26% of the 1,365 patients reported a urinary adverse event.
However, events related to urinary voiding dysfunction, which have a plausible biological relation to retigabine’s pharmacology, occurred in less than 4% of retigabine-treated patients in the phase III trials alone and in all phase II and III trial populations combined. Urinary hesitation was the most common of these in the entire phase II and III study group, occurring in 3.1%, followed by urinary retention in 1.9%. Catheterization was required in four patients taking retigabine and in one patient on placebo.
"These all occurred in a smaller number of patients and provide a better guide as to how many patents are potentially at risk. It is also important to note that these events were for the most part ‘mild’ in intensity and the patients were able to continue treatment," Dr. Brickel said in an interview.
Dr. Brickel also said that GSK is "communicating these risks to potential prescribers and encouraging them to advise patients of the symptoms relating to difficulty in passing urine that could range from discomfort on bladder emptying, hesitancy, poor flow through to a complete inability to void (acute urinary retention). GSK is keen to proactively manage this."
The study was funded by GSK and Valeant Pharmaceuticals International, and all of the investigators are employees of either company.
FROM THE ANNUAL MEETING OF THE AMERICAN EPILEPSY SOCIETY
Major Finding: Urinary hesitation occurred in 3.1% of the entire phase II/III study group, and urinary retention occurred in 1.9%.
Data Source: Safety data collected from three phase III, randomized, double-blind, placebo-controlled trials, as well as from four phase II studies and six long-term open-label extension studies, involving 1,365 patients in all.
Disclosures: The study was funded by GlaxoSmithKline and Valeant Pharmaceuticals International. All of the investigators are employees of either company.
Combining MRI With Prostate Ultrasound Biopsy Bests Biopsy Alone
CHICAGO – Fusing MRI with real-time, three-dimensional ultrasound allows for more targeted prostate biopsies and finds additional cancers, compared with standard systematic biopsies.
"This may lead to fewer total biopsies, improved yield and improved confidence for active surveillance," Dr. Daniel J.A. Margolis said at the annual meeting of the Radiological Society of North America.
Direct MRI-guided biopsy is not universally available, leaving most centers to use two-dimensional ultrasound to systematically biopsy 12 areas of the prostate, whether they are all suspicious or not. Not surprising, roughly 30% of systematic core biopsies are false negative, explained radiologist Dr. Margolis of the University of California, Los Angeles.
Researchers at UCLA departments and the medical device company Eigen have been using external-array 3 Tesla MRI scans, including T2-weighted, diffusion-weighted, and dynamic contrast-enhanced images to identify suspicious areas in the prostate. The areas are scored on a 5-point scale by a radiologist based on cancer risk, and the data are used to create a 3-D contoured reconstruction that is fused with real-time, transrectal ultrasound during biopsy.
Early results were promising in the two groups of men most likely to benefit from the new imaging technology – those with a prior negative biopsy and elevated prostate-specific antigen (PSA) levels and those with low-risk prostate cancer under active surveillance. In 47 such men, the biopsy-positivity rate was 33% with MRI-fusion ultrasound vs. 7% for systematic, nontargeted biopsy (Urol. Oncol. 2011;29:334-42).
At the meeting, Dr. Margolis presented data from 57 consecutive men with a previous biopsy, in whom MRI-fusion ultrasound identified 101 suspicious areas. In all, 22 men had 28 positive MRI targets.
Positive biopsies were found in 12 patients on targets only. Nine patients had positive lesions on both MRI-fusion ultrasound and systematic biopsy. In one additional patient, the positive systematic core was changed from Gleason 3+3 to 3+4 disease with the targeted biopsy.
Seven patients had positive biopsies found on systematic biopsy only, although all were Gleason score 3+3, less than 4 mm in size and less than 25% of the core, Dr. Margolis said.
A separate study presented in the same session suggests that fusing MRI with transrectal ultrasound biopsy may also be useful in identifying aggressive tumors in men with no prior prostate biopsy or suspicious digital rectal exam and a PSA of 3-10 ng/mL (mean 8 ng/mL).
The overall cancer detection rate was 52% among 323 (168/323) such men, 73% within MRI targets (144/197) and 19% with sextant biopsy (24/126), reported Dr. François Cornud, a consultant radiologist at René Descartes University, Paris.
In 98 patients with both MRI targeted- and sextant-positive biopsies, targeted biopsies identified significantly more cancers with a Gleason score greater than 6 (44% vs. 25%), with a length in any core of more than 7 mm (50% vs. 25.5%) and with a longer mean length (5.3 mm vs. 0.8 mm).
Interestingly, performance was similar whether the multiparametric MRI data were fused with the real-time ultrasound images visually or by a computerized electromagnetic navigator system.
"Targeted biopsies definitely provide better evaluation of tumor burden and Gleason score," Dr. Cornud said, adding that "a negative MRI prior to biopsy may mean no cancer or indolent cancer and may suggest that in these patients biopsy may be deferred."
Both studies were well received, although one attendee questioned whether the researchers have been able to convince frequently reluctant urologists that targeted biopsies are worth it. Dr. Margolis said his project was actually instigated by an urologist. Dr. Cornud said the majority of his urologists are now requesting an MRI and its findings.
Dr. Margolis reported a research grant from Siemens AG and a coauthor reported conflicts with several pharmaceutical and device firms. Dr. Cornud and his coauthors reported no relevant disclosures.
Direct MRI-guided biopsy, prostate, Eigen, prior negative biopsy, elevated prostate-specific antigen (PSA) levels,
CHICAGO – Fusing MRI with real-time, three-dimensional ultrasound allows for more targeted prostate biopsies and finds additional cancers, compared with standard systematic biopsies.
"This may lead to fewer total biopsies, improved yield and improved confidence for active surveillance," Dr. Daniel J.A. Margolis said at the annual meeting of the Radiological Society of North America.
Direct MRI-guided biopsy is not universally available, leaving most centers to use two-dimensional ultrasound to systematically biopsy 12 areas of the prostate, whether they are all suspicious or not. Not surprising, roughly 30% of systematic core biopsies are false negative, explained radiologist Dr. Margolis of the University of California, Los Angeles.
Researchers at UCLA departments and the medical device company Eigen have been using external-array 3 Tesla MRI scans, including T2-weighted, diffusion-weighted, and dynamic contrast-enhanced images to identify suspicious areas in the prostate. The areas are scored on a 5-point scale by a radiologist based on cancer risk, and the data are used to create a 3-D contoured reconstruction that is fused with real-time, transrectal ultrasound during biopsy.
Early results were promising in the two groups of men most likely to benefit from the new imaging technology – those with a prior negative biopsy and elevated prostate-specific antigen (PSA) levels and those with low-risk prostate cancer under active surveillance. In 47 such men, the biopsy-positivity rate was 33% with MRI-fusion ultrasound vs. 7% for systematic, nontargeted biopsy (Urol. Oncol. 2011;29:334-42).
At the meeting, Dr. Margolis presented data from 57 consecutive men with a previous biopsy, in whom MRI-fusion ultrasound identified 101 suspicious areas. In all, 22 men had 28 positive MRI targets.
Positive biopsies were found in 12 patients on targets only. Nine patients had positive lesions on both MRI-fusion ultrasound and systematic biopsy. In one additional patient, the positive systematic core was changed from Gleason 3+3 to 3+4 disease with the targeted biopsy.
Seven patients had positive biopsies found on systematic biopsy only, although all were Gleason score 3+3, less than 4 mm in size and less than 25% of the core, Dr. Margolis said.
A separate study presented in the same session suggests that fusing MRI with transrectal ultrasound biopsy may also be useful in identifying aggressive tumors in men with no prior prostate biopsy or suspicious digital rectal exam and a PSA of 3-10 ng/mL (mean 8 ng/mL).
The overall cancer detection rate was 52% among 323 (168/323) such men, 73% within MRI targets (144/197) and 19% with sextant biopsy (24/126), reported Dr. François Cornud, a consultant radiologist at René Descartes University, Paris.
In 98 patients with both MRI targeted- and sextant-positive biopsies, targeted biopsies identified significantly more cancers with a Gleason score greater than 6 (44% vs. 25%), with a length in any core of more than 7 mm (50% vs. 25.5%) and with a longer mean length (5.3 mm vs. 0.8 mm).
Interestingly, performance was similar whether the multiparametric MRI data were fused with the real-time ultrasound images visually or by a computerized electromagnetic navigator system.
"Targeted biopsies definitely provide better evaluation of tumor burden and Gleason score," Dr. Cornud said, adding that "a negative MRI prior to biopsy may mean no cancer or indolent cancer and may suggest that in these patients biopsy may be deferred."
Both studies were well received, although one attendee questioned whether the researchers have been able to convince frequently reluctant urologists that targeted biopsies are worth it. Dr. Margolis said his project was actually instigated by an urologist. Dr. Cornud said the majority of his urologists are now requesting an MRI and its findings.
Dr. Margolis reported a research grant from Siemens AG and a coauthor reported conflicts with several pharmaceutical and device firms. Dr. Cornud and his coauthors reported no relevant disclosures.
CHICAGO – Fusing MRI with real-time, three-dimensional ultrasound allows for more targeted prostate biopsies and finds additional cancers, compared with standard systematic biopsies.
"This may lead to fewer total biopsies, improved yield and improved confidence for active surveillance," Dr. Daniel J.A. Margolis said at the annual meeting of the Radiological Society of North America.
Direct MRI-guided biopsy is not universally available, leaving most centers to use two-dimensional ultrasound to systematically biopsy 12 areas of the prostate, whether they are all suspicious or not. Not surprising, roughly 30% of systematic core biopsies are false negative, explained radiologist Dr. Margolis of the University of California, Los Angeles.
Researchers at UCLA departments and the medical device company Eigen have been using external-array 3 Tesla MRI scans, including T2-weighted, diffusion-weighted, and dynamic contrast-enhanced images to identify suspicious areas in the prostate. The areas are scored on a 5-point scale by a radiologist based on cancer risk, and the data are used to create a 3-D contoured reconstruction that is fused with real-time, transrectal ultrasound during biopsy.
Early results were promising in the two groups of men most likely to benefit from the new imaging technology – those with a prior negative biopsy and elevated prostate-specific antigen (PSA) levels and those with low-risk prostate cancer under active surveillance. In 47 such men, the biopsy-positivity rate was 33% with MRI-fusion ultrasound vs. 7% for systematic, nontargeted biopsy (Urol. Oncol. 2011;29:334-42).
At the meeting, Dr. Margolis presented data from 57 consecutive men with a previous biopsy, in whom MRI-fusion ultrasound identified 101 suspicious areas. In all, 22 men had 28 positive MRI targets.
Positive biopsies were found in 12 patients on targets only. Nine patients had positive lesions on both MRI-fusion ultrasound and systematic biopsy. In one additional patient, the positive systematic core was changed from Gleason 3+3 to 3+4 disease with the targeted biopsy.
Seven patients had positive biopsies found on systematic biopsy only, although all were Gleason score 3+3, less than 4 mm in size and less than 25% of the core, Dr. Margolis said.
A separate study presented in the same session suggests that fusing MRI with transrectal ultrasound biopsy may also be useful in identifying aggressive tumors in men with no prior prostate biopsy or suspicious digital rectal exam and a PSA of 3-10 ng/mL (mean 8 ng/mL).
The overall cancer detection rate was 52% among 323 (168/323) such men, 73% within MRI targets (144/197) and 19% with sextant biopsy (24/126), reported Dr. François Cornud, a consultant radiologist at René Descartes University, Paris.
In 98 patients with both MRI targeted- and sextant-positive biopsies, targeted biopsies identified significantly more cancers with a Gleason score greater than 6 (44% vs. 25%), with a length in any core of more than 7 mm (50% vs. 25.5%) and with a longer mean length (5.3 mm vs. 0.8 mm).
Interestingly, performance was similar whether the multiparametric MRI data were fused with the real-time ultrasound images visually or by a computerized electromagnetic navigator system.
"Targeted biopsies definitely provide better evaluation of tumor burden and Gleason score," Dr. Cornud said, adding that "a negative MRI prior to biopsy may mean no cancer or indolent cancer and may suggest that in these patients biopsy may be deferred."
Both studies were well received, although one attendee questioned whether the researchers have been able to convince frequently reluctant urologists that targeted biopsies are worth it. Dr. Margolis said his project was actually instigated by an urologist. Dr. Cornud said the majority of his urologists are now requesting an MRI and its findings.
Dr. Margolis reported a research grant from Siemens AG and a coauthor reported conflicts with several pharmaceutical and device firms. Dr. Cornud and his coauthors reported no relevant disclosures.
Direct MRI-guided biopsy, prostate, Eigen, prior negative biopsy, elevated prostate-specific antigen (PSA) levels,
Direct MRI-guided biopsy, prostate, Eigen, prior negative biopsy, elevated prostate-specific antigen (PSA) levels,
FROM THE ANNUAL MEETING OF THE RADIOLOGICAL SOCIETY OF NORTH AMERICA
Major Finding: Among 57 consecutive men with a previous biopsy in whom MRI-fusion ultrasound identified 101 suspicious areas, 22 men had 28 positive MRI targets. In a second study, fusing MRI with transrectal ultrasound biopsy was useful in identifying aggressive tumors in men with no prior prostate biopsy or suspicious digital rectal exam and a PSA of 3-10 ng/mL. The overall cancer detection rate was 52% among 323 (168/323) such men, 73% within MRI targets (144/197) and 19% with sextant biopsy (24/126).
Data Source: Prospective study in 57 men with a prior prostate biopsy, and a prospective study in 323 men with no prior biopsy and PSA levels of 3-10 ng/mL.
Disclosures: Dr. Margolis reported a research grant from Siemens AG, and a coauthor reported conflicts with several pharmaceutical and device firms. Dr. Cornud and his coauthors reported no relevant disclosures.
FDA Panel Votes on Peginesatide for Some Anemia
SILVER SPRING, MD. – The majority of a Food and Drug Administration advisory panel on Dec. 7 voted that peginesatide, a new erythropoiesis-stimulating agent, has a favorable risk-benefit profile when used to treat anemia associated with chronic renal failure in patients who are on dialysis.
At a meeting of the FDA’s Oncologic Drugs Advisory Committee, it voted 15-1 with one abstention on the risk-benefit question. One of the two cardiologists on the panel voted no; the other abstained. The panel did not vote specifically on whether to recommend approval.
Affymax Inc., the manufacturer of peginesatide – a long-acting erythropoiesis-stimulating agent (ESA) administered intravenously or subcutaneously – has proposed that it be approved for the treatment of anemia associated with chronic kidney disease (CKD) in adults who are on dialysis, but not in CKD patients who are not on dialysis or in cancer patients. Peginesatide is administered once a month, which is less frequently than the two ESAs approved in the United States: epoetin alfa, marketed as Epogen and Procrit, and darbepoetin alfa, marketed as Aranesp. A pegylated epoetin beta (Mircera) is not available in the United States.
In two phase III studies of patients with anemia due to CKD who were on dialysis, 1,066 subjects were treated with peginesatide once a month and 542 were treated with epoetin administered one to three times a week. The effectiveness of peginesatide in maintaining hemoglobin levels at 10-12 g/dL through weeks 29-36 of treatment was similar to that of epoetin, with similar low transfusion rates in both groups. The safety profiles, including serious adverse events, deaths during the study, and adverse events resulting in permanent discontinuation of treatment, were similar in both treatment groups.
But in two phase III studies of patients with anemia due to CKD who were not on dialysis, peginesatide appeared to be less safe than darbepoetin. In these two studies, the rates of adverse events, serious adverse events, deaths during the study, adverse events leading to discontinuation as well as adverse cardiovascular outcomes were higher among those on peginesatide than among those on darbepoetin.
Panelists expressed concerned about this safety signal in the patients not on dialysis, but those who voted in favor of peginesatide for the proposed indication agreed that the safety and efficacy of peginesatide were comparable with epoetin in two large randomized studies and cited the convenience of once-a-month dosing for patients. They recommended postmarketing follow-up of these patients to provide longer-term data, since the average length of ESA treatment in patients with CKD is about 5 years and patients in the studies were followed for a little more than 1 year.
The two cardiologists on the panel cited concerns about the studies not being blinded and the possibility that the differences in toxicity in the two patient groups might not be explained pathophysiologically but instead might be the result of stricter entry criteria in the study of patients on dialysis. They also cited the potential for misuse of these drugs in patients.
Targeting higher hemoglobin levels with ESA treatment has been associated with an increased risk of MI and other adverse cardiovascular outcomes in several large clinical trials, which led to changes in treatment recommendations.
In 2007, a boxed warning about the increased risk of deaths and serious cardiovascular events when higher hemoglobin levels are targeted was added to the labels of ESAs, with the recommendation to individualize dosing to target and maintain hemoglobin levels within the 10-12 g/dL range. In 2009, a warning about the increased risk of stroke based on the results of a study of patients with diabetes and CKD who were not on dialysis was added to ESA labels.
The FDA usually follows the recommendations of its advisory panels, which are not binding. Panelists have been cleared of potential conflicts of interest related to the topic of the meeting. Occasionally, a panelist may be given a waiver, but not at this meeting.
SILVER SPRING, MD. – The majority of a Food and Drug Administration advisory panel on Dec. 7 voted that peginesatide, a new erythropoiesis-stimulating agent, has a favorable risk-benefit profile when used to treat anemia associated with chronic renal failure in patients who are on dialysis.
At a meeting of the FDA’s Oncologic Drugs Advisory Committee, it voted 15-1 with one abstention on the risk-benefit question. One of the two cardiologists on the panel voted no; the other abstained. The panel did not vote specifically on whether to recommend approval.
Affymax Inc., the manufacturer of peginesatide – a long-acting erythropoiesis-stimulating agent (ESA) administered intravenously or subcutaneously – has proposed that it be approved for the treatment of anemia associated with chronic kidney disease (CKD) in adults who are on dialysis, but not in CKD patients who are not on dialysis or in cancer patients. Peginesatide is administered once a month, which is less frequently than the two ESAs approved in the United States: epoetin alfa, marketed as Epogen and Procrit, and darbepoetin alfa, marketed as Aranesp. A pegylated epoetin beta (Mircera) is not available in the United States.
In two phase III studies of patients with anemia due to CKD who were on dialysis, 1,066 subjects were treated with peginesatide once a month and 542 were treated with epoetin administered one to three times a week. The effectiveness of peginesatide in maintaining hemoglobin levels at 10-12 g/dL through weeks 29-36 of treatment was similar to that of epoetin, with similar low transfusion rates in both groups. The safety profiles, including serious adverse events, deaths during the study, and adverse events resulting in permanent discontinuation of treatment, were similar in both treatment groups.
But in two phase III studies of patients with anemia due to CKD who were not on dialysis, peginesatide appeared to be less safe than darbepoetin. In these two studies, the rates of adverse events, serious adverse events, deaths during the study, adverse events leading to discontinuation as well as adverse cardiovascular outcomes were higher among those on peginesatide than among those on darbepoetin.
Panelists expressed concerned about this safety signal in the patients not on dialysis, but those who voted in favor of peginesatide for the proposed indication agreed that the safety and efficacy of peginesatide were comparable with epoetin in two large randomized studies and cited the convenience of once-a-month dosing for patients. They recommended postmarketing follow-up of these patients to provide longer-term data, since the average length of ESA treatment in patients with CKD is about 5 years and patients in the studies were followed for a little more than 1 year.
The two cardiologists on the panel cited concerns about the studies not being blinded and the possibility that the differences in toxicity in the two patient groups might not be explained pathophysiologically but instead might be the result of stricter entry criteria in the study of patients on dialysis. They also cited the potential for misuse of these drugs in patients.
Targeting higher hemoglobin levels with ESA treatment has been associated with an increased risk of MI and other adverse cardiovascular outcomes in several large clinical trials, which led to changes in treatment recommendations.
In 2007, a boxed warning about the increased risk of deaths and serious cardiovascular events when higher hemoglobin levels are targeted was added to the labels of ESAs, with the recommendation to individualize dosing to target and maintain hemoglobin levels within the 10-12 g/dL range. In 2009, a warning about the increased risk of stroke based on the results of a study of patients with diabetes and CKD who were not on dialysis was added to ESA labels.
The FDA usually follows the recommendations of its advisory panels, which are not binding. Panelists have been cleared of potential conflicts of interest related to the topic of the meeting. Occasionally, a panelist may be given a waiver, but not at this meeting.
SILVER SPRING, MD. – The majority of a Food and Drug Administration advisory panel on Dec. 7 voted that peginesatide, a new erythropoiesis-stimulating agent, has a favorable risk-benefit profile when used to treat anemia associated with chronic renal failure in patients who are on dialysis.
At a meeting of the FDA’s Oncologic Drugs Advisory Committee, it voted 15-1 with one abstention on the risk-benefit question. One of the two cardiologists on the panel voted no; the other abstained. The panel did not vote specifically on whether to recommend approval.
Affymax Inc., the manufacturer of peginesatide – a long-acting erythropoiesis-stimulating agent (ESA) administered intravenously or subcutaneously – has proposed that it be approved for the treatment of anemia associated with chronic kidney disease (CKD) in adults who are on dialysis, but not in CKD patients who are not on dialysis or in cancer patients. Peginesatide is administered once a month, which is less frequently than the two ESAs approved in the United States: epoetin alfa, marketed as Epogen and Procrit, and darbepoetin alfa, marketed as Aranesp. A pegylated epoetin beta (Mircera) is not available in the United States.
In two phase III studies of patients with anemia due to CKD who were on dialysis, 1,066 subjects were treated with peginesatide once a month and 542 were treated with epoetin administered one to three times a week. The effectiveness of peginesatide in maintaining hemoglobin levels at 10-12 g/dL through weeks 29-36 of treatment was similar to that of epoetin, with similar low transfusion rates in both groups. The safety profiles, including serious adverse events, deaths during the study, and adverse events resulting in permanent discontinuation of treatment, were similar in both treatment groups.
But in two phase III studies of patients with anemia due to CKD who were not on dialysis, peginesatide appeared to be less safe than darbepoetin. In these two studies, the rates of adverse events, serious adverse events, deaths during the study, adverse events leading to discontinuation as well as adverse cardiovascular outcomes were higher among those on peginesatide than among those on darbepoetin.
Panelists expressed concerned about this safety signal in the patients not on dialysis, but those who voted in favor of peginesatide for the proposed indication agreed that the safety and efficacy of peginesatide were comparable with epoetin in two large randomized studies and cited the convenience of once-a-month dosing for patients. They recommended postmarketing follow-up of these patients to provide longer-term data, since the average length of ESA treatment in patients with CKD is about 5 years and patients in the studies were followed for a little more than 1 year.
The two cardiologists on the panel cited concerns about the studies not being blinded and the possibility that the differences in toxicity in the two patient groups might not be explained pathophysiologically but instead might be the result of stricter entry criteria in the study of patients on dialysis. They also cited the potential for misuse of these drugs in patients.
Targeting higher hemoglobin levels with ESA treatment has been associated with an increased risk of MI and other adverse cardiovascular outcomes in several large clinical trials, which led to changes in treatment recommendations.
In 2007, a boxed warning about the increased risk of deaths and serious cardiovascular events when higher hemoglobin levels are targeted was added to the labels of ESAs, with the recommendation to individualize dosing to target and maintain hemoglobin levels within the 10-12 g/dL range. In 2009, a warning about the increased risk of stroke based on the results of a study of patients with diabetes and CKD who were not on dialysis was added to ESA labels.
The FDA usually follows the recommendations of its advisory panels, which are not binding. Panelists have been cleared of potential conflicts of interest related to the topic of the meeting. Occasionally, a panelist may be given a waiver, but not at this meeting.
FROM A MEETING OF THE FOOD AND DRUG ADMINISTRATION'S ONCOLOGIC DRUGS ADVISORY COMMITTEE
Panel Endorses Surveillance for Low-Risk Prostate Cancer
An independent panel convened by the U.S. National Institutes of Health has endorsed the use of active surveillance and delay of treatment for men with localized, low-risk prostate cancer.
"Our panel found that many men with localized, low-risk prostate cancer should be closely monitored, permitting their treatment to be delayed until disease progression warrants it. Some of the men affected by prostate cancer will benefit from immediate treatment and others will benefit from observation," panel and conference chairperson Dr. Patricia A. Ganz said in a telebriefing.
However, monitoring or observational strategies, sometimes referred to as "watchful waiting" or "active surveillance," have not been uniformly studied, and available data do not yet point to clear follow-up protocols, said Dr. Ganz, director of the division of cancer prevention and control research at the Jonsson Comprehensive Cancer Center, University of California, Los Angeles.
The panel identified the combination of a prostate-specific antigen (PSA) level less than 10 ng/dL and a Gleason score of 6 or lower as the emerging consensus definition of "low-risk" disease. "Using this definition, we estimate that more than 100,000 men diagnosed with prostate cancer each year in the United States would be candidates for active monitoring rather than immediate treatment," she said.
Additional research will be needed to determine the point at which treatment might ultimately be needed for men who are being closely monitored. The panel recommended that such studies receiving federal funding should not be done at a single site, but rather should be multicenter studies with large patient populations.
The consensus about active surveillance came from increasing evidence that outcomes for men with low-risk disease are not better for those who undergo surgery or radiation therapy. Among the evidence considered was the newly reported findings of the Prostate Cancer Intervention Versus Observation Trial (PIVOT), the only randomized controlled trial conducted on men identified via PSA screening that compared watchful waiting with radical prostatectomy.
With a median follow-up of 10 years, there were no statistically significant differences in prostate cancer mortality or all-cause mortality. "Supporting data from additional cohort studies give us confidence that the risk of death is minimal in a low risk population," the panel wrote.
According to the document, only about 10% of men eligible for observational strategies choose this approach. The reasons for this are probably due both to physician communication that favors treatment and to patient expectations. "When men are given a diagnosis of cancer, it’s very difficult to decline the standard therapy for this disease, which is either surgery or radiation therapy," Dr. Ganz noted.
For this reason, the panel also endorsed consideration of a name change to remove the anxiety-provoking term cancer to describe this low-risk condition, much as was done with cervical intraepithelial neoplasia for early-stage cervical neoplasms and ductal carcinoma in situ for lower-risk breast lesions.
"Some of the men affected by prostate cancer will benefit from immediate treatment and others will benefit from observation."
They also discussed ways in which urologists and primary care physicians ordering PSA tests could be educated to counsel patients about the potential benefits of active surveillance and delaying treatment, Dr. Ganz said.
"Anything we can do to bring this into the consultation room so that the patient feels comfortable raising this issue with his physician ... [Stakeholders] now have an NIH-vetted document that describes this as a reasonable approach to the management of prostate cancer and for those reasons it can be very powerful," she said.
The 14-member state-of-the-science panel included experts in the fields of cancer prevention and control, urology, pathology, epidemiology, genetics, transplantation, bioethics, economics, health services research, shared decision-making, health communication, and community engagement.
Panel members were compensated for travel to the conference, but were not otherwise paid and have no additional conflicts of interest. The statement does not constitute federal policy.
The draft statement is posted online, and the final version will be posted by mid to late January 2012.
An independent panel convened by the U.S. National Institutes of Health has endorsed the use of active surveillance and delay of treatment for men with localized, low-risk prostate cancer.
"Our panel found that many men with localized, low-risk prostate cancer should be closely monitored, permitting their treatment to be delayed until disease progression warrants it. Some of the men affected by prostate cancer will benefit from immediate treatment and others will benefit from observation," panel and conference chairperson Dr. Patricia A. Ganz said in a telebriefing.
However, monitoring or observational strategies, sometimes referred to as "watchful waiting" or "active surveillance," have not been uniformly studied, and available data do not yet point to clear follow-up protocols, said Dr. Ganz, director of the division of cancer prevention and control research at the Jonsson Comprehensive Cancer Center, University of California, Los Angeles.
The panel identified the combination of a prostate-specific antigen (PSA) level less than 10 ng/dL and a Gleason score of 6 or lower as the emerging consensus definition of "low-risk" disease. "Using this definition, we estimate that more than 100,000 men diagnosed with prostate cancer each year in the United States would be candidates for active monitoring rather than immediate treatment," she said.
Additional research will be needed to determine the point at which treatment might ultimately be needed for men who are being closely monitored. The panel recommended that such studies receiving federal funding should not be done at a single site, but rather should be multicenter studies with large patient populations.
The consensus about active surveillance came from increasing evidence that outcomes for men with low-risk disease are not better for those who undergo surgery or radiation therapy. Among the evidence considered was the newly reported findings of the Prostate Cancer Intervention Versus Observation Trial (PIVOT), the only randomized controlled trial conducted on men identified via PSA screening that compared watchful waiting with radical prostatectomy.
With a median follow-up of 10 years, there were no statistically significant differences in prostate cancer mortality or all-cause mortality. "Supporting data from additional cohort studies give us confidence that the risk of death is minimal in a low risk population," the panel wrote.
According to the document, only about 10% of men eligible for observational strategies choose this approach. The reasons for this are probably due both to physician communication that favors treatment and to patient expectations. "When men are given a diagnosis of cancer, it’s very difficult to decline the standard therapy for this disease, which is either surgery or radiation therapy," Dr. Ganz noted.
For this reason, the panel also endorsed consideration of a name change to remove the anxiety-provoking term cancer to describe this low-risk condition, much as was done with cervical intraepithelial neoplasia for early-stage cervical neoplasms and ductal carcinoma in situ for lower-risk breast lesions.
"Some of the men affected by prostate cancer will benefit from immediate treatment and others will benefit from observation."
They also discussed ways in which urologists and primary care physicians ordering PSA tests could be educated to counsel patients about the potential benefits of active surveillance and delaying treatment, Dr. Ganz said.
"Anything we can do to bring this into the consultation room so that the patient feels comfortable raising this issue with his physician ... [Stakeholders] now have an NIH-vetted document that describes this as a reasonable approach to the management of prostate cancer and for those reasons it can be very powerful," she said.
The 14-member state-of-the-science panel included experts in the fields of cancer prevention and control, urology, pathology, epidemiology, genetics, transplantation, bioethics, economics, health services research, shared decision-making, health communication, and community engagement.
Panel members were compensated for travel to the conference, but were not otherwise paid and have no additional conflicts of interest. The statement does not constitute federal policy.
The draft statement is posted online, and the final version will be posted by mid to late January 2012.
An independent panel convened by the U.S. National Institutes of Health has endorsed the use of active surveillance and delay of treatment for men with localized, low-risk prostate cancer.
"Our panel found that many men with localized, low-risk prostate cancer should be closely monitored, permitting their treatment to be delayed until disease progression warrants it. Some of the men affected by prostate cancer will benefit from immediate treatment and others will benefit from observation," panel and conference chairperson Dr. Patricia A. Ganz said in a telebriefing.
However, monitoring or observational strategies, sometimes referred to as "watchful waiting" or "active surveillance," have not been uniformly studied, and available data do not yet point to clear follow-up protocols, said Dr. Ganz, director of the division of cancer prevention and control research at the Jonsson Comprehensive Cancer Center, University of California, Los Angeles.
The panel identified the combination of a prostate-specific antigen (PSA) level less than 10 ng/dL and a Gleason score of 6 or lower as the emerging consensus definition of "low-risk" disease. "Using this definition, we estimate that more than 100,000 men diagnosed with prostate cancer each year in the United States would be candidates for active monitoring rather than immediate treatment," she said.
Additional research will be needed to determine the point at which treatment might ultimately be needed for men who are being closely monitored. The panel recommended that such studies receiving federal funding should not be done at a single site, but rather should be multicenter studies with large patient populations.
The consensus about active surveillance came from increasing evidence that outcomes for men with low-risk disease are not better for those who undergo surgery or radiation therapy. Among the evidence considered was the newly reported findings of the Prostate Cancer Intervention Versus Observation Trial (PIVOT), the only randomized controlled trial conducted on men identified via PSA screening that compared watchful waiting with radical prostatectomy.
With a median follow-up of 10 years, there were no statistically significant differences in prostate cancer mortality or all-cause mortality. "Supporting data from additional cohort studies give us confidence that the risk of death is minimal in a low risk population," the panel wrote.
According to the document, only about 10% of men eligible for observational strategies choose this approach. The reasons for this are probably due both to physician communication that favors treatment and to patient expectations. "When men are given a diagnosis of cancer, it’s very difficult to decline the standard therapy for this disease, which is either surgery or radiation therapy," Dr. Ganz noted.
For this reason, the panel also endorsed consideration of a name change to remove the anxiety-provoking term cancer to describe this low-risk condition, much as was done with cervical intraepithelial neoplasia for early-stage cervical neoplasms and ductal carcinoma in situ for lower-risk breast lesions.
"Some of the men affected by prostate cancer will benefit from immediate treatment and others will benefit from observation."
They also discussed ways in which urologists and primary care physicians ordering PSA tests could be educated to counsel patients about the potential benefits of active surveillance and delaying treatment, Dr. Ganz said.
"Anything we can do to bring this into the consultation room so that the patient feels comfortable raising this issue with his physician ... [Stakeholders] now have an NIH-vetted document that describes this as a reasonable approach to the management of prostate cancer and for those reasons it can be very powerful," she said.
The 14-member state-of-the-science panel included experts in the fields of cancer prevention and control, urology, pathology, epidemiology, genetics, transplantation, bioethics, economics, health services research, shared decision-making, health communication, and community engagement.
Panel members were compensated for travel to the conference, but were not otherwise paid and have no additional conflicts of interest. The statement does not constitute federal policy.
The draft statement is posted online, and the final version will be posted by mid to late January 2012.
Major Finding: Many men with localized, low-risk prostate cancer should be closely monitored, permitting their treatment to be delayed until disease progression warrants it.
Data Source: Newly reported findings of the Prostate Cancer Intervention Versus Observation Trial (PIVOT).
Disclosures: Panel members were compensated for travel to the conference, but were not otherwise paid and have no additional conflicts of interest. The statement does not constitute federal policy.
Chronic Kidney Disease: Protecting Against Progressive Nephropathy
Chronic kidney disease (CKD) is the silent epidemic. It often follows hypertension, diabetes, and obesity. Patients with CKD are not seen exclusively in a nephrology practice. Often, they are not referred to a nephrologist until their glomerular filtration rate (GFR) is less than 60 mL/min/1.73m2.
CKD affects 11.5% of the US population ages 20 and older,1 which translates to some 23 million people. According to the National Institute of Diabetes and Digestive and Kidney Diseases, more than 382,000 people were receiving dialysis in 2008,1 and this number is expected to more than double by 2020. It is essential for the practitioner in any specialty to be aware of their patients’ renal status and any adjustments that may entail.
Q: In diabetic patients with incipient nephropathy (microalbuminuria, but GFR > 90 mL/min, more specifically), do evidence-based recommendations exist that suggest renal dosing parameters be followed in order to protect against progressive nephropathy? This, of course, would constitute a secondary prevention strategy, since (as most clinicians know) glycemic and blood pressure control are the most important primary steps toward prevention.
A patient with microalbuminuria and preserved renal function, according to the estimated GFR (eGFR), would be classified as having CKD stage 2. Currently, no medication dosing guidelines are available for patients at this stage of CKD; however, as providers, we should be aware that the presence of microalbuminuria has important clinical implications.
Microalbuminuria is associated with increased risk for cardiovascular disease and progression of CKD.4 Accordingly, medication selection should be targeted at avoiding renal insult, limiting progression of nephropathy through the use of ACE inhibitors or angiotensin receptor blockers (ARBs), and promptly addressing and modifying the risk factors for cardiovascular disease.
Blood pressure (BP), glycemic control, and lipids should all be managed aggressively. The JNC7 (Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure)5 and the K/DOQI (Kidney Disease Outcomes Quality Initiative) guidelines6 recommend a target BP below 130/80 mm Hg for CKD patients with proteinuria of less than 1 g/24 h; and below 120/80 mm Hg for patients with proteinuria greater than 1 g/24 h. Unpublished guidelines from “Kidney Disease: Improving Global Outcomes” (see www.kdigo.org) are encouraging practitioners to give nonhypertensive diabetic patients with proteinuria an ACE inhibitor or an ARB.
Nephrotoxins, such as NSAIDs and aminoglycosides, should be avoided. Contrast dye and nephrotoxic chemotherapeutic agents should be used with caution and only if clinically justified. If they are used, renal precautions should be taken, including preprocedure and postprocedure hydration.7
Whenever possible, an ACE inhibitor or an ARB should be initiated early, as the greatest benefit in slowing the progression of nephrosclerosis is realized when these medications are initiated before irreversible scarring has occurred.5,6 Initiating one of these agents is preferable while the serum creatinine level is below 1.2 mg/dL.8 Creatinine and potassium should be checked within two weeks of initiating or increasing ACE inhibitor or ARB dosing.
As CKD progresses, patients are at risk for acidemia and hyperkalemia. Typically, these concerns are greater in CKD stages 4 to 5 but can be seen as early as CKD stage 3. Metformin should be prescribed with caution in patients with CKD stage 2 and changed to an alternate antihyperglycemic agent for men whose serum creatinine exceeds 1.5 mg/dL and for women with serum creatinine greater than 1.4 mg/dL.8
Clinically, it is recommended that metformin be avoided in patients whose eGFR is below 60 to 70 mL/min.9 The eGFR is a better indicator of CKD stage than serum creatinine; using serum creatinine alone to calculate the CKD stage could lead to a very wrong result. For example, a thin, elderly white woman can have a very low eGFR but a serum creatinine of 1.4 mg/dL or less (which is essentially normal).
Diabetic patients in particular are susceptible to hyperkalemia, so spironolactone, potassium, and sulfamethoxazole should be prescribed with caution. Potassium levels should be monitored in patients receiving b-blockers, ACE inhibitors, or ARBs.
Renal medication dosing guidelines, as noted in the FDA information in each drug’s package insert, generally offer recommendations for adjustment in CKD stage 4 or 5, which correspond to an eGFR of < 30 mL/min or < 15 mL/min, respectively. FDA renal dosing guidelines are often based on serum creatinine, but eGFR can vary widely for a particular serum creatinine level, depending upon patient variables such as age, weight, race, and gender (as in the example above).
In summary, although no specific published guidelines exist for patients with CKD stage 2, the presence of microalbuminuria is an important clinical indicator that should inform the provider’s approach to patient management.
Alexis Chettiar, ACNP, East Bay Nephrology Medical Group, Oakland, CA
References
1. National Kidney and Urologic Diseases Information Clearinghouse (NKUDIC). http://kidney.niddk.nih.gov. Accessed November 18, 2011.
2. Thorp ML, Morris CD, Bagby SP. A crossover study of gabapentin in treatment of restless legs syndrome among hemodialysis patients. Am J Kidney Dis. 2001;38(1):104-108.
3. Blommel ML, Blommel AL. Pregabalin: an antiepileptic agent useful for neuropathic pain. Am J Health-System Pharm. 2007;64(14):1475-1482.
4. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA. 2001;286(4):421-426.
5. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289(19):2560-2572.
6. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004; 43(5 suppl 1):S1-290.
7. National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF KDOQI™). www.kidney.org/professionals/kdoqi. Accessed November 18, 2011.
8. Post TW, Rose BD. Overview of management of chronic kidney disease in adults. www.uptodate.com/contents/overview-of-the-management-of-chronic-kidney-disease-in-adults/contributors. Accessed November 18, 2011.
9. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med. 1999;131(4):281-303.
Chronic kidney disease (CKD) is the silent epidemic. It often follows hypertension, diabetes, and obesity. Patients with CKD are not seen exclusively in a nephrology practice. Often, they are not referred to a nephrologist until their glomerular filtration rate (GFR) is less than 60 mL/min/1.73m2.
CKD affects 11.5% of the US population ages 20 and older,1 which translates to some 23 million people. According to the National Institute of Diabetes and Digestive and Kidney Diseases, more than 382,000 people were receiving dialysis in 2008,1 and this number is expected to more than double by 2020. It is essential for the practitioner in any specialty to be aware of their patients’ renal status and any adjustments that may entail.
Q: In diabetic patients with incipient nephropathy (microalbuminuria, but GFR > 90 mL/min, more specifically), do evidence-based recommendations exist that suggest renal dosing parameters be followed in order to protect against progressive nephropathy? This, of course, would constitute a secondary prevention strategy, since (as most clinicians know) glycemic and blood pressure control are the most important primary steps toward prevention.
A patient with microalbuminuria and preserved renal function, according to the estimated GFR (eGFR), would be classified as having CKD stage 2. Currently, no medication dosing guidelines are available for patients at this stage of CKD; however, as providers, we should be aware that the presence of microalbuminuria has important clinical implications.
Microalbuminuria is associated with increased risk for cardiovascular disease and progression of CKD.4 Accordingly, medication selection should be targeted at avoiding renal insult, limiting progression of nephropathy through the use of ACE inhibitors or angiotensin receptor blockers (ARBs), and promptly addressing and modifying the risk factors for cardiovascular disease.
Blood pressure (BP), glycemic control, and lipids should all be managed aggressively. The JNC7 (Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure)5 and the K/DOQI (Kidney Disease Outcomes Quality Initiative) guidelines6 recommend a target BP below 130/80 mm Hg for CKD patients with proteinuria of less than 1 g/24 h; and below 120/80 mm Hg for patients with proteinuria greater than 1 g/24 h. Unpublished guidelines from “Kidney Disease: Improving Global Outcomes” (see www.kdigo.org) are encouraging practitioners to give nonhypertensive diabetic patients with proteinuria an ACE inhibitor or an ARB.
Nephrotoxins, such as NSAIDs and aminoglycosides, should be avoided. Contrast dye and nephrotoxic chemotherapeutic agents should be used with caution and only if clinically justified. If they are used, renal precautions should be taken, including preprocedure and postprocedure hydration.7
Whenever possible, an ACE inhibitor or an ARB should be initiated early, as the greatest benefit in slowing the progression of nephrosclerosis is realized when these medications are initiated before irreversible scarring has occurred.5,6 Initiating one of these agents is preferable while the serum creatinine level is below 1.2 mg/dL.8 Creatinine and potassium should be checked within two weeks of initiating or increasing ACE inhibitor or ARB dosing.
As CKD progresses, patients are at risk for acidemia and hyperkalemia. Typically, these concerns are greater in CKD stages 4 to 5 but can be seen as early as CKD stage 3. Metformin should be prescribed with caution in patients with CKD stage 2 and changed to an alternate antihyperglycemic agent for men whose serum creatinine exceeds 1.5 mg/dL and for women with serum creatinine greater than 1.4 mg/dL.8
Clinically, it is recommended that metformin be avoided in patients whose eGFR is below 60 to 70 mL/min.9 The eGFR is a better indicator of CKD stage than serum creatinine; using serum creatinine alone to calculate the CKD stage could lead to a very wrong result. For example, a thin, elderly white woman can have a very low eGFR but a serum creatinine of 1.4 mg/dL or less (which is essentially normal).
Diabetic patients in particular are susceptible to hyperkalemia, so spironolactone, potassium, and sulfamethoxazole should be prescribed with caution. Potassium levels should be monitored in patients receiving b-blockers, ACE inhibitors, or ARBs.
Renal medication dosing guidelines, as noted in the FDA information in each drug’s package insert, generally offer recommendations for adjustment in CKD stage 4 or 5, which correspond to an eGFR of < 30 mL/min or < 15 mL/min, respectively. FDA renal dosing guidelines are often based on serum creatinine, but eGFR can vary widely for a particular serum creatinine level, depending upon patient variables such as age, weight, race, and gender (as in the example above).
In summary, although no specific published guidelines exist for patients with CKD stage 2, the presence of microalbuminuria is an important clinical indicator that should inform the provider’s approach to patient management.
Alexis Chettiar, ACNP, East Bay Nephrology Medical Group, Oakland, CA
References
1. National Kidney and Urologic Diseases Information Clearinghouse (NKUDIC). http://kidney.niddk.nih.gov. Accessed November 18, 2011.
2. Thorp ML, Morris CD, Bagby SP. A crossover study of gabapentin in treatment of restless legs syndrome among hemodialysis patients. Am J Kidney Dis. 2001;38(1):104-108.
3. Blommel ML, Blommel AL. Pregabalin: an antiepileptic agent useful for neuropathic pain. Am J Health-System Pharm. 2007;64(14):1475-1482.
4. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA. 2001;286(4):421-426.
5. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289(19):2560-2572.
6. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004; 43(5 suppl 1):S1-290.
7. National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF KDOQI™). www.kidney.org/professionals/kdoqi. Accessed November 18, 2011.
8. Post TW, Rose BD. Overview of management of chronic kidney disease in adults. www.uptodate.com/contents/overview-of-the-management-of-chronic-kidney-disease-in-adults/contributors. Accessed November 18, 2011.
9. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med. 1999;131(4):281-303.
Chronic kidney disease (CKD) is the silent epidemic. It often follows hypertension, diabetes, and obesity. Patients with CKD are not seen exclusively in a nephrology practice. Often, they are not referred to a nephrologist until their glomerular filtration rate (GFR) is less than 60 mL/min/1.73m2.
CKD affects 11.5% of the US population ages 20 and older,1 which translates to some 23 million people. According to the National Institute of Diabetes and Digestive and Kidney Diseases, more than 382,000 people were receiving dialysis in 2008,1 and this number is expected to more than double by 2020. It is essential for the practitioner in any specialty to be aware of their patients’ renal status and any adjustments that may entail.
Q: In diabetic patients with incipient nephropathy (microalbuminuria, but GFR > 90 mL/min, more specifically), do evidence-based recommendations exist that suggest renal dosing parameters be followed in order to protect against progressive nephropathy? This, of course, would constitute a secondary prevention strategy, since (as most clinicians know) glycemic and blood pressure control are the most important primary steps toward prevention.
A patient with microalbuminuria and preserved renal function, according to the estimated GFR (eGFR), would be classified as having CKD stage 2. Currently, no medication dosing guidelines are available for patients at this stage of CKD; however, as providers, we should be aware that the presence of microalbuminuria has important clinical implications.
Microalbuminuria is associated with increased risk for cardiovascular disease and progression of CKD.4 Accordingly, medication selection should be targeted at avoiding renal insult, limiting progression of nephropathy through the use of ACE inhibitors or angiotensin receptor blockers (ARBs), and promptly addressing and modifying the risk factors for cardiovascular disease.
Blood pressure (BP), glycemic control, and lipids should all be managed aggressively. The JNC7 (Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure)5 and the K/DOQI (Kidney Disease Outcomes Quality Initiative) guidelines6 recommend a target BP below 130/80 mm Hg for CKD patients with proteinuria of less than 1 g/24 h; and below 120/80 mm Hg for patients with proteinuria greater than 1 g/24 h. Unpublished guidelines from “Kidney Disease: Improving Global Outcomes” (see www.kdigo.org) are encouraging practitioners to give nonhypertensive diabetic patients with proteinuria an ACE inhibitor or an ARB.
Nephrotoxins, such as NSAIDs and aminoglycosides, should be avoided. Contrast dye and nephrotoxic chemotherapeutic agents should be used with caution and only if clinically justified. If they are used, renal precautions should be taken, including preprocedure and postprocedure hydration.7
Whenever possible, an ACE inhibitor or an ARB should be initiated early, as the greatest benefit in slowing the progression of nephrosclerosis is realized when these medications are initiated before irreversible scarring has occurred.5,6 Initiating one of these agents is preferable while the serum creatinine level is below 1.2 mg/dL.8 Creatinine and potassium should be checked within two weeks of initiating or increasing ACE inhibitor or ARB dosing.
As CKD progresses, patients are at risk for acidemia and hyperkalemia. Typically, these concerns are greater in CKD stages 4 to 5 but can be seen as early as CKD stage 3. Metformin should be prescribed with caution in patients with CKD stage 2 and changed to an alternate antihyperglycemic agent for men whose serum creatinine exceeds 1.5 mg/dL and for women with serum creatinine greater than 1.4 mg/dL.8
Clinically, it is recommended that metformin be avoided in patients whose eGFR is below 60 to 70 mL/min.9 The eGFR is a better indicator of CKD stage than serum creatinine; using serum creatinine alone to calculate the CKD stage could lead to a very wrong result. For example, a thin, elderly white woman can have a very low eGFR but a serum creatinine of 1.4 mg/dL or less (which is essentially normal).
Diabetic patients in particular are susceptible to hyperkalemia, so spironolactone, potassium, and sulfamethoxazole should be prescribed with caution. Potassium levels should be monitored in patients receiving b-blockers, ACE inhibitors, or ARBs.
Renal medication dosing guidelines, as noted in the FDA information in each drug’s package insert, generally offer recommendations for adjustment in CKD stage 4 or 5, which correspond to an eGFR of < 30 mL/min or < 15 mL/min, respectively. FDA renal dosing guidelines are often based on serum creatinine, but eGFR can vary widely for a particular serum creatinine level, depending upon patient variables such as age, weight, race, and gender (as in the example above).
In summary, although no specific published guidelines exist for patients with CKD stage 2, the presence of microalbuminuria is an important clinical indicator that should inform the provider’s approach to patient management.
Alexis Chettiar, ACNP, East Bay Nephrology Medical Group, Oakland, CA
References
1. National Kidney and Urologic Diseases Information Clearinghouse (NKUDIC). http://kidney.niddk.nih.gov. Accessed November 18, 2011.
2. Thorp ML, Morris CD, Bagby SP. A crossover study of gabapentin in treatment of restless legs syndrome among hemodialysis patients. Am J Kidney Dis. 2001;38(1):104-108.
3. Blommel ML, Blommel AL. Pregabalin: an antiepileptic agent useful for neuropathic pain. Am J Health-System Pharm. 2007;64(14):1475-1482.
4. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA. 2001;286(4):421-426.
5. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289(19):2560-2572.
6. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004; 43(5 suppl 1):S1-290.
7. National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF KDOQI™). www.kidney.org/professionals/kdoqi. Accessed November 18, 2011.
8. Post TW, Rose BD. Overview of management of chronic kidney disease in adults. www.uptodate.com/contents/overview-of-the-management-of-chronic-kidney-disease-in-adults/contributors. Accessed November 18, 2011.
9. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med. 1999;131(4):281-303.
Chronic Kidney Disease: Treating Peripheral Neuropathy Caused by Diabetes
Chronic kidney disease (CKD) is the silent epidemic. It often follows hypertension, diabetes, and obesity. Patients with CKD are not seen exclusively in a nephrology practice. Often, they are not referred to a nephrologist until their glomerular filtration rate (GFR) is less than 60 mL/min/1.73m2.
CKD affects 11.5% of the US population ages 20 and older,1 which translates to some 23 million people. According to the National Institute of Diabetes and Digestive and Kidney Diseases, more than 382,000 people were receiving dialysis in 2008,1 and this number is expected to more than double by 2020. It is essential for the practitioner in any specialty to be aware of their patients’ renal status and any adjustments that may entail.
Q: In my primary care office, I saw a dialysis patient with peripheral neuropathy caused by her diabetes. I treated her with gabapentin 300 mg qd with an increase of 100 mg each week until her symptoms resolved. I received a note from the nephrology group that the dose I ordered was way too high, and they adjusted it down to 100 mg/d. Is that right? Would it have been better for me to prescribe pregabalin? I was trying to use an inexpensive medication because this patient has to take so many.
Gabapentin has been used for years in the dialysis unit to treat patients with diabetes-related peripheral neuropathy.2 It is one of the most commonly prescribed drugs for this population of patients due to its effectiveness and low adverse-effect profile. However, because gabapentin is cleared solely by renal excretion, it is recommended that patients on dialysis receive 200 to 300 mg after each four-hour hemodialysis session. This dose should be reached with gradual titration to avoid adverse effects, which include dizziness, ataxia, sedation, euphoria, ankle edema, and weight gain.
The risk for altered consciousness and myoclonus associated with gabapentin is increased in the dialysis population. When these adverse effects occur, the drug should be stopped. Doses above the recommended 200 to 300 mg per dialysis session have not been shown to provide any added analgesic effect and may increase adverse effects, putting patients at greater risk for falls, in addition to other side effects. Gabapentin has a much longer half-life in patients on dialysis, compared with those who have normal kidney function. These patients will benefit from a minimal dosing schedule as well as the prolonged pain control with gabapentin.
The efficacy of pregabalin (Lyrica®) in the management of painful diabetic neuropathy has been established in several controlled clinical trials.3 Because it has not been in use as long as gabapentin, its safety profile has not yet been established. Pregabalin has better gastrointestinal absorption than gabapentin and offers more rapid pain relief; it can be administered twice daily. Pregabalin is cleared rapidly by dialysis and has a short half-life; therefore, an extra dose is required after each dialysis session. Pregabalin dosing must also be adjusted for creatinine clearance.*
Pregabalin is a Schedule V controlled substance because of its potential for abuse. It cannot be prescribed by advanced practitioners in all states.
Dawn McCombs, CRNP, Nephrology Associates, PC, Birmingham, AL
* Several sources are available on the Internet to determine creatinine clearance or stage of kidney disease if your lab does not calculate it for you.
References
1. National Kidney and Urologic Diseases Information Clearinghouse (NKUDIC). http://kidney.niddk.nih.gov. Accessed November 18, 2011.
2. Thorp ML, Morris CD, Bagby SP. A crossover study of gabapentin in treatment of restless legs syndrome among hemodialysis patients. Am J Kidney Dis. 2001;38(1):104-108.
3. Blommel ML, Blommel AL. Pregabalin: an antiepileptic agent useful for neuropathic pain. Am J Health-System Pharm. 2007;64(14):1475-1482.
4. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA. 2001;286(4):421-426.
5. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289(19):2560-2572.
6. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004; 43(5 suppl 1):S1-290.
7. National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF KDOQI™). www.kidney.org/professionals/kdoqi. Accessed November 18, 2011.
8. Post TW, Rose BD. Overview of management of chronic kidney disease in adults. www.uptodate.com/contents/overview-of-the-management-of-chronic-kidney-disease-in-adults/contributors. Accessed November 18, 2011.
9. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med. 1999;131(4):281-303.
Chronic kidney disease (CKD) is the silent epidemic. It often follows hypertension, diabetes, and obesity. Patients with CKD are not seen exclusively in a nephrology practice. Often, they are not referred to a nephrologist until their glomerular filtration rate (GFR) is less than 60 mL/min/1.73m2.
CKD affects 11.5% of the US population ages 20 and older,1 which translates to some 23 million people. According to the National Institute of Diabetes and Digestive and Kidney Diseases, more than 382,000 people were receiving dialysis in 2008,1 and this number is expected to more than double by 2020. It is essential for the practitioner in any specialty to be aware of their patients’ renal status and any adjustments that may entail.
Q: In my primary care office, I saw a dialysis patient with peripheral neuropathy caused by her diabetes. I treated her with gabapentin 300 mg qd with an increase of 100 mg each week until her symptoms resolved. I received a note from the nephrology group that the dose I ordered was way too high, and they adjusted it down to 100 mg/d. Is that right? Would it have been better for me to prescribe pregabalin? I was trying to use an inexpensive medication because this patient has to take so many.
Gabapentin has been used for years in the dialysis unit to treat patients with diabetes-related peripheral neuropathy.2 It is one of the most commonly prescribed drugs for this population of patients due to its effectiveness and low adverse-effect profile. However, because gabapentin is cleared solely by renal excretion, it is recommended that patients on dialysis receive 200 to 300 mg after each four-hour hemodialysis session. This dose should be reached with gradual titration to avoid adverse effects, which include dizziness, ataxia, sedation, euphoria, ankle edema, and weight gain.
The risk for altered consciousness and myoclonus associated with gabapentin is increased in the dialysis population. When these adverse effects occur, the drug should be stopped. Doses above the recommended 200 to 300 mg per dialysis session have not been shown to provide any added analgesic effect and may increase adverse effects, putting patients at greater risk for falls, in addition to other side effects. Gabapentin has a much longer half-life in patients on dialysis, compared with those who have normal kidney function. These patients will benefit from a minimal dosing schedule as well as the prolonged pain control with gabapentin.
The efficacy of pregabalin (Lyrica®) in the management of painful diabetic neuropathy has been established in several controlled clinical trials.3 Because it has not been in use as long as gabapentin, its safety profile has not yet been established. Pregabalin has better gastrointestinal absorption than gabapentin and offers more rapid pain relief; it can be administered twice daily. Pregabalin is cleared rapidly by dialysis and has a short half-life; therefore, an extra dose is required after each dialysis session. Pregabalin dosing must also be adjusted for creatinine clearance.*
Pregabalin is a Schedule V controlled substance because of its potential for abuse. It cannot be prescribed by advanced practitioners in all states.
Dawn McCombs, CRNP, Nephrology Associates, PC, Birmingham, AL
* Several sources are available on the Internet to determine creatinine clearance or stage of kidney disease if your lab does not calculate it for you.
References
1. National Kidney and Urologic Diseases Information Clearinghouse (NKUDIC). http://kidney.niddk.nih.gov. Accessed November 18, 2011.
2. Thorp ML, Morris CD, Bagby SP. A crossover study of gabapentin in treatment of restless legs syndrome among hemodialysis patients. Am J Kidney Dis. 2001;38(1):104-108.
3. Blommel ML, Blommel AL. Pregabalin: an antiepileptic agent useful for neuropathic pain. Am J Health-System Pharm. 2007;64(14):1475-1482.
4. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA. 2001;286(4):421-426.
5. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289(19):2560-2572.
6. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004; 43(5 suppl 1):S1-290.
7. National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF KDOQI™). www.kidney.org/professionals/kdoqi. Accessed November 18, 2011.
8. Post TW, Rose BD. Overview of management of chronic kidney disease in adults. www.uptodate.com/contents/overview-of-the-management-of-chronic-kidney-disease-in-adults/contributors. Accessed November 18, 2011.
9. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med. 1999;131(4):281-303.
Chronic kidney disease (CKD) is the silent epidemic. It often follows hypertension, diabetes, and obesity. Patients with CKD are not seen exclusively in a nephrology practice. Often, they are not referred to a nephrologist until their glomerular filtration rate (GFR) is less than 60 mL/min/1.73m2.
CKD affects 11.5% of the US population ages 20 and older,1 which translates to some 23 million people. According to the National Institute of Diabetes and Digestive and Kidney Diseases, more than 382,000 people were receiving dialysis in 2008,1 and this number is expected to more than double by 2020. It is essential for the practitioner in any specialty to be aware of their patients’ renal status and any adjustments that may entail.
Q: In my primary care office, I saw a dialysis patient with peripheral neuropathy caused by her diabetes. I treated her with gabapentin 300 mg qd with an increase of 100 mg each week until her symptoms resolved. I received a note from the nephrology group that the dose I ordered was way too high, and they adjusted it down to 100 mg/d. Is that right? Would it have been better for me to prescribe pregabalin? I was trying to use an inexpensive medication because this patient has to take so many.
Gabapentin has been used for years in the dialysis unit to treat patients with diabetes-related peripheral neuropathy.2 It is one of the most commonly prescribed drugs for this population of patients due to its effectiveness and low adverse-effect profile. However, because gabapentin is cleared solely by renal excretion, it is recommended that patients on dialysis receive 200 to 300 mg after each four-hour hemodialysis session. This dose should be reached with gradual titration to avoid adverse effects, which include dizziness, ataxia, sedation, euphoria, ankle edema, and weight gain.
The risk for altered consciousness and myoclonus associated with gabapentin is increased in the dialysis population. When these adverse effects occur, the drug should be stopped. Doses above the recommended 200 to 300 mg per dialysis session have not been shown to provide any added analgesic effect and may increase adverse effects, putting patients at greater risk for falls, in addition to other side effects. Gabapentin has a much longer half-life in patients on dialysis, compared with those who have normal kidney function. These patients will benefit from a minimal dosing schedule as well as the prolonged pain control with gabapentin.
The efficacy of pregabalin (Lyrica®) in the management of painful diabetic neuropathy has been established in several controlled clinical trials.3 Because it has not been in use as long as gabapentin, its safety profile has not yet been established. Pregabalin has better gastrointestinal absorption than gabapentin and offers more rapid pain relief; it can be administered twice daily. Pregabalin is cleared rapidly by dialysis and has a short half-life; therefore, an extra dose is required after each dialysis session. Pregabalin dosing must also be adjusted for creatinine clearance.*
Pregabalin is a Schedule V controlled substance because of its potential for abuse. It cannot be prescribed by advanced practitioners in all states.
Dawn McCombs, CRNP, Nephrology Associates, PC, Birmingham, AL
* Several sources are available on the Internet to determine creatinine clearance or stage of kidney disease if your lab does not calculate it for you.
References
1. National Kidney and Urologic Diseases Information Clearinghouse (NKUDIC). http://kidney.niddk.nih.gov. Accessed November 18, 2011.
2. Thorp ML, Morris CD, Bagby SP. A crossover study of gabapentin in treatment of restless legs syndrome among hemodialysis patients. Am J Kidney Dis. 2001;38(1):104-108.
3. Blommel ML, Blommel AL. Pregabalin: an antiepileptic agent useful for neuropathic pain. Am J Health-System Pharm. 2007;64(14):1475-1482.
4. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA. 2001;286(4):421-426.
5. Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289(19):2560-2572.
6. Kidney Disease Outcomes Quality Initiative (K/DOQI). K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis. 2004; 43(5 suppl 1):S1-290.
7. National Kidney Foundation Kidney Disease Outcomes Quality Initiative (NKF KDOQI™). www.kidney.org/professionals/kdoqi. Accessed November 18, 2011.
8. Post TW, Rose BD. Overview of management of chronic kidney disease in adults. www.uptodate.com/contents/overview-of-the-management-of-chronic-kidney-disease-in-adults/contributors. Accessed November 18, 2011.
9. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mellitus. Ann Intern Med. 1999;131(4):281-303.
Inadequate differential proves fatal ... Death by fentanyl patch and methadone ... more
Culture results go undiscussed, man suffers stroke
TWO WEEKS AFTER PROSTATE SURGERY, a 76-year-old man went to the ED because he was having trouble urinating. The ED physician catheterized the patient, ordered a urine culture, and discharged him.
The culture results, showing methicillin-resistant Staphylococcus aureus, were sent to a printer in the ED twice, as was the usual practice, but evidently no one saw them.
The patient returned to the ED 2 weeks after his initial visit with the same complaint of difficult urination and was seen by the same physician. The physician again discharged him with a catheter but without mentioning the culture results. Two days later, the patient suffered a stroke, which paralyzed his left side.
PLAINTIFF’S CLAIM The bacteria had spread from the patient’s urine to his bloodstream, sparking a cascade of events that led to the stroke.
THE DEFENSE No information about the defense is available.
VERDICT $2.25 million New Jersey settlement.
COMMENT The repeated missed opportunities to diagnose and treat this patient’s infection were regrettable—and costly.
Inadequate differential proves fatal
SHORTNESS OF BREATH led a 52-year-old woman to visit her medical group, where she was a long-time patient. The family practitioner who saw her noted tachycardia and ordered an electrocardiogram, which was abnormal. The physician also ordered a chest x-ray and, because the woman had a history of anemia, a complete blood count and a number of other blood tests. He subsequently called the patient at home to tell her that the blood tests were normal and she didn’t have anemia.
Three days later, the patient went to an urgent care center complaining of shortness of breath and tightness in her chest. A pulmonary embolism was diagnosed, and she was transferred to a hospital ED. Later that evening, a code blue was called and the patient was resuscitated. She died the following day.
PLAINTIFF’S CLAIM The doctor assumed that the patient had anemia and failed to develop a differential diagnosis. The patient had risk factors for pulmonary embolism—obesity and the use of an ethinyl estradiol-etonogestrel vaginal contraceptive ring—which should have prompted the doctor to consider that possibility. If he had done so, the pulmonary embolism would have been diagnosed and the patient’s death prevented.
THE DEFENSE The patient’s presentation wasn’t typical for pulmonary embolism, and there wasn’t any way to know whether an earlier diagnosis would have resulted in survival.
VERDICT $1.9 million California verdict.
COMMENT Although pulmonary embolism can be a challenging diagnosis to make, it needs to be considered carefully in all patients with shortness of breath, chest pain, or poorly defined pulmonary or cardiac symptoms.
The correct diagnosis comes too late
FLU-LIKE SYMPTOMS AND AN IRREGULAR HEART RATE prompted a man to go to the ED, where the physician diagnosed a viral infection, prescribed pain medication, and discharged him. The following day, a laboratory report indicating a staph infection was sent to an ED secretary, but the patient wasn’t told the results.
The patient returned to the hospital 2 days later in a confused state. Tests revealed a staph infection and meningitis, for which the patient received antibiotics. A week later, the patient suffered a stroke, resulting in diminished cognitive ability, impaired vision, and right-sided motor deficits.
PLAINTIFF’S CLAIM The white blood cell count and C-reactive protein level measured at the patient’s first visit to the ED would have led to a diagnosis of bacterial infection. The patient should have been admitted to the hospital and given antibiotics at that time.
THE DEFENSE The original diagnosis was reasonable.
VERDICT Confidential settlement with the hospital. $900,000 net verdict against the physician in New Jersey.
COMMENT Lab reports gone awry and the lack of a fail-safe for abnormal tests result in a $900,000 judgment. Do you have adequate systems in place to avoid a communication failure like this one?
Slow response turns a bad situation into a disaster
A 66-YEAR-OLD MAN on warfarin therapy for chronic atrial fibrillation and a transient ischemic attack underwent lithotripsy for kidney stones. Three days after the lithotripsy, he went to the ED complaining of severe flank pain. A computed tomography (CT) scan of the abdomen showed a large retroperitoneal hematoma and prominent perinephric and pararenal hemorrhages.
The patient remained on a gurney in the hallway of the ED in deteriorating condition until he was admitted to the intensive care unit, by which time his condition was critical. He died the next day.
PLAINTIFF’S CLAIM The ED physician and admitting urologists failed to monitor and treat the patient’s active hemorrhage for 9 hours. They didn’t order coagulation studies or respond to signs of escalating hemorrhagic shock. They failed to seek timely consults from surgery and interventional radiology.
THE DEFENSE No information about the defense is available.
VERDICT $825,000 Virginia settlement.
COMMENT Preventing complications of anticoagulation is hard enough; the lack of a timely response in this case made a bad outcome disastrous.
Were steps taken quickly enough?
SEVERE LOWER ABDOMINAL PAIN prompted a 52-year-old woman to go to the ED. She said she hadn’t had a bowel movement in almost a week. The ED physician, in consultation with the attending physician, admitted her to the hospital and ordered intravenous fluids and a soap suds enema, which didn’t relieve the constipation. The patient’s vital signs deteriorated, and she was crying and restless.
When the attending physician saw the patient almost 3 hours after admission, she had a fever of 101.4°F. He ordered additional tests, a computed tomography (CT) scan, and antibiotics, but didn’t order them STAT.
About 1½ hours later, a house physician examined the patient, and, after speaking with the attending physician, transferred her to a step-down telemetry unit. About 1½ hours after the transfer, a nurse called the house physician to report that the patient’s condition was worsening. The house physician ordered pain relievers and a second enema but didn’t come to the hospital.
Because the patient wasn’t in the intensive care unit, no one checked on her again for 3½ hours. When the nurse did check, she found the patient pale, cold, and turning blue. The nurse called the house physician, who came to the hospital. The patient had a fever of 102.4°F and her blood pressure couldn’t be measured.
After speaking with the attending physician, the house physician had the patient admitted to the ICU and also ordered a STAT surgical consultation and CT scan. In the meantime, the patient went into cardiac arrest and couldn’t be revived. Death was caused by peritonitis with sepsis resulting from a large intestinal obstruction.
PLAINTIFF’S CLAIM The patient showed early signs of sepsis. She should have undergone testing sooner and been transferred to the ICU earlier.
THE DEFENSE The doctors claimed that all their actions were appropriate and that the actions suggested by the plaintiff wouldn’t have resulted in the patient’s survival.
VERDICT $3.8 million Pennsylvania verdict.
COMMENT Prompt evaluation and monitoring of this patient might have prevented death and a substantial verdict.
2 analgesic calamities: Death by fentanyl patch …
AFTER A WEEK OF INCREASING BACK PAIN, which had begun to shoot down his right leg, a 37-year-old man went to the ED. He was examined and given prescriptions for pain killers, including acetaminophen and hydrocodone, and muscle relaxants and discharged with instructions to return in 3 days for magnetic resonance imaging (MRI).
While he was at the hospital for the MRI, the patient returned to the ED because he was still in pain and his acetaminophen-hydrocodone prescription was running out. The ED physician prescribed a 0.75-mg fentanyl transdermal patch and instructed the patient to put it on his chest.
Three days later, the patient filled the prescription and applied the patch. The following day, his girlfriend found him dead in bed. Postmortem toxicology results showed a blood fentanyl level of 9.85 ng/mL, markedly higher than the therapeutic level. Respiratory failure caused by fentanyl toxicity was cited as the cause of death.
PLAINTIFF’S CLAIM The ED physician prescribed an excessive dose of fentanyl.
THE DEFENSE A defective patch or misuse of the patch caused the patient’s death.
VERDICT $1.2 million Indiana verdict.
… and methadone
A 36-YEAR-OLD MAN started treatment with a pain specialist for pain arising from a back problem, for which he had taken pain medication previously. The pain specialist prescribed methadone, 360 10-mg tablets. The prescription limited the patient to 2 tablets every 4 hours for a maximum dosage of 12 tablets (120 mg) per day.
Three days after the patient filled the prescription, he was found dead. An autopsy determined the cause of death to be drug toxicity from methadone. At the time the patient died, the bottle of methadone tablets contained 342 tablets, indicating that he had taken only 18 tablets, well within the maximum dosage authorized by the prescription.
PLAINTIFF’S CLAIM The prescribed methadone dosage was excessive for a patient just beginning to use the drug. A proper initial dosage is between 2.5 and 10 mg every 8 to 12 hours for a maximum of 30 mg per day.
THE DEFENSE No information about the defense is available.
VERDICT Confidential Utah settlement.
COMMENT These 2 cases have a common thread. The effects of opioids are often idiosyncratic. A plan for careful monitoring and follow-up should be prepared at initiation of treatment and when escalating the dosage.
Culture results go undiscussed, man suffers stroke
TWO WEEKS AFTER PROSTATE SURGERY, a 76-year-old man went to the ED because he was having trouble urinating. The ED physician catheterized the patient, ordered a urine culture, and discharged him.
The culture results, showing methicillin-resistant Staphylococcus aureus, were sent to a printer in the ED twice, as was the usual practice, but evidently no one saw them.
The patient returned to the ED 2 weeks after his initial visit with the same complaint of difficult urination and was seen by the same physician. The physician again discharged him with a catheter but without mentioning the culture results. Two days later, the patient suffered a stroke, which paralyzed his left side.
PLAINTIFF’S CLAIM The bacteria had spread from the patient’s urine to his bloodstream, sparking a cascade of events that led to the stroke.
THE DEFENSE No information about the defense is available.
VERDICT $2.25 million New Jersey settlement.
COMMENT The repeated missed opportunities to diagnose and treat this patient’s infection were regrettable—and costly.
Inadequate differential proves fatal
SHORTNESS OF BREATH led a 52-year-old woman to visit her medical group, where she was a long-time patient. The family practitioner who saw her noted tachycardia and ordered an electrocardiogram, which was abnormal. The physician also ordered a chest x-ray and, because the woman had a history of anemia, a complete blood count and a number of other blood tests. He subsequently called the patient at home to tell her that the blood tests were normal and she didn’t have anemia.
Three days later, the patient went to an urgent care center complaining of shortness of breath and tightness in her chest. A pulmonary embolism was diagnosed, and she was transferred to a hospital ED. Later that evening, a code blue was called and the patient was resuscitated. She died the following day.
PLAINTIFF’S CLAIM The doctor assumed that the patient had anemia and failed to develop a differential diagnosis. The patient had risk factors for pulmonary embolism—obesity and the use of an ethinyl estradiol-etonogestrel vaginal contraceptive ring—which should have prompted the doctor to consider that possibility. If he had done so, the pulmonary embolism would have been diagnosed and the patient’s death prevented.
THE DEFENSE The patient’s presentation wasn’t typical for pulmonary embolism, and there wasn’t any way to know whether an earlier diagnosis would have resulted in survival.
VERDICT $1.9 million California verdict.
COMMENT Although pulmonary embolism can be a challenging diagnosis to make, it needs to be considered carefully in all patients with shortness of breath, chest pain, or poorly defined pulmonary or cardiac symptoms.
The correct diagnosis comes too late
FLU-LIKE SYMPTOMS AND AN IRREGULAR HEART RATE prompted a man to go to the ED, where the physician diagnosed a viral infection, prescribed pain medication, and discharged him. The following day, a laboratory report indicating a staph infection was sent to an ED secretary, but the patient wasn’t told the results.
The patient returned to the hospital 2 days later in a confused state. Tests revealed a staph infection and meningitis, for which the patient received antibiotics. A week later, the patient suffered a stroke, resulting in diminished cognitive ability, impaired vision, and right-sided motor deficits.
PLAINTIFF’S CLAIM The white blood cell count and C-reactive protein level measured at the patient’s first visit to the ED would have led to a diagnosis of bacterial infection. The patient should have been admitted to the hospital and given antibiotics at that time.
THE DEFENSE The original diagnosis was reasonable.
VERDICT Confidential settlement with the hospital. $900,000 net verdict against the physician in New Jersey.
COMMENT Lab reports gone awry and the lack of a fail-safe for abnormal tests result in a $900,000 judgment. Do you have adequate systems in place to avoid a communication failure like this one?
Slow response turns a bad situation into a disaster
A 66-YEAR-OLD MAN on warfarin therapy for chronic atrial fibrillation and a transient ischemic attack underwent lithotripsy for kidney stones. Three days after the lithotripsy, he went to the ED complaining of severe flank pain. A computed tomography (CT) scan of the abdomen showed a large retroperitoneal hematoma and prominent perinephric and pararenal hemorrhages.
The patient remained on a gurney in the hallway of the ED in deteriorating condition until he was admitted to the intensive care unit, by which time his condition was critical. He died the next day.
PLAINTIFF’S CLAIM The ED physician and admitting urologists failed to monitor and treat the patient’s active hemorrhage for 9 hours. They didn’t order coagulation studies or respond to signs of escalating hemorrhagic shock. They failed to seek timely consults from surgery and interventional radiology.
THE DEFENSE No information about the defense is available.
VERDICT $825,000 Virginia settlement.
COMMENT Preventing complications of anticoagulation is hard enough; the lack of a timely response in this case made a bad outcome disastrous.
Were steps taken quickly enough?
SEVERE LOWER ABDOMINAL PAIN prompted a 52-year-old woman to go to the ED. She said she hadn’t had a bowel movement in almost a week. The ED physician, in consultation with the attending physician, admitted her to the hospital and ordered intravenous fluids and a soap suds enema, which didn’t relieve the constipation. The patient’s vital signs deteriorated, and she was crying and restless.
When the attending physician saw the patient almost 3 hours after admission, she had a fever of 101.4°F. He ordered additional tests, a computed tomography (CT) scan, and antibiotics, but didn’t order them STAT.
About 1½ hours later, a house physician examined the patient, and, after speaking with the attending physician, transferred her to a step-down telemetry unit. About 1½ hours after the transfer, a nurse called the house physician to report that the patient’s condition was worsening. The house physician ordered pain relievers and a second enema but didn’t come to the hospital.
Because the patient wasn’t in the intensive care unit, no one checked on her again for 3½ hours. When the nurse did check, she found the patient pale, cold, and turning blue. The nurse called the house physician, who came to the hospital. The patient had a fever of 102.4°F and her blood pressure couldn’t be measured.
After speaking with the attending physician, the house physician had the patient admitted to the ICU and also ordered a STAT surgical consultation and CT scan. In the meantime, the patient went into cardiac arrest and couldn’t be revived. Death was caused by peritonitis with sepsis resulting from a large intestinal obstruction.
PLAINTIFF’S CLAIM The patient showed early signs of sepsis. She should have undergone testing sooner and been transferred to the ICU earlier.
THE DEFENSE The doctors claimed that all their actions were appropriate and that the actions suggested by the plaintiff wouldn’t have resulted in the patient’s survival.
VERDICT $3.8 million Pennsylvania verdict.
COMMENT Prompt evaluation and monitoring of this patient might have prevented death and a substantial verdict.
2 analgesic calamities: Death by fentanyl patch …
AFTER A WEEK OF INCREASING BACK PAIN, which had begun to shoot down his right leg, a 37-year-old man went to the ED. He was examined and given prescriptions for pain killers, including acetaminophen and hydrocodone, and muscle relaxants and discharged with instructions to return in 3 days for magnetic resonance imaging (MRI).
While he was at the hospital for the MRI, the patient returned to the ED because he was still in pain and his acetaminophen-hydrocodone prescription was running out. The ED physician prescribed a 0.75-mg fentanyl transdermal patch and instructed the patient to put it on his chest.
Three days later, the patient filled the prescription and applied the patch. The following day, his girlfriend found him dead in bed. Postmortem toxicology results showed a blood fentanyl level of 9.85 ng/mL, markedly higher than the therapeutic level. Respiratory failure caused by fentanyl toxicity was cited as the cause of death.
PLAINTIFF’S CLAIM The ED physician prescribed an excessive dose of fentanyl.
THE DEFENSE A defective patch or misuse of the patch caused the patient’s death.
VERDICT $1.2 million Indiana verdict.
… and methadone
A 36-YEAR-OLD MAN started treatment with a pain specialist for pain arising from a back problem, for which he had taken pain medication previously. The pain specialist prescribed methadone, 360 10-mg tablets. The prescription limited the patient to 2 tablets every 4 hours for a maximum dosage of 12 tablets (120 mg) per day.
Three days after the patient filled the prescription, he was found dead. An autopsy determined the cause of death to be drug toxicity from methadone. At the time the patient died, the bottle of methadone tablets contained 342 tablets, indicating that he had taken only 18 tablets, well within the maximum dosage authorized by the prescription.
PLAINTIFF’S CLAIM The prescribed methadone dosage was excessive for a patient just beginning to use the drug. A proper initial dosage is between 2.5 and 10 mg every 8 to 12 hours for a maximum of 30 mg per day.
THE DEFENSE No information about the defense is available.
VERDICT Confidential Utah settlement.
COMMENT These 2 cases have a common thread. The effects of opioids are often idiosyncratic. A plan for careful monitoring and follow-up should be prepared at initiation of treatment and when escalating the dosage.
Culture results go undiscussed, man suffers stroke
TWO WEEKS AFTER PROSTATE SURGERY, a 76-year-old man went to the ED because he was having trouble urinating. The ED physician catheterized the patient, ordered a urine culture, and discharged him.
The culture results, showing methicillin-resistant Staphylococcus aureus, were sent to a printer in the ED twice, as was the usual practice, but evidently no one saw them.
The patient returned to the ED 2 weeks after his initial visit with the same complaint of difficult urination and was seen by the same physician. The physician again discharged him with a catheter but without mentioning the culture results. Two days later, the patient suffered a stroke, which paralyzed his left side.
PLAINTIFF’S CLAIM The bacteria had spread from the patient’s urine to his bloodstream, sparking a cascade of events that led to the stroke.
THE DEFENSE No information about the defense is available.
VERDICT $2.25 million New Jersey settlement.
COMMENT The repeated missed opportunities to diagnose and treat this patient’s infection were regrettable—and costly.
Inadequate differential proves fatal
SHORTNESS OF BREATH led a 52-year-old woman to visit her medical group, where she was a long-time patient. The family practitioner who saw her noted tachycardia and ordered an electrocardiogram, which was abnormal. The physician also ordered a chest x-ray and, because the woman had a history of anemia, a complete blood count and a number of other blood tests. He subsequently called the patient at home to tell her that the blood tests were normal and she didn’t have anemia.
Three days later, the patient went to an urgent care center complaining of shortness of breath and tightness in her chest. A pulmonary embolism was diagnosed, and she was transferred to a hospital ED. Later that evening, a code blue was called and the patient was resuscitated. She died the following day.
PLAINTIFF’S CLAIM The doctor assumed that the patient had anemia and failed to develop a differential diagnosis. The patient had risk factors for pulmonary embolism—obesity and the use of an ethinyl estradiol-etonogestrel vaginal contraceptive ring—which should have prompted the doctor to consider that possibility. If he had done so, the pulmonary embolism would have been diagnosed and the patient’s death prevented.
THE DEFENSE The patient’s presentation wasn’t typical for pulmonary embolism, and there wasn’t any way to know whether an earlier diagnosis would have resulted in survival.
VERDICT $1.9 million California verdict.
COMMENT Although pulmonary embolism can be a challenging diagnosis to make, it needs to be considered carefully in all patients with shortness of breath, chest pain, or poorly defined pulmonary or cardiac symptoms.
The correct diagnosis comes too late
FLU-LIKE SYMPTOMS AND AN IRREGULAR HEART RATE prompted a man to go to the ED, where the physician diagnosed a viral infection, prescribed pain medication, and discharged him. The following day, a laboratory report indicating a staph infection was sent to an ED secretary, but the patient wasn’t told the results.
The patient returned to the hospital 2 days later in a confused state. Tests revealed a staph infection and meningitis, for which the patient received antibiotics. A week later, the patient suffered a stroke, resulting in diminished cognitive ability, impaired vision, and right-sided motor deficits.
PLAINTIFF’S CLAIM The white blood cell count and C-reactive protein level measured at the patient’s first visit to the ED would have led to a diagnosis of bacterial infection. The patient should have been admitted to the hospital and given antibiotics at that time.
THE DEFENSE The original diagnosis was reasonable.
VERDICT Confidential settlement with the hospital. $900,000 net verdict against the physician in New Jersey.
COMMENT Lab reports gone awry and the lack of a fail-safe for abnormal tests result in a $900,000 judgment. Do you have adequate systems in place to avoid a communication failure like this one?
Slow response turns a bad situation into a disaster
A 66-YEAR-OLD MAN on warfarin therapy for chronic atrial fibrillation and a transient ischemic attack underwent lithotripsy for kidney stones. Three days after the lithotripsy, he went to the ED complaining of severe flank pain. A computed tomography (CT) scan of the abdomen showed a large retroperitoneal hematoma and prominent perinephric and pararenal hemorrhages.
The patient remained on a gurney in the hallway of the ED in deteriorating condition until he was admitted to the intensive care unit, by which time his condition was critical. He died the next day.
PLAINTIFF’S CLAIM The ED physician and admitting urologists failed to monitor and treat the patient’s active hemorrhage for 9 hours. They didn’t order coagulation studies or respond to signs of escalating hemorrhagic shock. They failed to seek timely consults from surgery and interventional radiology.
THE DEFENSE No information about the defense is available.
VERDICT $825,000 Virginia settlement.
COMMENT Preventing complications of anticoagulation is hard enough; the lack of a timely response in this case made a bad outcome disastrous.
Were steps taken quickly enough?
SEVERE LOWER ABDOMINAL PAIN prompted a 52-year-old woman to go to the ED. She said she hadn’t had a bowel movement in almost a week. The ED physician, in consultation with the attending physician, admitted her to the hospital and ordered intravenous fluids and a soap suds enema, which didn’t relieve the constipation. The patient’s vital signs deteriorated, and she was crying and restless.
When the attending physician saw the patient almost 3 hours after admission, she had a fever of 101.4°F. He ordered additional tests, a computed tomography (CT) scan, and antibiotics, but didn’t order them STAT.
About 1½ hours later, a house physician examined the patient, and, after speaking with the attending physician, transferred her to a step-down telemetry unit. About 1½ hours after the transfer, a nurse called the house physician to report that the patient’s condition was worsening. The house physician ordered pain relievers and a second enema but didn’t come to the hospital.
Because the patient wasn’t in the intensive care unit, no one checked on her again for 3½ hours. When the nurse did check, she found the patient pale, cold, and turning blue. The nurse called the house physician, who came to the hospital. The patient had a fever of 102.4°F and her blood pressure couldn’t be measured.
After speaking with the attending physician, the house physician had the patient admitted to the ICU and also ordered a STAT surgical consultation and CT scan. In the meantime, the patient went into cardiac arrest and couldn’t be revived. Death was caused by peritonitis with sepsis resulting from a large intestinal obstruction.
PLAINTIFF’S CLAIM The patient showed early signs of sepsis. She should have undergone testing sooner and been transferred to the ICU earlier.
THE DEFENSE The doctors claimed that all their actions were appropriate and that the actions suggested by the plaintiff wouldn’t have resulted in the patient’s survival.
VERDICT $3.8 million Pennsylvania verdict.
COMMENT Prompt evaluation and monitoring of this patient might have prevented death and a substantial verdict.
2 analgesic calamities: Death by fentanyl patch …
AFTER A WEEK OF INCREASING BACK PAIN, which had begun to shoot down his right leg, a 37-year-old man went to the ED. He was examined and given prescriptions for pain killers, including acetaminophen and hydrocodone, and muscle relaxants and discharged with instructions to return in 3 days for magnetic resonance imaging (MRI).
While he was at the hospital for the MRI, the patient returned to the ED because he was still in pain and his acetaminophen-hydrocodone prescription was running out. The ED physician prescribed a 0.75-mg fentanyl transdermal patch and instructed the patient to put it on his chest.
Three days later, the patient filled the prescription and applied the patch. The following day, his girlfriend found him dead in bed. Postmortem toxicology results showed a blood fentanyl level of 9.85 ng/mL, markedly higher than the therapeutic level. Respiratory failure caused by fentanyl toxicity was cited as the cause of death.
PLAINTIFF’S CLAIM The ED physician prescribed an excessive dose of fentanyl.
THE DEFENSE A defective patch or misuse of the patch caused the patient’s death.
VERDICT $1.2 million Indiana verdict.
… and methadone
A 36-YEAR-OLD MAN started treatment with a pain specialist for pain arising from a back problem, for which he had taken pain medication previously. The pain specialist prescribed methadone, 360 10-mg tablets. The prescription limited the patient to 2 tablets every 4 hours for a maximum dosage of 12 tablets (120 mg) per day.
Three days after the patient filled the prescription, he was found dead. An autopsy determined the cause of death to be drug toxicity from methadone. At the time the patient died, the bottle of methadone tablets contained 342 tablets, indicating that he had taken only 18 tablets, well within the maximum dosage authorized by the prescription.
PLAINTIFF’S CLAIM The prescribed methadone dosage was excessive for a patient just beginning to use the drug. A proper initial dosage is between 2.5 and 10 mg every 8 to 12 hours for a maximum of 30 mg per day.
THE DEFENSE No information about the defense is available.
VERDICT Confidential Utah settlement.
COMMENT These 2 cases have a common thread. The effects of opioids are often idiosyncratic. A plan for careful monitoring and follow-up should be prepared at initiation of treatment and when escalating the dosage.
Urinary Sodium Excretion Levels Flag CV Risk
High urinary sodium excretion, a proxy for high sodium intake, was associated with an increased risk of cardiovascular mortality, myocardial infarction, stroke, and hospitalization for heart failure in a study in the Nov. 23/30 JAMA.
However, low urinary sodium excretion also was associated with increased risk of cardiovascular mortality and hospitalization for heart failure.
"Our findings emphasize the burden of CV disease associated with excess sodium intake and the importance of population-based programs to reduce sodium intake in populations consuming high-sodium diets," Dr. Martin J. O’Donnell of the Population Health Research Institute, McMaster University, Hamilton, Ont., and his associates.
Importantly, the association between sodium intake and adverse CV outcomes did not become significant until intake exceeded 6.5 g/day. This threshold is higher than that recommended by the World Health Organization and by many national guidelines. This highlights "the urgent need to establish a safe range for sodium intake," they noted.
"We found a J-shaped association between estimated sodium excretion and CV events," the researchers noted.
Compared with the "moderate" sodium excretion of 4-5.99 g/day, both higher (more than 7 g/day) and lower (less than 3 g/day) sodium excretion were linked to adverse CV events.
They examined the relationship between sodium intake and CV events using data from two large clinical trials in which patients at high risk for CV events provided urinary samples and were followed for a median of 56 months. Data from the two trials were combined because both recruited subjects from the same areas during the same time period (2001-2004) using the same eligibility criteria and the same methods of obtaining baseline clinical data and monitoring outcomes.
A total of 28,880 study subjects followed at 733 medical centers in 40 countries were included. The subjects were aged 55 and older and either had established CV disease or high-risk diabetes.
Sodium and potassium concentrations were determined from a single morning fasting urine sample from each subject, and this was used to estimate 24-hour sodium and potassium excretion. Overall, the mean estimated 24-hour sodium excretion, a proxy for sodium intake, was 4.77 g, and potassium excretion, a proxy for potassium intake, was 2.19 g.
The composite outcome of CV death, MI, stroke, and hospitalization for heart failure occurred in 4,729 study subjects (16%) during follow-up through 2008. Overall mortality was 12%, the rate of MI was 5%, that of stroke and heart failure hospitalization were each about 4%.
Compared with the moderate excretion of 4-5.99 g/day, higher excretion (more than 7 g/day) was associated with a hazard ratio of 1.15-1.49 for the composite end point. And those with lower excretion (less than 3 g/day), had a hazard ratio of 1.16-1.21 for the composite end point.
In addition, high sodium excretion was significantly associated with each of the individual components of this composite outcome, Dr. O’Donnell and his colleagues said (JAMA 2011;306:2229-38).
"We found a J-shaped association between estimated sodium excretion and CV events."
Lower sodium excretion also was associated with increased risk of heart failure hospitalization.
These results remained robust in several sensitivity and subgroup analyses.
No association was found between potassium excretion and any of the CV outcomes, except that higher potassium excretion was related to a reduced risk of stroke.
"Clearly, large randomized controlled trials evaluating the effect of reduced sodium intake ... on CV outcomes in those with moderate sodium intake are needed urgently. Pending the results of such trials, a more cautious approach to policy on sodium intake may be appropriate, one that targets sodium reduction in populations consuming high sodium levels and reflects the uncertainty in those with moderate sodium diets, which includes the majority of the population."
This study was limited in that it involved patients who volunteered to participate in clinical trials, who may have different lifestyles and behaviors than those who declined to participate. Also, urinary samples were taken at a single time point, which may not accurately reflect subjects’ typical sodium and potassium excretion, Dr. O’Donnell and his associates said.
This study was supported by Boehringer Ingelheim, a manufacturer of angiotensin II receptor blockers. Dr. O’Donnell and several associates reported ties to Boehringer Ingelheim, and his associates reported ties to numerous other industry sources.
High urinary sodium excretion, a proxy for high sodium intake, was associated with an increased risk of cardiovascular mortality, myocardial infarction, stroke, and hospitalization for heart failure in a study in the Nov. 23/30 JAMA.
However, low urinary sodium excretion also was associated with increased risk of cardiovascular mortality and hospitalization for heart failure.
"Our findings emphasize the burden of CV disease associated with excess sodium intake and the importance of population-based programs to reduce sodium intake in populations consuming high-sodium diets," Dr. Martin J. O’Donnell of the Population Health Research Institute, McMaster University, Hamilton, Ont., and his associates.
Importantly, the association between sodium intake and adverse CV outcomes did not become significant until intake exceeded 6.5 g/day. This threshold is higher than that recommended by the World Health Organization and by many national guidelines. This highlights "the urgent need to establish a safe range for sodium intake," they noted.
"We found a J-shaped association between estimated sodium excretion and CV events," the researchers noted.
Compared with the "moderate" sodium excretion of 4-5.99 g/day, both higher (more than 7 g/day) and lower (less than 3 g/day) sodium excretion were linked to adverse CV events.
They examined the relationship between sodium intake and CV events using data from two large clinical trials in which patients at high risk for CV events provided urinary samples and were followed for a median of 56 months. Data from the two trials were combined because both recruited subjects from the same areas during the same time period (2001-2004) using the same eligibility criteria and the same methods of obtaining baseline clinical data and monitoring outcomes.
A total of 28,880 study subjects followed at 733 medical centers in 40 countries were included. The subjects were aged 55 and older and either had established CV disease or high-risk diabetes.
Sodium and potassium concentrations were determined from a single morning fasting urine sample from each subject, and this was used to estimate 24-hour sodium and potassium excretion. Overall, the mean estimated 24-hour sodium excretion, a proxy for sodium intake, was 4.77 g, and potassium excretion, a proxy for potassium intake, was 2.19 g.
The composite outcome of CV death, MI, stroke, and hospitalization for heart failure occurred in 4,729 study subjects (16%) during follow-up through 2008. Overall mortality was 12%, the rate of MI was 5%, that of stroke and heart failure hospitalization were each about 4%.
Compared with the moderate excretion of 4-5.99 g/day, higher excretion (more than 7 g/day) was associated with a hazard ratio of 1.15-1.49 for the composite end point. And those with lower excretion (less than 3 g/day), had a hazard ratio of 1.16-1.21 for the composite end point.
In addition, high sodium excretion was significantly associated with each of the individual components of this composite outcome, Dr. O’Donnell and his colleagues said (JAMA 2011;306:2229-38).
"We found a J-shaped association between estimated sodium excretion and CV events."
Lower sodium excretion also was associated with increased risk of heart failure hospitalization.
These results remained robust in several sensitivity and subgroup analyses.
No association was found between potassium excretion and any of the CV outcomes, except that higher potassium excretion was related to a reduced risk of stroke.
"Clearly, large randomized controlled trials evaluating the effect of reduced sodium intake ... on CV outcomes in those with moderate sodium intake are needed urgently. Pending the results of such trials, a more cautious approach to policy on sodium intake may be appropriate, one that targets sodium reduction in populations consuming high sodium levels and reflects the uncertainty in those with moderate sodium diets, which includes the majority of the population."
This study was limited in that it involved patients who volunteered to participate in clinical trials, who may have different lifestyles and behaviors than those who declined to participate. Also, urinary samples were taken at a single time point, which may not accurately reflect subjects’ typical sodium and potassium excretion, Dr. O’Donnell and his associates said.
This study was supported by Boehringer Ingelheim, a manufacturer of angiotensin II receptor blockers. Dr. O’Donnell and several associates reported ties to Boehringer Ingelheim, and his associates reported ties to numerous other industry sources.
High urinary sodium excretion, a proxy for high sodium intake, was associated with an increased risk of cardiovascular mortality, myocardial infarction, stroke, and hospitalization for heart failure in a study in the Nov. 23/30 JAMA.
However, low urinary sodium excretion also was associated with increased risk of cardiovascular mortality and hospitalization for heart failure.
"Our findings emphasize the burden of CV disease associated with excess sodium intake and the importance of population-based programs to reduce sodium intake in populations consuming high-sodium diets," Dr. Martin J. O’Donnell of the Population Health Research Institute, McMaster University, Hamilton, Ont., and his associates.
Importantly, the association between sodium intake and adverse CV outcomes did not become significant until intake exceeded 6.5 g/day. This threshold is higher than that recommended by the World Health Organization and by many national guidelines. This highlights "the urgent need to establish a safe range for sodium intake," they noted.
"We found a J-shaped association between estimated sodium excretion and CV events," the researchers noted.
Compared with the "moderate" sodium excretion of 4-5.99 g/day, both higher (more than 7 g/day) and lower (less than 3 g/day) sodium excretion were linked to adverse CV events.
They examined the relationship between sodium intake and CV events using data from two large clinical trials in which patients at high risk for CV events provided urinary samples and were followed for a median of 56 months. Data from the two trials were combined because both recruited subjects from the same areas during the same time period (2001-2004) using the same eligibility criteria and the same methods of obtaining baseline clinical data and monitoring outcomes.
A total of 28,880 study subjects followed at 733 medical centers in 40 countries were included. The subjects were aged 55 and older and either had established CV disease or high-risk diabetes.
Sodium and potassium concentrations were determined from a single morning fasting urine sample from each subject, and this was used to estimate 24-hour sodium and potassium excretion. Overall, the mean estimated 24-hour sodium excretion, a proxy for sodium intake, was 4.77 g, and potassium excretion, a proxy for potassium intake, was 2.19 g.
The composite outcome of CV death, MI, stroke, and hospitalization for heart failure occurred in 4,729 study subjects (16%) during follow-up through 2008. Overall mortality was 12%, the rate of MI was 5%, that of stroke and heart failure hospitalization were each about 4%.
Compared with the moderate excretion of 4-5.99 g/day, higher excretion (more than 7 g/day) was associated with a hazard ratio of 1.15-1.49 for the composite end point. And those with lower excretion (less than 3 g/day), had a hazard ratio of 1.16-1.21 for the composite end point.
In addition, high sodium excretion was significantly associated with each of the individual components of this composite outcome, Dr. O’Donnell and his colleagues said (JAMA 2011;306:2229-38).
"We found a J-shaped association between estimated sodium excretion and CV events."
Lower sodium excretion also was associated with increased risk of heart failure hospitalization.
These results remained robust in several sensitivity and subgroup analyses.
No association was found between potassium excretion and any of the CV outcomes, except that higher potassium excretion was related to a reduced risk of stroke.
"Clearly, large randomized controlled trials evaluating the effect of reduced sodium intake ... on CV outcomes in those with moderate sodium intake are needed urgently. Pending the results of such trials, a more cautious approach to policy on sodium intake may be appropriate, one that targets sodium reduction in populations consuming high sodium levels and reflects the uncertainty in those with moderate sodium diets, which includes the majority of the population."
This study was limited in that it involved patients who volunteered to participate in clinical trials, who may have different lifestyles and behaviors than those who declined to participate. Also, urinary samples were taken at a single time point, which may not accurately reflect subjects’ typical sodium and potassium excretion, Dr. O’Donnell and his associates said.
This study was supported by Boehringer Ingelheim, a manufacturer of angiotensin II receptor blockers. Dr. O’Donnell and several associates reported ties to Boehringer Ingelheim, and his associates reported ties to numerous other industry sources.
FROM JAMA
Major Finding: Compared with the moderate excretion of 4-5.99 g/day, higher excretion (more than 7 g/day) was associated with a hazard ratio of 1.15-1.49 for the composite end point of CV mortality, MI, stroke, and hospitalization for heart failure. And those with lower excretion (less than 3 g/day), had a hazard ratio of 1.16-1.21 for the composite end point.
Data Source: A secondary analysis of data obtained in 2 large clinical trials in which subjects at high risk for CV events contributed urine samples from which sodium and potassium excretion were calculated, and were followed for a median of 56 months.
Disclosures: This study was supported by Boehringer Ingelheim, a manufacturer of angiotensin II receptor blockers. Dr. O’Donnell and several associates reported ties to Boehringer Ingelheim, and his associates reported ties to numerous other industry sources.
Too Much, Too Little Glucose Risky in Diabetics on Dialysis
PHILADELPHIA – Poor glycemic control is associated with decreased survival in diabetes patients on maintenance hemodialysis, according to a study presented at Kidney Week 2011.
Not only does too much glucose increase the risk of all-cause and cardiovascular mortality in this patient population, too little does as well, according to Dr. Kamyar Kalantar-Zadeh of the Harbor-UCLA Medical Center in Torrance, Calif.
Although some guidelines, such as those from the National Kidney Foundation Kidney Disease Outcomes Quality Initiative, recommend that diabetic dialysis patients follow the American Diabetes Association targets for glycemic control (hemoglobin A1c less than 7%), "there is no consistent evidence to support these targets in dialysis patients," Dr. Kalantar-Zadeh said, noting that recent studies with different methodologies have reached opposing conclusions.
To determine the predictive value of glycemic control on mortality, Dr. Kalantar-Zadeh and his colleagues evaluated the association between HbA1c and random serum glucose and survival in a large, contemporary cohort of diabetes patients treated at DaVita dialysis clinics from July 2001 to June 2006 with follow-up through June 2007.
Of 164,789 hemodialysis patients, the investigators identified 54,757 with diabetes for whom HbA1c data were available, and analyzed death hazard ratios according to HbA1c values and serum glucose levels, Dr. Kalantar-Zadeh explained. The adjusted all-cause death hazard ratio for baseline HbA1c increments of 8.0%-8.9%, 9.0%-9.9%, and 10% or greater, respectively, were 1.06, 1.05, and 1.19, compared with a reference HbA1c value of 7.0%-7.9%; the respective all-cause death hazard ratios for time-averaged HbA1c were 1.11, 1.36, and 1.59, he said, noting that "a similarly consistent increase in cardiovascular mortality" was also observed.
Not only does too much glucose increase the risk of all-cause and cardiovascular mortality in this patient population, too little does as well, according to Dr. Kamyar Kalantar-Zadeh.
In fully adjusted models, decreased survival was also directly associated with time-averaged HbA1c levels in the low range, with all-cause-death hazard ratios of 1.05, 1.08, and 1.35, respectively, for baseline HbA1c values of 6.0-6.9%, 5.0-5.9%, and less than 5%, compared with 7.0-7.9%, Dr. Kalantar-Zadeh said. Further, the adjusted all-cause-death hazard ratios for time-averaged blood glucose levels of 175-199 mg/dL, 200-249 mg/dL, 250-299 mg/dL, and 300 mg/dL or higher were 1.03, 1.14, 1.30, and 1.66, respectively, compared with the reference range of 150-175 mg/dL.
The results are limited by the fact that HbA1c levels are known to significantly underestimate glycemic control in hemodialysis patients, Dr. Kalantar-Zadeh acknowledged at the meeting, sponsored by the American Society of Nephrology. Also, "the information on comorbidity in our study was limited to that obtained from Medical Evidence Form 2728, in which comorbid conditions are significantly underestimated," he said. Lastly, "the study data did not include information on diabetes medication used at home or treatment adherence."
Limitations notwithstanding, the findings indicate that poor glycemic control (defined as HbA1c of 8% or greater or a serum glucose level of 200mg/dL) is associated with death in diabetes patients on maintenance hemodialysis, and, unlike most diabetic patients, "those with kidney failure do not benefit from HbA1c levels lower than 7%," Dr. Kalantar-Zadeh said. It appears that the most beneficial percentage is somewhere inside 6-8% in this patient population, "although controlled clinical trials to target HbA1c at 6-8% are needed to determine the optimal range," he concluded.
Dr. Kalantar-Zadeh disclosed financial relationships with Abbott, Amgen, DaVita, Fresenius, Otsuka, Shire, Vifor, the National Institutes of Health, and the National Kidney Foundation. The study was supported by the National Institute of Diabetes and Digestive and Kidney Diseases.
PHILADELPHIA – Poor glycemic control is associated with decreased survival in diabetes patients on maintenance hemodialysis, according to a study presented at Kidney Week 2011.
Not only does too much glucose increase the risk of all-cause and cardiovascular mortality in this patient population, too little does as well, according to Dr. Kamyar Kalantar-Zadeh of the Harbor-UCLA Medical Center in Torrance, Calif.
Although some guidelines, such as those from the National Kidney Foundation Kidney Disease Outcomes Quality Initiative, recommend that diabetic dialysis patients follow the American Diabetes Association targets for glycemic control (hemoglobin A1c less than 7%), "there is no consistent evidence to support these targets in dialysis patients," Dr. Kalantar-Zadeh said, noting that recent studies with different methodologies have reached opposing conclusions.
To determine the predictive value of glycemic control on mortality, Dr. Kalantar-Zadeh and his colleagues evaluated the association between HbA1c and random serum glucose and survival in a large, contemporary cohort of diabetes patients treated at DaVita dialysis clinics from July 2001 to June 2006 with follow-up through June 2007.
Of 164,789 hemodialysis patients, the investigators identified 54,757 with diabetes for whom HbA1c data were available, and analyzed death hazard ratios according to HbA1c values and serum glucose levels, Dr. Kalantar-Zadeh explained. The adjusted all-cause death hazard ratio for baseline HbA1c increments of 8.0%-8.9%, 9.0%-9.9%, and 10% or greater, respectively, were 1.06, 1.05, and 1.19, compared with a reference HbA1c value of 7.0%-7.9%; the respective all-cause death hazard ratios for time-averaged HbA1c were 1.11, 1.36, and 1.59, he said, noting that "a similarly consistent increase in cardiovascular mortality" was also observed.
Not only does too much glucose increase the risk of all-cause and cardiovascular mortality in this patient population, too little does as well, according to Dr. Kamyar Kalantar-Zadeh.
In fully adjusted models, decreased survival was also directly associated with time-averaged HbA1c levels in the low range, with all-cause-death hazard ratios of 1.05, 1.08, and 1.35, respectively, for baseline HbA1c values of 6.0-6.9%, 5.0-5.9%, and less than 5%, compared with 7.0-7.9%, Dr. Kalantar-Zadeh said. Further, the adjusted all-cause-death hazard ratios for time-averaged blood glucose levels of 175-199 mg/dL, 200-249 mg/dL, 250-299 mg/dL, and 300 mg/dL or higher were 1.03, 1.14, 1.30, and 1.66, respectively, compared with the reference range of 150-175 mg/dL.
The results are limited by the fact that HbA1c levels are known to significantly underestimate glycemic control in hemodialysis patients, Dr. Kalantar-Zadeh acknowledged at the meeting, sponsored by the American Society of Nephrology. Also, "the information on comorbidity in our study was limited to that obtained from Medical Evidence Form 2728, in which comorbid conditions are significantly underestimated," he said. Lastly, "the study data did not include information on diabetes medication used at home or treatment adherence."
Limitations notwithstanding, the findings indicate that poor glycemic control (defined as HbA1c of 8% or greater or a serum glucose level of 200mg/dL) is associated with death in diabetes patients on maintenance hemodialysis, and, unlike most diabetic patients, "those with kidney failure do not benefit from HbA1c levels lower than 7%," Dr. Kalantar-Zadeh said. It appears that the most beneficial percentage is somewhere inside 6-8% in this patient population, "although controlled clinical trials to target HbA1c at 6-8% are needed to determine the optimal range," he concluded.
Dr. Kalantar-Zadeh disclosed financial relationships with Abbott, Amgen, DaVita, Fresenius, Otsuka, Shire, Vifor, the National Institutes of Health, and the National Kidney Foundation. The study was supported by the National Institute of Diabetes and Digestive and Kidney Diseases.
PHILADELPHIA – Poor glycemic control is associated with decreased survival in diabetes patients on maintenance hemodialysis, according to a study presented at Kidney Week 2011.
Not only does too much glucose increase the risk of all-cause and cardiovascular mortality in this patient population, too little does as well, according to Dr. Kamyar Kalantar-Zadeh of the Harbor-UCLA Medical Center in Torrance, Calif.
Although some guidelines, such as those from the National Kidney Foundation Kidney Disease Outcomes Quality Initiative, recommend that diabetic dialysis patients follow the American Diabetes Association targets for glycemic control (hemoglobin A1c less than 7%), "there is no consistent evidence to support these targets in dialysis patients," Dr. Kalantar-Zadeh said, noting that recent studies with different methodologies have reached opposing conclusions.
To determine the predictive value of glycemic control on mortality, Dr. Kalantar-Zadeh and his colleagues evaluated the association between HbA1c and random serum glucose and survival in a large, contemporary cohort of diabetes patients treated at DaVita dialysis clinics from July 2001 to June 2006 with follow-up through June 2007.
Of 164,789 hemodialysis patients, the investigators identified 54,757 with diabetes for whom HbA1c data were available, and analyzed death hazard ratios according to HbA1c values and serum glucose levels, Dr. Kalantar-Zadeh explained. The adjusted all-cause death hazard ratio for baseline HbA1c increments of 8.0%-8.9%, 9.0%-9.9%, and 10% or greater, respectively, were 1.06, 1.05, and 1.19, compared with a reference HbA1c value of 7.0%-7.9%; the respective all-cause death hazard ratios for time-averaged HbA1c were 1.11, 1.36, and 1.59, he said, noting that "a similarly consistent increase in cardiovascular mortality" was also observed.
Not only does too much glucose increase the risk of all-cause and cardiovascular mortality in this patient population, too little does as well, according to Dr. Kamyar Kalantar-Zadeh.
In fully adjusted models, decreased survival was also directly associated with time-averaged HbA1c levels in the low range, with all-cause-death hazard ratios of 1.05, 1.08, and 1.35, respectively, for baseline HbA1c values of 6.0-6.9%, 5.0-5.9%, and less than 5%, compared with 7.0-7.9%, Dr. Kalantar-Zadeh said. Further, the adjusted all-cause-death hazard ratios for time-averaged blood glucose levels of 175-199 mg/dL, 200-249 mg/dL, 250-299 mg/dL, and 300 mg/dL or higher were 1.03, 1.14, 1.30, and 1.66, respectively, compared with the reference range of 150-175 mg/dL.
The results are limited by the fact that HbA1c levels are known to significantly underestimate glycemic control in hemodialysis patients, Dr. Kalantar-Zadeh acknowledged at the meeting, sponsored by the American Society of Nephrology. Also, "the information on comorbidity in our study was limited to that obtained from Medical Evidence Form 2728, in which comorbid conditions are significantly underestimated," he said. Lastly, "the study data did not include information on diabetes medication used at home or treatment adherence."
Limitations notwithstanding, the findings indicate that poor glycemic control (defined as HbA1c of 8% or greater or a serum glucose level of 200mg/dL) is associated with death in diabetes patients on maintenance hemodialysis, and, unlike most diabetic patients, "those with kidney failure do not benefit from HbA1c levels lower than 7%," Dr. Kalantar-Zadeh said. It appears that the most beneficial percentage is somewhere inside 6-8% in this patient population, "although controlled clinical trials to target HbA1c at 6-8% are needed to determine the optimal range," he concluded.
Dr. Kalantar-Zadeh disclosed financial relationships with Abbott, Amgen, DaVita, Fresenius, Otsuka, Shire, Vifor, the National Institutes of Health, and the National Kidney Foundation. The study was supported by the National Institute of Diabetes and Digestive and Kidney Diseases.
FROM KIDNEY WEEK 2011
Major Finding: The risk for all-cause mortality among diabetes patients on maintenance hemodialysis was 11%-59% higher in those with HbA1c greater than 8% than in those with HbA1c of 7.0-7.9%.
Data Source: Retrospective analysis of a contemporary cohort of 54,757 diabetes patients treated at DaVita dialysis clinics from July 2001 to June 2006 with follow-up through June 2007.
Disclosures: Dr. Kalantar-Zadeh disclosed financial relationships with Abbott, Amgen, DaVita, Fresenius, Otsuka, Shire, Vifor, the National Institutes of Health, and the National Kidney Foundation. The study was supported by the National Institute of Diabetes and Digestive and Kidney Diseases.



