9
Pharmacokinetics of bumetanide in critically ill infants Object&: Define the pharmacokinetics of bumetanide after single intravenous doses in volume-overloaded critically ill infants. Methods: A prospective, open-label study was carried out in a group of 58 infants aged 0 to 6 months who required diuretic therapy. Each patient received a single dose of intravenous bumetanide. Doses selected in sequential order ranged from 0.005 to 0.10 mg/kg. Hematologic and serum chemistry studies were performed before and at 6 and 24 hours after bumetanide administration. Determinations of urine volume and chemistries were performed before (collected from - 2 to -4 hours to time 0) and at 1,2,3,4,6, and 12 hours after bumetanide dosing. Serum samples collected at time 0 and at 5,15,30,60,120,180,240, 360, and 480 minutes and urine collected at time 0 and at 0 to 1, 1 to 2, 2 to 3, 3 to 4,4 to 6, and 6 to 12 hours were analyzed for bumetanide concentration. Data were evaluated by standard noncompartmen- tal pharmacokinetic techniques. Resdts: Peak serum bumetanide concentrations occurred at 5 minutes after bumetanide administration. Area under the curve and peak serum bumetanide concentrations showed linear increases over the twen- tyfold dose range; whereas beta volume of distribution, volume of distribution at steady state, clearance, renal clearance, half-life, and mean residence time values were independent of dose. Peak urinary excretion rates of bumetanide increased linearly with increasing doses. The mean percent of bumetanide recovered in the urine from 0 to 12 hours was 40% + 15% of the administered dose. Conclwions: Distribution and elimination kinetics of bumetanide were similar in all patients. Elimination kinetics were first order over the dose range of 0.005 to 0.10 mg/kg. Pharmacokinetic parameter estimates (beta volume of distribution, volume of distribution at steady state, clearance, renal clearance, half-life, and mean residence time) were independent of the dose of bumetanide administered. Single doses of bumetanide up to 0.10 mg/kg appear to be well tolerated in acutely ill volume-overloaded infants aged 0 to 6 months. (Clin Pharmacol Ther 1996;60:405-13.) Janice E. Sullivan, MD,” Madolin K. Witte, MD,b Toyoko S. Yamashita, PhD, Carolyn M. Myers, PhD, and Jeffrey L. Blumer, PhD, MD Cleveland, Ohio From the Departments of Pediatrics, Pharmacology, and Epide- miology and Biostatistics, Case Western Reserve University; and the Division of Pediatric Pharmacology and Critical Care, Rainbow Babies and Childrens Hospital. Supported in part by a grant from Roche Laboratories, through the Children’s Research Foundation of Cleveland and Pediat- ric Pharmacology Research Unit Grant (HD 31323-02). Portions of this work were presented at Society for Pediatric Research (Washington, D.C., 1986), International Congress of Pharmacology (Stockholm, 1986 and Montreal, 1994), and American Society for Clinical Pharmacology and Therapeutics (New Orleans, 1994 and San Diego, 1995). Received for publication June 27, 1995; accepted May 20, 1996. Reprint requests: Jeffrey L. Blumer, PhD, MD, Rainbow Babies and Childrens Hospital, Department of Pediatrics, Division of Pediatric Pharmacology and Critical Care, Mailstop 6010, 11100 Euclid Ave., Cleveland, OH 44106. aPresent address: Department of Pediatrics, University of Louis- ville, Louisville, KY 40292. Pathologic fluid retention in critically ill infants is frequently associated with congestive heart failure, pulmonary disease, renal disease, or sepsis with cap- illary 1eak.l Diuresis is often required to improve hemodynamics, facilitate weaning a patient from mechanical ventilation, and maintain or establish adequate urinary output in a patient with oliguria. Although medical treatment should focus primarily on correcting the underlying disorder causing fluid retention, judicious administration of diuretic agents is often needed to supplement meticulous bPresent address: Department of Pediatrics, University of Utah Medical Center, Salt Lake City, UT 84132. Copyright 0 1996 by Mosby-Year Book, Inc. 0009-9236/96/$5.00 + 0 13/l/75214 405

Pharmacokinetics of bumetanide in critically ill infants*

Embed Size (px)

Citation preview

Page 1: Pharmacokinetics of bumetanide in critically ill infants*

Pharmacokinetics of bumetanide in critically ill infants

Object&: Define the pharmacokinetics of bumetanide after single intravenous doses in volume-overloaded critically ill infants. Methods: A prospective, open-label study was carried out in a group of 58 infants aged 0 to 6 months who required diuretic therapy. Each patient received a single dose of intravenous bumetanide. Doses selected in sequential order ranged from 0.005 to 0.10 mg/kg. Hematologic and serum chemistry studies were performed before and at 6 and 24 hours after bumetanide administration. Determinations of urine volume and chemistries were performed before (collected from - 2 to -4 hours to time 0) and at 1,2,3,4,6, and 12 hours after bumetanide dosing. Serum samples collected at time 0 and at 5,15,30,60,120,180,240, 360, and 480 minutes and urine collected at time 0 and at 0 to 1, 1 to 2, 2 to 3, 3 to 4,4 to 6, and 6 to 12 hours were analyzed for bumetanide concentration. Data were evaluated by standard noncompartmen- tal pharmacokinetic techniques. Resdts: Peak serum bumetanide concentrations occurred at 5 minutes after bumetanide administration. Area under the curve and peak serum bumetanide concentrations showed linear increases over the twen- tyfold dose range; whereas beta volume of distribution, volume of distribution at steady state, clearance, renal clearance, half-life, and mean residence time values were independent of dose. Peak urinary excretion rates of bumetanide increased linearly with increasing doses. The mean percent of bumetanide recovered in the urine from 0 to 12 hours was 40% + 15% of the administered dose. Conclwions: Distribution and elimination kinetics of bumetanide were similar in all patients. Elimination kinetics were first order over the dose range of 0.005 to 0.10 mg/kg. Pharmacokinetic parameter estimates (beta volume of distribution, volume of distribution at steady state, clearance, renal clearance, half-life, and mean residence time) were independent of the dose of bumetanide administered. Single doses of bumetanide up to 0.10 mg/kg appear to be well tolerated in acutely ill volume-overloaded infants aged 0 to 6 months. (Clin Pharmacol Ther 1996;60:405-13.)

Janice E. Sullivan, MD,” Madolin K. Witte, MD,b Toyoko S. Yamashita, PhD, Carolyn M. Myers, PhD, and Jeffrey L. Blumer, PhD, MD Cleveland, Ohio

From the Departments of Pediatrics, Pharmacology, and Epide- miology and Biostatistics, Case Western Reserve University; and the Division of Pediatric Pharmacology and Critical Care, Rainbow Babies and Childrens Hospital.

Supported in part by a grant from Roche Laboratories, through the Children’s Research Foundation of Cleveland and Pediat- ric Pharmacology Research Unit Grant (HD 31323-02).

Portions of this work were presented at Society for Pediatric Research (Washington, D.C., 1986), International Congress of Pharmacology (Stockholm, 1986 and Montreal, 1994), and American Society for Clinical Pharmacology and Therapeutics (New Orleans, 1994 and San Diego, 1995).

Received for publication June 27, 1995; accepted May 20, 1996. Reprint requests: Jeffrey L. Blumer, PhD, MD, Rainbow Babies

and Childrens Hospital, Department of Pediatrics, Division of Pediatric Pharmacology and Critical Care, Mailstop 6010, 11100 Euclid Ave., Cleveland, OH 44106.

aPresent address: Department of Pediatrics, University of Louis- ville, Louisville, KY 40292.

Pathologic fluid retention in critically ill infants is frequently associated with congestive heart failure, pulmonary disease, renal disease, or sepsis with cap- illary 1eak.l Diuresis is often required to improve hemodynamics, facilitate weaning a patient from mechanical ventilation, and maintain or establish adequate urinary output in a patient with oliguria. Although medical treatment should focus primarily on correcting the underlying disorder causing fluid retention, judicious administration of diuretic agents is often needed to supplement meticulous

bPresent address: Department of Pediatrics, University of Utah Medical Center, Salt Lake City, UT 84132.

Copyright 0 1996 by Mosby-Year Book, Inc. 0009-9236/96/$5.00 + 0 13/l/75214

405

Page 2: Pharmacokinetics of bumetanide in critically ill infants*

406 Sullivan et al. CLINICAL PHAPAfACOLOGY &THERAPEUTICS

OCTOBER 1996

fluid and electrolyte management to attain or main- tain homeostasis in these patients.‘,’

Loop diuretics are the most frequently adminis- tered diuretic agents in volume-overloaded infants because they prevent reabsorption of ~15% of fil- tered sodium and produce a greater diuresis than other types of diuretics.3 The prototype of this class, furosemide, is used most commonly to treat fluid overload in infants and children.2 However, bu- metanide (3-n-butylamino-4-phenoxy-5-sulfamoyl- benzoic acid), a loop diuretic chemically related to furosemide, may be less toxic4 Other potential ther- apeutic advantages of bumetanide include greater potency than furosemide,5,6 decreased displacement of bilirubin from albumin at presumed therapeutic concentrations of each drug,7-9 less potential for ototoxicity,lO~ll and significantly lower potassium ex- cretion.5,12 Except for one study of infants receiving extracorporeal membrane oxygenation,13 the phar- macokinetics of bumetanide in children are de- scribed only in abstracts of studies performed in preterm or term newborn infants.14’15 Present pub- lished data are inadequate to permit design of an effective, safe diuretic regimen for infants.

This is the first of three articles describing the pharmacokinetics and pharmacodynamics of bumet- anide and factors affecting the response to this agent in acutely ill, fluid-overloaded infants up to 6 months of age. Here, we report the pharmacokinet- its of bumetanide over a twentyfold dosage range.

METHODS Patients. Critically ill infants from birth to 6

months of age with fluid retention and clinical indications for diuretic therapy were considered for enrollment. Fluid retention was evidenced by the presence of edema, excessive weight gain, signs of congestive heart failure, or pulmonary edema. Primary physicians determined the need for diuretic therapy before patient enrollment. Patients were screened by an investigator who obtained written consent for participation from their parents or legal guardians. Exclusion criteria included: treatment with a diuretic agent within the previous 24 hours, New York Heart Associa- tion cardiac status/prognosis Class IV,16 history of hearing impairment or concurrent exposure to other ototoxic agents (e.g., aminoglycosides, am- photericin), clinically significant hypotension (~2 standard deviations below normal for age), poly- cythemia (hematocrit r60%), allergy to sulfon- amide derivatives, significant acid-base or electro-

lyte imbalance, clinical and laboratory evidence of significant renal impairment (serum creatinine ~2.5 mg/dl, anuria, or both), or evidence of mul- tiorgan system failure. After approval of the pro- tocol by the Institutional Review Board of the University Hospitals of Cleveland, this study was per- formed in the Pediatric Intensive Care Unit at Rain- bow Babies and Childrens Hospital, Cleveland, Ohio.

Study design. In this open-label, dose-ranging study protocol, each patient received a single dose of bumetanide intravenously over 1 to 2 minutes. Doses were selected in a sequential order starting with 0.005 mg/kg and proceeding sequentially to 0.01, 0.015, 0.02, 0.025, 0.03, 0.035, 0.05, and 0.10 mg/kg. The maximal dose (0.10 mg/kg) was based on the usual pediatric dose of furosemide (1 to 2 mg/ kg), reportedly twentyfold to fortyfold less potent than bumetanide in adults.5

Blood samples for serum chemistries and hema- tologic studies were drawn before the administra- tion of bumetanide and repeated 6 and 24 hours thereafter to assess renal and hepatic status and drug safety. Analyses included a complete blood count with differential and determinations of serum sodium, potassium, chloride, bicarbonate, urea ni- trogen, creatinine, aspartate aminotransferase, ala- nine aminotransferase, lactate dehydrogenase, alka- line phosphatase, total and direct bilirubin, total protein, albumin, calcium, magnesium, glucose, and uric acid levels and osmolality.

Serial blood samples for analysis of serum bumet- anide concentration were drawn from an indwelling arterial or venous catheter before and at 5, 15, 30, 60, 120, 180, 240, 360, and 480 minutes after the bumetanide bolus injection. Serum obtained after centrifugation of these samples was stored at -70” C for later processing and assay.

Urine was collected for a 2- to 4-hour period before the bumetanide dose and for timed intervals thereafter (0 to 1, 1 to 2, 2 to 3, 3 to 4, 4 to 6, and 6 to 12 hours). Urine volumes were recorded, and aliquots were analyzed for sodium, potassium, chlo- ride, calcium, magnesium, and creatinine levels and osmolality. Aliquots of urine for bumetanide con- centration determination were stored at -70” C un- til they were analyzed. Most patients had Foley catheters in place during the study. Diapers from each patient were weighed before and after use to assure accurate urine collection.

Creatinine clearance (CL,,) was used to estimate the glomerular filtration rate for each patient ac- cording to the following formula:

Page 3: Pharmacokinetics of bumetanide in critically ill infants*

CLINICAL PHARMACOLOGY &THERAPEUTICS VOLUME 60, NUMBER4 Sullivan et al. 407

[UCR] CkR (ml/min/1.73 m’) = ,ScRl X

Urine output X 1.73 TXBSA

in which [U,,] and [S,,] represent urine and serum concentrations of creatinine (in milligrams per de- ciliter), respectively, T is the collection time (in minutes), and BSA is body surface area (in square meters). Creatinine clearance was calculated on the basis of both body weight and body surface area. Creatinine clearances determined by using pretreat- ment, posttreatment, and combined pretreatment and posttreatment urine samples did not differ sig- nificantly from each other (p > 0.05); accordingly, CL,, was calculated from the combined pretreat- ment and posttreatment urine samples for each pa- tient. Timed urine samples for CL,, determination were collected for a minimum of 6 hours. Values calculated for CL, were compared with normal values reported for infants of the same age.17’18

Blood pressure, heart rate, and respiratory rate were monitored continuously in each patient and recorded hourly for 2 to 4 hours before the dose of bumetanide was administered and for at least 6 hours after bumetanide administration. Fluid man- agement for each patient before and during the study period was determined by the primary physi- cian.

Determination of bumetanide concentrations in se- rum and urine. Bumetanide analyses were per- formed by HPLC. Serum samples (0.4 ml) were combined with 0.1 ml of internal standard (0.5 @ml R021-1825 supplied by Roche Laboratories, Nutley, N.J.) and 1.0 ml of 1.0 mol/L potassium citrate, pH 5.0, in 13 X 100 mm screw cap borosili- cate tubes. Mixtures were vortexed and allowed to sit for 1 minute, after which 4.0 ml of ethyl acetate/ cyclohexane (70:30) was added. The contents were mixed for 10 minutes on a Rototorque mixer (Cole Parmer Instrument Co., Niles, Ill.) at 60 rpm and centrifuged at 11508 for 1 minute. The upper or- ganic layer was removed and evaporated to dryness under reduced pressure. The solid residue was re- dissolved in 0.125 ml of mobile phase, and 0.05 ml was injected onto the HPLC column for analysis. The assay was standardized against identically pro- cessed sera to which known amounts of bumetanide were added.

Urine samples were centrifuged for 1 minute at 16,OOOg in an Eppendorf microcentrifuge (Eppen- dorf North America Inc., Madison, Wis.) to remove particulate matter. One volume of urine supernatant was mixed with 1 volume of 0.8 mol/L potassium

formate, pH 3.9, and the resulting solution was an- alyzed for bumetanide concentration. This assay was standardized by processing urine samples to which known amounts of bumetanide were added before storage at -70” C.

Bumetanide concentrations were determined by chromatography on a Varian Instruments model 5500 liquid chromatograph (Varian Sample Prepa- ration Products, Harbor City, Calif.) equipped with a 30 cm X 4 mm Zorbax C-8 reversed-phase column (MAC-MOD Analytical Inc., Chadds Ford, Pa.) maintained at 40” C. Peaks were eluted with a mo- bile phase of methanol (0.03 mol/L potassium phos- phate, pH 2.5 [63:37, vol/vol] at a flow rate of 1 ml/min) and detected with a Varian 9070 fluores- cence detector set at a wavelength of 340 nm for excitation and 440 nm for emission. Peak areas were integrated by a DS 604 computer (Varian Analytical Instruments, San Fernando, Calif.) and printed on a Hewlett-Packard Think Jet recorder (Hewlett- Packard Co., Palo Alto, Calif.). Retention times were approximately 6.8 and 8.6 minutes for bumet- anide and the internal standard, respectively.

Serum and urine assays were linear with respect to bumetanide concentrations from 3 to 200 rig/ml (r = 0.999) and from 10 to 500 rig/ml (Y = 0.997), respectively. Bumetanide recovery was 74.3% + 5.9% from serum and 97.1% 2 7.2% from urine. The mean intraassay and interassay coefficients of variation were 4.6% and 6.3% for serum and 5.6% and 4.7% for urine.

Pharmacokinetic analysis. The time-dependent disposition of bumetanide was subjected to standard noncompartmental pharmacokinetic analysis.” Se- rum bumetanide concentration versus 0- to g-hour time curves were plotted for each patient on a semi- logarithmic scale. To compare the serum concentra- tion versus time patterns of the nine dose groups of bumetanide, a composite serum concentration ver- sus time curve was constructed. To accomplish this, the peak bumetanide concentration for each patient was arbitrarily assigned the value of 100%. Each subsequent serum concentration was then expressed as a percentage of the peak for that patient, after which the mean percent for each dose group at each time point was determined. The mean percent of maximal bumetanide concentration for each dose group at each time point was then used to determine the mean t SD for the entire study population at each time point, and these values were then plotted on a graph as a function of time. This graph was not used to derive pharmacokinetic parameters but sim-

Page 4: Pharmacokinetics of bumetanide in critically ill infants*

408 Sdlivan et al. CLINICAL PJSARMA COLOGY & THERAPEUTICS

OCTOBER 1996

Table I. Patient characteristics (n = 58) 1400 1 1200-

" lOOO- E B 5 800-

ii E 600- .

0

I 0.02 0.04 0.06 0.08 0.10 0.12

Dose (mglkg)

Fig. 1. Dose-dependence of peak bumetanide serum concentrations. Bumetanide serum concentration was maximal in all patients when the first sample was obtained 5 minutes after bumetanide administration. Peak serum concentrations (C,,) were plotted against dose of bumet- anide administered (in milligrams per kilogram) for each patient. The number of points is actually the same as the number of patients shown in Table II for each dose group but may appear to be lower because of substantial overlap of similar C,, values. C,, was linear and proportional to dose over the entire dose range (R2 = 0.66;~ < 0.001).

ply to illustrate the serum concentration versus time curve for all patients.

Pharmacokinetic parameters were estimated for each patient by the following methods. The area under the serum drug concentration-time curve (AUC) was determined for each patient by the lin- ear trapezoidal rule up to the time that the final concentration was measured and was then extrapo- lated to infinity [AUC(,_,)]. Linear regression was used to estimate the elimination rate constant (k,) from the postdistributive terminal slope of the log serum concentration versus time plot. Elimination half-life (t& and mean residence time (MRT) were calculated as 0.693/k, and AUMC/AUC, respec- tively, in which AUMC represents the area under the moment curve.

Total (CLr) and renal (C&J clearances of bu- metanide were determined for each patient by the following equations:

CL, = Dose/[AUC(O-m)]

and

Parameters Mean + SD Range

Age (mo> 2.16 5 1.63 0.13-5.78 Sex (M/F) 38120 NA Weight (kg) 4.0 + 1.3 2.3-7.5 BUN (mgidl) 10 ? 6 1-24 Creatinine (mg/dl) 0.5 2 0.2 0.1-1.3 Albumin (mg/dl) 3.2 r 0.7 1.8-5.1 CL,, (ml/minikg) 2.2 + 1.7 0.5-8.1 CL,, (ml/min/1.73 m2) 64 -+ 50 15-248

NA, Not applicable; BUN, Blood urea nitrogen; CL,,, creatinine clear- ance.

CL, = AJ[AUC(O-a)]

in which A, represents the cumulative amount ex- creted during the sampling interval. A, was interpo- lated for the S-hour time point because urine was collected from 6 to 12 hours, whereas serum bumet- anide concentration was determined at 8 hours after the dose was given. Nonrenal clearance (CL& was estimated by subtracting renal clearance from total clearance. Beta volume of distribution (VP) and vol- ume of distribution at steady state (V,,) were esti- mated, respectively, from the following equations:

and v, = C&/k,

V,, = (Dose X AUMC)/AUC2

Values for Cl+ CL,, CL,,, V,, and Vss were corrected for body weight.

The excretion rate of bumetanide (in micrograms per kilogram per hour) for each collection interval was determined by dividing the amount of bumet- anide in the urine during the sampling interval by the collection time and weight of each patient. The cumulative amount of drug eliminated in the urine from 0 to 12 hours was divided by the total intrave- nous dose to determine the percent of the dose excreted in the urine over that time interval.

Statistical analysis. Pharmacokinetic parameter estimates (V,, Vss, tm+, MRT, CLr, CLn, and CL& for each patient were graphed as a function of dose and evaluated by regression and one-way analysis of variance to determine their relationship to dose. The accepted level of significance wasp < 0.05. By using power analysis2’ and on the basis of our pharmacokinetic data, we estimated that six patients were required per dose group (80% power to detect a difference of 20% between the 9 treat- ment groups, (Y = 0.05).

Page 5: Pharmacokinetics of bumetanide in critically ill infants*

CLINICAL PHARMA COLOGY &THERAPEUTICS VOLUME 60, NUMBER4 Sullivan etal. 409

1 I 0 2 4 6 a

Time (hours)

Fig. 2. Composite serum bumetanide concentration ver- sus time curve. Peak bumetanide concentrations for each patient (from Fig. 1) were assigned the value of lOO%, and the curve was constructed as explained in Methods. Bu- metanide elimination appeared to follow first-order kinet- ics over the dose range studied.

RESULTS Study population. Of the 71 patients initially en-

rolled in the study, 13 were eliminated for the fol- lowing reasons: inadequate urine collections (n = 9), age >6 months (TZ = l), oliguria with worsening congestive heart failure requiring additional diuretic therapy during the sampling period (n = l), phar- macokinetic data >3 standard deviations from the mean of other patients in the same dose group (n = l), and inadvertent double enrollment (n = 1). In the last instance, only data from the first study are included.

The final study group consisted of 58 infants (Ta- ble I) who fell into three categories: postoperative repair or palliation of congenital heart defects (n = 31), pulmonary disorders (n = 22), or other disor- ders (n = 5), each associated with volume overload requiring parenteral diuretic therapy. However, all patients had normal renal and hepatic function on entry into the study as determined by CL,,, serum transaminase, bilirubin values, and hepatic synthetic function (prothrombin time and partial thrombo- plastin time). The study population was also nutri- tionally replete, as evidenced by normal values for serum albumin and total protein. Four patients had serum albumin values ~2.5 mg/dl, but only one of these patients had a value ~2.0 mg/dl; low serum

0.02 0.04 0.06 0.08 0.10 0.12

Dose (mglkg)

Fig. 3. Dose dependence of peak urinary bumetanide ex- cretion, rate. Peak bumetanide urinary excretion rate for each patient was plotted against dose of bumetanide ad- ministered (in milligrams per kilogram). Peak bumetanide excretion rates appeared linear and proportional to dose (P = 0.74; p < 0.001).

albumin concentrations were attributed to volume overload rather than to poor nutritional status. No adverse effects of bumetanide (e.g., hemodynamic instability, serum electrolyte abnormalities, signifi- cant elevations of bilirubin, or allergic reactions) were noted in any patient.

Pharmacokinetics. Both peak serum concentra- tions of bumetanide (Fig. 1; R2 = 0.66, p < 0.001) and AUC (data not shown) showed linear and pro- portional increases with respect to dose over the twentyfold dose range. Serum bumetanide concen- trations were maximal in all patients 5 minutes after bumetanide administration and decreased rapidly, irrespective of the dose administered, to approxi- mately 25% of the peak concentration by 1 hour after the dose (Fig. 2). Bumetanide elimination fol- lowed first-order kinetics in all patients over the dosage range studied.

Pharmacokinetic parameter estimates for V,, Vss, Cb, Ch, tliz, and MRT did not differ significantly among dose groups (Table II). Peak serum concentra- tions and V, did not differ significantly among patients with serum albumin ~2.5 mg/dl compared with those with serum albumin >2.5 mg/dl (p > 0.65). Estimates of pharmacokinetic parameters showed marked inter- patient and intergroup variations, which did not reach statistical significance.

Urinary excretion of bumetanide achieved maxi- mal values in all patients by 3 hours after the dose,

Page 6: Pharmacokinetics of bumetanide in critically ill infants*

4 10 Sullivan et al. CLINICAL PHARMA COLOGY & THERAPEUTICS

OCTOBER 1996

Table II. Estimates of pharmacokinetic parameters by dose group after a single intravenous dose of bumetanide

Subjects Dose (4 h&d (L2g)

CL, (mllminlkg)

CL, (mllminlkg)

t1/2

(W MRT @r)

6 0.005 0.34 t 0.14 0.25 ? 0.11 2.63 ? 1.23 1.04 + 0.45 1.62 2 0.47 1.71 2 0.51 6 0.010 0.26 ? 0.07 0.20 2 0.07 1.40 +- 0.75 0.72 2 0.69 2.65 + 1.41 2.92 k 2.05 6 0.015 0.30 2 0.10 0.25 2 0.10 1.63 ? 0.84 0.63 2 0.36 3.03 + 2.48 3.73 + 3.34 6 0.020 0.24 k 0.04 0.20 i. 0.02 1.94 t 1.24 0.85 2 0.81 2.03 + 1.35 2.72 + 2.26 6 0.025 0.43 t 0.21 0.35 + 0.14 2.68 k 1.44 1.30 If: 0.76 2.06 2 0.78 2.59 +- 1.35 6 0.030 0.42 2 0.15 0.35 + 0.12 3.14 k 2.55 1.10 t 0.77 2.52 k 1.73 3.23 2 2.40 7 0.035 0.39 2 0.10 0.32 !I 0.09 3.34 2 1.54 1.09 2 0.90 1.57 + 0.75 1.84 2 0.90 8 0.050 0.60 t 0.42 0.39 2 0.17 3.82 2 3.14 1.61 21 1.46 2.55 2 1.49 3.05 c 2.38 7 0.100 0.43 2 0.17 0.28 ? 0.07 3.49 2 2.27 1.33 t 0.83 2.12 2 1.47 2.43 5 2.28

Mean 2 SD 0.39 t 0.21 0.29 + 0.12 2.74 2 1.95 1.10 2 0.86 2.34 2 1.41 2.68 -t- 2.04

V,, Beta volume of distribution; Ch, total clearance; CL, renal clearance; t,,2, half-life; MRT, mean residence time.

and in the majority of patients, by 1 or 2 hours after the dose. Peak urinary bumetanide excretion rates (in micrograms per kilogram per hour) demon- strated a linear relationship to dose (I?’ = 0.74;~ < O.OOl), and showed no evidence of approaching a maximum (Fig. 3). The greatest percentage of the administered dose of bumetanide was recovered in the urine within the first hour after bumetanide administration and declined steadily thereafter. The mean percent of bumetanide recovered in the urine for all patients by 12 hours was 40% ? 15% of the administered dose with a range of 10% to 83% (Fig. 4). No relationship was evident between the dose administered and the total percent of bumetanide recovered by 12 hours.

DISCUSSION This is the first study of the effect of dose on the

pharmacokinetics of bumetanide in volume- overloaded critically ill infants during the first 6 months of life. The large size of our patient popu- lation and the number of patients in each dosage group permit identification of certain pharmacoki- netic patterns despite large interindividual and in- tergroup variations in estimates of pharmacokinetic parameters in this heterogeneous population. The primary findings were that peak serum concentra- tion, AUC, and bumetanide excretion rate increased linearly with increasing doses of bumetanide; whereas V,, Vss, CLr, CL,, t,,,, and MRT did not vary significantly. Elimination kinetics of bumet- anide were first order. About 40% of the drug was cleared through renal elimination, and 10% to 83% of the administered dose was recovered in the urine during the 12 hours immediately after the dose.

Studies in adult patients have demonstrated about 50% to 70% recovery of unchanged bumetanide in the urine over 24 to 48 hours after intravenous administration of bumetanide.21‘23 Because bumet- anide was still detected in the 6- to 12-hour urine sample from most of our patients, total urinary re- covery of bumetanide may have been underesti- mated. The percent of drug recovered might have been similar to that reported for adults if we had collected urine for 24 hours after administration of bumetanide.

Estimates of certain pharmacokinetic parameters in our study differ from others published in abstract form. Lopez et a1.15 reported serum concentrations of bumetanide twofold to sixfold higher than we found with comparable doses and identical sampling intervals during the bumetanide elimination phase. Their values are also much greater than those re- ported by Cook et a1.24 in adults who received an average of 0.045 mg/kg. Estimated CLcu was 30% of adult values, and CL, of bumetanide was much less (range, 0.2 to 1.1 ml/minikg) in the infants studied by Lopez et al. l5 than in either our patients or adults24 (2.74 t 1.95 ml/min/kg and 2.55 + 0.54 ml/mm/kg, respectively). A reduction in clearance (about fourfold) may account for the large differ- ences in reported serum concentrations between groups. Similar to our findings, Lopez et a1.13 also reported large interpatient variability in serum bu- metanide concentrations in their infant population. Peak serum bumetanide concentrations in our pa- tients showed a linear increase with increasing dose and substantial variability within each dose group. Other investigators14715 found values for V, and Vs, similar to those found in our patients, but their

Page 7: Pharmacokinetics of bumetanide in critically ill infants*

CLINICAL P HARMACOLOGY &THERAPEUTICS VOLUME 60, NUMBER4 Sullivan etal. 411

o-1 l-2 2-3 3-4 4-6 6-12 o-12

Time (hours)

Fig. 4. Urinary recovery of bumetanide. Light bars on the left represent percentages of total dose of administered bumetanide recovered per hour for each collection interval. The dark bar on the right depicts the mean t SD of the total percentage of bumetanide dose recovered from 0 to 12 hours (40% +- 15%) after administration of bumetanide (n = 58).

bumetanide clearances were reduced (0.83 2 0.83 ml/min/kg versus 2.74 + 1.95 ml/min/kg, respective- ly), and plasma t,,, values were increased (5.8 t 0.7 versus 2.34 t 1.41 hours, respectively). These dif- ferences may reflect the relative immaturity of bu- metanide clearance mechanisms manifested by pre- term infants included in their studies. The actual dose of bumetanide administered in their studies was not validated by measurement of bumetanide excretion rates, as was done in our study.

Bumetanide clearances for infants in our study were similar to those reported for healthy adults (2.74 + 1.95 ml/mm/kg versus 2.55 + 0.54 mUmin/ kg), 21,24,25 although the variability was much greater. This may reflect the ontogeny of renal and hepatic function. However, the volume of distribution (Vs,) was about twofold greater in our infants (0.29 ? 0.12 L/kg versus 0.13 +- 0.03 L/kg), thereby largely ac- counting for the longer observed elimination t,,, (2.34 t 1.41 hours versus 0.8 + 0.2 hours). The reason for the larger volume of distribution for bu- metanide in infants as compared with adults is not known. Protein binding of bumetanide (97%) is re- portedly similar in the two populations, but even an experimentally undetectable decrease in bumet- anide binding to plasma proteins could substantially increase values for this parameter in our patients.26 Also, the greater volume of extracellular fluid per kilogram of body weight available for bumetanide distribution in infants as compared with adults (350

to 440 ml/kg versus 200 ml/kg, respectively)27 may contribute to the larger volume of bumetanide dis- tribution in infants.

In infants major differences exist between the pharmacokinetics of bumetanide and furosemide, the prototype of loop diuretics. The t,,, of bumet- anide (2.34 + 1.41 hours) is much shorter than that of furosemide (13.4 +- 8.6 hours).28 This nearly six- fold difference is largely due to a greater clearance of bumetanide (2.74 2 1.95 ml/min/kg versus 0.20 t 0.16 ml/min/kg for furosemide28), even though smaller V, (0.39 + 0.21 versus 0.52 2 0.42 L/kg for furosemide2’) contributes to it.29”1 This suggests that more frequent drug administration is needed with bumetanide than furosemide, depending on their relative pharmacodynamic effects.

The highest single dose of bumetanide chosen for our study, 0.10 mg/kg, produced no adverse effects attributable to the drug, namely electrolyte abnor- malities, hemodynamic instability, hypovolemia, or significant elevations in bilirubin. Long-term man- agement (2 to 40 weeks) with oral doses of bumet- anide up to 0.1 mg/kg/day has been reported to be an effective adjunct to therapy designed to control congestive heart failure in pediatric patients, and without demonstrable toxicity.32 However, possible ototoxicity was not evaluated in either study.

In conclusion, bumetanide exhibited first-order elimination kinetics after single doses ranging from 0.005 to 0.10 mg/kg in acutely ill, fluid-overloaded

Page 8: Pharmacokinetics of bumetanide in critically ill infants*

412 Sadivan et al,

infants aged 4 days to 6 months. Pharmacokinetic parameter estimates were independent of the bu- metanide dose administered, whereas peak urinary excretion rates of bumetanide were proportional to dose over the range studied with no evidence for saturation of bumetanide excretion mechanisms. No toxicity attributable to bumetanide was observed. The large interpatient and intergroup variability found for pharmacokinetic parameter estimates may, at least in part, reflect different disease states and the ontogeny of hepatic and renal excretory mechanisms in this patient population.

We thank Dr. Leslie T. Webster, Jr., Dr. Lia Lowrie, and Dr. Michael D. Reed for their contributions during the performance of this study and the preparation of this manuscript. We also thank the research nurses for assist- ing in patient enrollment, data collection, and collection and storage of serum and urine specimens; and Anita Pettigrew for performing HPLC bumetanide analyses.

References 1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

Baliga R, Lewy JE. Pathogenesis and treatment of edema. Pediatr Clin North Am 1987;34:639-48. Witte MK, Stork JE, Blumer JL. Diuretic therapeutics in the pediatric patient. Am J Cardiol 1986;57:44A- 53A. Wells TG. The pharmacology and therapeutics of diuretics in the pediatric patient. Pediatr Clin North Am 1990;37:463-503. Tuzel IH. Comparison of adverse reactions to bumet- anide and furosemide. J Clin Pharmacol 1981;21: 615-9. Ramsay LE, McInnes GT, Hettiarachchi J, Shelton J, Scott P. Bumetanide and furosemide: a comparison of dose-response curves in healthy men. Br J Clin Phar- macol 1978;5:243-7. Asbury MJ, Gatenby PBB, O’Sullivan S, Bourke E. Bumetanide: potent new “loop” diuretic. Br Med J 1972;1:211-3. Aranda JV, Turmen T, Sasyniuk BI. Pharmacokinet- its of diuretics and methylxanthines in the neonate. Eur J Clin Pharmacol 1980;18:55-63. Turmen T, Thorn P, Louridas, AT, LeMorvan P, Aranda JV. Protein binding and bilirubin displacing properties of bumetanide and furosemide. J Clin Pharmacol 1982;22:551-6. Robertson A, Karp W. Albumin binding of bumet- anide. Dev Pharmacol Ther 1986;9:241-8. Bourke E. Frusemide, bumetanide, and ototoxicity. Lancet 1976;1:917-8. Brown RD, Manno JE, Daigneault EA, Manno BR. Comparative acute ototoxicity of intravenous bumet- anide and firrosemide in the pure-bred beagle. Toxi- co1 Appl Pharmacol 1979;48:157-69.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

CLINICAL P HARMACOLOGY &THERAPEUTICS OCTOBER 1996

Dunn FG, Kerr IC, McQueen MJ, Thomson RM. Comparison of intravenous bumetanide and frusemide during open heart surgery. Postgrad Med 1975;51(suppl 6):72-6. Wells TG, Fasules JW, Taylor BJ, Kearns GL. Phar- macokinetics and pharmacodynamics of bumetanide in neonates treated with extracorporeal membrane oxygenation. J Pediatr 1992;121:974-80. Shankaran S, Ilaqan N, Liang KC, Bhavsar V, Kauff- man R. Bumetanide pharmacokinetics in preterm ne- onates [abstract]. Pediatr Res 1989;25:72A. Lopez AM, Modi MW, Adams JA, Horner GP, De- Grazia FT, Goldberg RN. Pharmacokinetics of bu- metanide in the newborn infant [abstract]. Pediatr Res 1990;27:62A. Goldman L, Hashimoto B, Cook EF, Loscalzo A. Comparative reproducibility and validity of systems for assessing cardiovascular functional class: advan- tages of a new specific activity scale. Circulation 1981; 64: 1227-34. Papadopoulou ZL, Tina LU, Sandler P, Jose PA, Calcagno PL. Size and function of the kidneys. In: Johnson TR, Moore WM, Jeffries JE, editors. Chil- dren are different. Columbus: Ross Laboratories, 1978:98-103. Schwartz GJ, Brion LP, Spitzer A. The use of plasma creatinine concentration for estimating glomerular fil- tration rate in infants, children and adolescents. Pe- diatr Clin North Am 1987;34:571-90. Gibaldi M, Perrier D. Noncompartmental analysis based on statistical moment theory. In: Gibaldi M, Perrier D, editors. Pharmacokinetics. New York: Marcel Dekker, 1982:409-18. Borenstein M, Cohen J. Statistical power analysis: a computer program. Hillsdale, NJ: Lawence Erlbaum Associates Inc, 1988. Lau HS, Hyneck ML, Berardi RR, Swartz RD, Smith DE. Kinetics, dynamics, and bioavailability of bumet- anide in healthy subjects and patients with chronic renal failure. Clin Pharmacol Ther 1986;39:635-45. Halladay SC, Sipes IG, Carter DE. Diuretic effect and metabolism of bumetanide in man. Clin Pharmacol Ther 1977;22:179-87. Holazo AA, Colburn WA, Gustafson JH, Young RL, Parsonnet M. Pharmacokinetics of bumetanide fol- lowing intravenous, intramuscular and oral adminis- trations to normal subjects. J Pharm Sci 1984;73:1108- 13. Cook JA, Smith DE, Cornish LA, Tankanow RM, Nicklas JM, Hyneck ML. Kinetics, dynamics and bio- availability of bumetanide in healthy subjects and pa- tients with congestive heart failure. Clin Pharmacol Ther 1988;44:487-500. Benet LZ, Williams RL. Design and optimization of dosage regimens: pharmacokinetic data. In: Gilman AG, Rall TW, Nies AS, Taylor P, editors. The phar-

Page 9: Pharmacokinetics of bumetanide in critically ill infants*

CLINICAL P HARMACOLOGY &THERAPEUTICS VOLUME 60, NUMBER4

macologic basis of therapeutics. New York: Pergamon Press, 1990:1650-735.

26. Walker PC, Berry NS, Edwards DJ. Protein binding characteristics of bumetanide. Dev Pharmacol Ther 1989;12:13-8.

27. Blumer JL, Reed MD. Principles of neonatal phar- macology. In: Yaffe SJ, Aranda JV, editors. Pediatric pharmacology. Philadelphia: WB Saunders, 1992:164- 77.

28. Vert P, Broquaire M, Legagneur M, Morselli PL. Pharmacokinetics of furosemide in neonates. Eur J Clin Pharmacol 1982;22:39-45.

Sullivan et al. 413

29. Peterson RG, Simmons MA, Rumack BH, Levine RL, Brooks JG. Pharmacology of furosemide in the premature newborn infant. J Pediatr 1980;97:139-43.

30. Aranda JV, Perez J, Sitar DS, Collinge J, Portuguez- Malavasi A, Duffy B, et al. Pharmacokinetic disposi- tion and protein binding of furosemide in newborn infants. J Pediatr 1978;93:507-11.

31. Tuck S, Morselli P, Broquaire M, Vert P. Plasma and urinary kinetics of furosemide in newborn infants. J Pediatr 1983;103:481-5.

32. Ward OC, Lam LKT. Bumetanide in heart failure in infancy. Arch Dis Child 1977;52:877-82.