throbber
Page 1
`
`rnRRATSRRASRRRaa
`
`
`
`1985
`W. B. SAUNDERS COMPANY
`Philadelphia London Toronto Mexico City RiodeJaneiro Sydney Tokyo HongKong
`
`ARGENTUM EX1021
`ARGENTUM EX1021
`
`Sol S. Zimmerman, M.D.
`Associate Professor of Clinical Pediatrics
`NewYork University School of Medicine
`Assistant Director of Pediatrics
`Director, Pediatric Intensive Care Unit
`University Hospital
`New York University Medical Center
`New York, New York
`
`ASSOCIATE EDITOR
`
`Joan Holter Gildea, R.N., B.S., M.A.
`Clinical Assistant Director of Nursing
`New York University Medical Center
`NewYork, New York
`
`
`
`EEELLY,CENRESCTaSSEF5
`0ee,
`
`CRITICAL CARE
`PEDIATRICS
`
`Page 1
`
`

`

`
`
`N®DoOa
`
`Page 2
`
`

`

`
`
`CLINICAL PHARMACOLOGYIN THE CRITICALLY ILL CHILD / 91
`
`R x ty,
`————
`C., =
`0.7 X Va x Wt.
`
`(
`
`14.4
`
`)
`
`where R is expressed in mg/hr and C,, is ex-
`pressed in mg/L.
`Equation 14.4 indicates that equilibrium drug
`concentration is related to three variables: half-
`life (t,,), volume of distribution (V,), and rate
`of administration (R, or D/T). Thus, doubling
`Vy has the same effect upon steady state con-
`centration as halving t,,. Either alteration will
`lead to a 50 percent reduction in drug concen-
`tration that can be exactly offset by doubling
`the dosage.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
` Egeaanepeean-
`
`
`
`
`
`ee
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`concentration; in 3.3 half-lives, it will be 90
`percent; and in five half-lives,
`it will be 97
`percent of the plateau level.
`Dosage recommendations for continuousin-
`fusions are designed to produce appropriate
`plasma concentrations at equilibrium. The phe-
`nomenonjust described, which is often termed
`drug accumulation, entails a delay in achieving this
`concentration. The magnitude of the delay is
`related to the half-life of the drug, whereas the
`ultimate concentration is determined by the Vj,
`the half-life, and the rate of administration.
`
`Plasma Drug Clearance
`
`The plasma clearance (Cl) of a drug is of
`primary importance in appreciating the rela-
`tionship between rate of drug administration
`and consequent drug concentration. Drug
`clearance, like creatinine or inulin clearances,
`is determinedbyrelating the rate of elimination
`(E) to the plasma concentration at equilibrium
`(C..):
`
`Cl = |=Ss
`
`At equilibrium, E = R. Thus,
`
`R
`Ccl= =

`C.
`
`or, with rearrangement,
`
`R
`==
`Cl
`
`C.
`
`3
`(14 )
`
`R=axc,
`
`°-
`
`(14.3A)
`
`where R= rate of administration.
`Equation 14.3A emphasizesthatit is drug clear-
`ance, rather than half-life, which determines
`the rate of administration (R) or dosage per
`interval (D/T) necessary to achieve a specific
`concentration. Ultimately, clearance is related
`to half-life and volume of distribution:
`
`cy 0.7 X Va XWe
`ty,
`
`44oO
`1nwo
`A2
`
`when Vj is expressed in L/kg, half-life (t,,) in
`hours, and clearance (Cl) in L/hr.
`With substitution into equation 14.3, this be-
`comes:
`
`Multiple Dose Kinetics
`
`The reader has been introduced to the phe-
`nomenon of drug accumulation as it occurs
`during continuous infusion. Drug accumula-
`tion also occurs with intermittent dosage
`schedules.
`Considera drug that is given by intermittent
`IVinjection. Whenthefirst dose is given, the
`concentration of drug is zero. Immediately after
`the dose, a peak concentration is recorded. The.
`concentration then declines at a rate deter-
`mined by the drug’s half-life. If the next dose
`is given before the concentration has once again
`reached zero, the second peak will be higher
`than the first. As this process continues, the
`peak(C,,,.) and trough (C,,,,) levels rise toward
`plateau values, as will the average concentra-
`tion, C,,-. This process is illustrated in Figure
`14-4. Drug accumulation occurs during inter-
`mittent administration when a second, or nth,
`dose is administered before all the previous
`dose has been eliminated. For mostclinical pur-
`poses, this condition is satisfied when the dos-
`ing interval is less than twice the half-life of
`the drug. As with continuousinfusions, 50 per-
`cent of a plateau concentration is achieved in
`one half-life; 97 percent is achieved after five
`half-lives.
`C.ve is analogous to the equilibrium concen-
`tration (C,,) that develops during continuous
`infusion. Thus, its value is determined only by
`the relationship between clearance (C1) and rate
`of administration (R; see Equation 14.3). The
`peak (C,,..) and trough (C,,,) concentrations
`fluctuate around the C,,, in a manner that is
`determined by the size of the dose and the
`length of the dosing interval. For example,
`theophylline may be administered by inter-
`mittent IV injection. In an adult, a standard
`regimen calls for 300 mg every 6 hours (1200
`
`
`
`Page 3
`
`

`

`
`
`
`
`
`92 / GENERAL PROBLEMS AND TECHNIQUES OF CRITICAL CARE
`
`
`
`20PPT?Trrirrrt
`
`
`14
`6
`8
`2
`
`
`Jos
`pot tt
`Jottd
`10 12 14 16 18 20 22 24 26 28 30. Hours
`3
`4
`5
`6
`7 Number of
`Half-lives
`
`2
`
`4
`1
`
`
`
`TheophyllineConcentration
`
`(mg/L)
`
`Concentration of theophylline during intermittent IV administration. Note that C,.a Cmins aNd Cz,. increase
`Figure 14-4.
`to equilibrium values. C,,,. = C,, that obtains during continuous administrationif the total daily dosagesare identical.
`
`
`
`
`
`
`
`
` mg/day). Alternatively, one may administer
`150 mg every 3 hours (1200 mg/day). Finally,
`some physicians administer a continuous in-
`
`fusion of 50 mg/hr (1200 mg/day). Both in-
`
`termittent regimens produce the same(C,,.),
`
`whichis equal to the C,, during the continuous
`
`To this point, the discussion has concerned
`
`infusion. This does not mean that the regimens
`
`first-order kinetic behavior in whichafixed pro-
`are equivalent. The 6-hour schedule produces
`
`portion of drug is eliminated per unit time. Zero-
`muchgreater fluctuation around C,,. than the
`
`order kinetics occurs under some conditions, no-
`3-hour schedule. With the 3-hour regimen, the
`
`tably when plasma drug concentrationsarerel-
`peaks and troughs lie closer to Cyye. This in-
`
`creases
`the
`likelihood of remaining within the therapeutic
`range throughout the dosing interval (Fig. 14—
`5).
`,
`In this regard, the half-life of a drug is a
`watershed. When a drugis given at an interval
`that is equalto its half-life (T = ty), Crnax/Crin
`is approximately 2. During more frequent
`administration (T < ty), Cmax/Cmin is less than
`2, and during less frequent administration
`(T > ty), Crax/Cmin is greater than 2.
`Thus, drugs with a long half-life, such as
`digoxin or phenobarbital, are often given once
`daily, because even with this schedule, T is less
`than ty, and the plasma concentration remains
`within the relatively narrow therapeutic range
`of these agents. Conversely, theophylline and
`quinidine have relatively short half-lives (3 to
`6 hours in children). When conventional for-
`mulations of these agents are administered,
`they requirerelatively frequent dosing (every
`3 to 6 hours) if the plasma concentrationis to
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`remain within the respective therapeutic ranges
`throughout the dosing interval.
`
`Nonlinear Kinetics
`
`
`
`
`
`Concentration(arbitraryunits)
`
`Dosage = D
`Interval = T = ty,
`
`
`
`Dosage = D/2
`interval = TI2 = ty,/2
`
`Time
`
`Effect of varying both dosage and dose
`Figure 14-5.
`interval upon peak(C,,,,) and trough (C,,,,) concentrations
`during steady state. The solid saw toothline indicates the
`time concentration curve that results with intermittent IV
`administration of dosage D at an interval T equal to the
`drug tv. Note that Cnad/Cmin = 2. The interrupted fine
`indicates the curve that results when dosage (D/2) and
`interval (T/2) are halved. Cya/Cmin = 1-5. The straight
`solid line indicates C.,,, which is the.same during both
`conditions, and is equal to C,,, which results when the
`sametotal daily dosage is administered by continuous IV
`infusion.
`
`age 4
`
`i P
`
`Page 4
`
`

`

`
`
`CLINICAL PHARMACOLOGY!N THE CRITICALLY ILL CHILD / 93
`
`MAINTENANCE DOSE
`
`The maintenance dose (MD) is the amount
`of drug (R for continuous infusion, D/Tfor an
`intermittent schedule) that is administered dur-
`ing equilibrium. Thus, from Equation 14.3A,
`maintenance dose, MD,is equal to the product
`of clearance, Cl, and desired steady state plasma
`concentration, C,,,
`(MD = Cl x C,). The
`maintenance dose is often determined by con-
`sulting standard reference material. In patients
`
`approaches V,,,,, and zero-order behavior oc-
`atively large. With zero order or nonlinear ki-
`curs.
`netics, a fixed amount of drug is eliminated per
`There are two important consequences of
`unit time. Ethanol is an extreme example, be-
`this kinetic behavior. The first is that an in-
`cause the usual dosage is large (gm amounts)
`crease in dosage produces an exponential, rather
`relative to other drugs (mg amounts). Within
`than a linear rise in concentration. This occurs
`the usual range of blood ethanol concentra-
`very often whentreating patients with phen-
`tions, humanseliminate about 120 mg per kg
`ytoin (Fig. 14-6); on occasion,this phenomenon
`per hrof the substance. Because the volume of
`is recognized during treatment with theophyl-
`distribution of ethanol is about 0.5 L per kg,
`line. It requires that dosage adjustments must
`blood ethanol levels decline at a fixed rate of
`be made cautiously and in small amounts. The
`20 to 25 mg/dl per hr. Therate of elimination
`second major consequence of Michaelis-Men-
`does not change with increases in concentra-
`ton kinetics is that the apparent plasmahalf-life
`tion. Consequently, increments in dosage pro-
`increases with the plasma concentration. The
`duce much greater changes in concentration
`greater the plasma concentration, the sloweris
`than would be the case for a drug eliminated
`the relative rate of elimination. Using repre-
`in accordance with first-order kinetics.
`sentative values of K,, and V,,.. for phenytoin,
`Manysubstances followafirst-order model
`one can estimate that at a concentration of 10
`at low plasma concentrations but a zero-order
`mg per L, the apparent t,, of phenytoin is 24
`modelat higher concentrations. Whenthetran-
`hours; at a concentration of 25 mg perL, the
`sition from first- to zero-order elimination oc-
`apparent t,,
`is 42 hours. This means: 1) in-
`curs at concentrations appreciably higher than
`creases in dosage cause lengthening of the ap-
`the usual therapeutic range, the pharmacoki-
`parent t,, (thus, Michaelis-Menton kinetics is
`netic treatment of the drug is uncomplicated
`sometimesreferred to as dose dependent kinetics),
`and a first-order kinetic model will be suffi-
`2) small increments in dosage can produce huge
`ciently accurate for mostclinical purposes.
`increases in drug concentration, and 3) intox-
`Unfortunately, a few commonly used drugs,
`ication with phenytoin will be prolonged, be-
`such as phenytoin and salicylate, exhibit this
`cause, at high concentrations, eliminationis ex-
`transition at concentrations within the thera-
`tremely slow relative to the amountof drugin
`peutic range.
`the body.
`A change from first- to zero-order kinetics
`as concentration increases is typical of an en-
`zyme-mediated process. This changeis due to
`saturation of the enzyme system that is re-
`sponsible for metabolic transformation of the
`drug. There is a limited amount of enzymeat
`the metabolic site; therefore, there is a maxi-
`mum rate at which transformation can occur
`(Vinax)» At concentrations that are low relative
`to Vn, first-order behavior predomiinates. As
`concentration increases, Vinax is approached.
`After V,,, has been achieved, further increases
`in concentration cannot augment the metabolic
`rate. Thus, a fixed amount of drug is metab-
`olized per unit time. This amount, of course,is
`equal to Via, Mathematically, this process is
`described by the Michaelis-Menton equation:
`
`14.5
`
`— Vanax X €
`(14.5)
`K, +C€
`E=
`whereE is the rate of elimination or metabo-
`lism; V,,, is the maximum rate of metabolism;
`K,, is the Michaelis-Menton constant, which
`defines the affinity of the enzyme for the drug;
`and C is plasma drug concentration.
`Note that when C is much less than K,,, E
`varies directly with C. This resemblesa first-
`order process. When C is greater than K,,, E
`
`
`
`Phenytoin
`(Michaelis-Menton)
`
` Gentamicin
`(First-order)
`
`|
`|
`
`Concentration
`
`
`
`Daily Dosage
`Effect of dosage upon plasma concentra-
`Figure 14-6.
`tion for drugs following first-order vs. Michaelis-Menton
`kinetics.
`
`Page 5
`
`Page 5
`
`

`

` 94 / GENERAL PROBLEMS AND TECHNIQUES OF CRITICAL CARE
`
`with abnormal drug disposition, better indi-
`vidualization of therapy is achieved if one ap-
`preciates the relationship between changes in
`Cl and consequent changes in C,,. Knowledge
`of a patient’s Cl can be used to calculate dosage
`requirements. This procedure is described in
`the case study at the conclusion of this chapter.
`It is not always appropriate to inifiale therapy
`with the maintenance dose. Both continuous
`and intermittent schedules produce a gradual
`rise from theinitial (usually zero) to the equi-
`librium concentration. Recall that 75 percent of
`the plateau is reached in two half-lives, and 97
`percent is reached in five half-lives. Thus, for
`drugs that have a long half-life, there will be
`a substantial delay in acquisition of the plateau
`concentration. Because the plateau concentra-
`tion may be close to the minimum effective
`concentration, this delay may be unacceptable
`in acutely ill patients. For example, an asth-
`matic who is simply placed on a theophylline
`infusion will not begin to experience relief for
`about8 hours.
`
`LOADING DOSE
`
`The solution to the problem of delay in
`achieving adequate levels is to administer a
`loading dose (LD). The loading dose is the
`amount of drug that will rapidly produce a
`therapeutic plasma concentration.
`If one is emphasizing a target concentration
`strategy, calculating the loading dose is simple,
`because the loading dose and the desired con-
`centration (C,,) are related through the volume
`of distribution (see Equation 14.2). With ap-
`propriate modifications,
`this expression be-
`comes:
`
`LD = V,(L/kg) x Wt(kg) x C..(mg/L)
`
`(14.6)
`is the desired equilibrium concen-
`where C,,
`tration. Thus, for a child weighing 10 kg in
`whom one wishes to achieve a plasma theo-
`phylline (Vz = 0.5 L/kg) concentration of 12
`mg per L, the correct loading dose is 60 mg.
`This amount of drug should be administered
`slowly, over about a 15-minute period. Im-
`mediately thereafter, the appropriate mainte-
`nance dose is initiated. The effect of a loading
`dose is shown in Figure 14-7.
`If one is using an empirically derived main-
`tenance dose and is not attempting to achieve
`a specific drug concentration (target effect
`strategy), the problem is less straightforward.
`In such cases, it is probably best to consult
`
`
`
`PlasmaConcentration
`
`ty
`
`4 ty,
`
`Time
`
`Figure 14-7.
`Administration of an appropriate loading
`doseeliminates the delay (4t,,) in achieving equilibrium
`concentration, C,,. Solid line-infusion alone, beginning at
`T = 0. Interrupted line—loading dose at T = 0, followed
`by continuous infusion.
`
`individual product information when designing
`the loading dose. Readers interested in a the-
`oretical approach to this issue should consult
`the suggested reading by Rowland and Tozer.
`When intravenous therapy is indicated, the
`loading dose is often given as a single rela-
`tively brief infusion. In the case of a drug with
`a narrow therapeutic range or a prolonged
`phaseof distribution, the physician may choose
`to divide the loading dose, as is commonly done
`with digoxin. In general, a loading dose is not
`indicated whenthe half-life is much less than
`the dosing interval (ie., drug accumulation
`does not occur) or when the therapeutic range
`is wide. Thus, penicillin therapy does not begin
`with a loading dose. Of course, a loading dose
`is not indicated when there is no urgency in
`achieving the equilibrium drug concentration.
`There is also no point in administering a load-
`ing dose when the half-life of a drug is very
`short, as with most pressor agents, because
`equilibrium conditions are reached in a matter
`of minutes during continuous maintenancein-
`fusion.
`The foregoing kinetic description applies to
`intravenous administration. Following intra-
`muscular or oral administration one must ex-
`tend the analysis by taking into account the
`rate and extent of absorption. Drugs that are
`completely and efficiently absorbed after in-
`tramuscular injection should maintain a similar
`C,yer although peak and trough levels may lie
`closer to C,,, (lower peak, higher trough). After
`oral administration, many drugseither are not
`completely absorbed from the gastrointestinal
`tract or once absorbedare efficiently extracted
`and then biotransformed on the first pass
`through the liver. This process effectively re-
`duces the dosage of drug that reaches the sys-
`temic circulation. Equations 14.2 through 14.4
`are modified by multiplying the dosage ad-
`
`Page 6
`
`
`
`Page 6
`
`

`

`Page 7
`
`
`
`CLINICAL PHARMACOLOGY IN THE CRITICALLY ILL CHILD / 95
`
`ministered (R, or D/T) byf,,.1, the fraction of
`orally administered drug that reaches the sys-
`temiccirculation. Severalof the suggested read-
`ings contain a table of representative f,,.4, values.
`
`KINETIC VARIATION—CLINICAL
`APPLICATION
`
`In the foregoing discussion, three variables
`that affect the plasma drug concentration have
`been identified: 1) the rate of administration
`(R, or D/T), 2) the volumeofdistribution (V4),
`and 3) the half-life (t,,). These three variables
`are related to one another through Equation
`14.4, Of these, only the rate of administration
`is under the physician’s control. Thus, the art
`of pharmacokinetics consists of altering this
`parameterin order to compensatefor individual
`differences in elimination or distribution.
`
`Altered Distribution
`
`Critical illness and the age of the patient af-
`fect the distribution of many drugs. Several
`mechanisms may be
`involved,
`including
`changes in the contentordistribution of body
`water, alterations in plasma protein binding,
`perturbations in regional blood flow, and dif-
`ferences in body fat content.
`
`AGE-RELATED CHANGES
`
`The water content of the body changes dra-
`matically with age. At 28 weeks gestation, the
`water content of the body is 85 percent. This
`figure decreases to 70 percent at term and to
`60 percent in adults. There is a concurrent in-
`crease in the amountof body fat from 1 percent
`of body weight at 28 weeks to 15 percent at
`term, as well as altered binding to protein. Dis-
`ease (tachypnea, dehydration),
`the environ-
`ment in which the infant is nursed, and the
`volume and composition of administered fluids
`produce fluctuations in body water. Thus,it is
`anticipated that drugs that are distributed
`mainly in the body water have a different, usu-
`ally greater, volumeof distribution (V4) in in-
`fants and young children than in adults. Table
`14-2 lists several drugs for which the V4q is
`knownto differ between newborn infants and
`adults. This information provides the rationale
`for many empirically determined dosage mod-
`ifications. A larger Vy does not necessarily in-
`volve a larger dosage. The actual determinant
`of the maintenance dosage requirementis clear-
`
`Effect on Vy
`
`Table 14-2, Selected Drugs With Altered Volume
`of Distribution (V,) in Neonates and Children
` Drug
`IN
`Diazepam
`tN,1C
`Digoxin
`1N,
`Furosemide
`tN tC
`Gentamicin
`tN
`Lidocaine
`1N
`Phenobarbital
`+N
`Phenytoin
`
`Theophylline
`tN
`t = V, larger; | = Va smaller; N = neonate; and C =
`children older than 1 month. The effect of changes in Vy
`may be modified by alterations in protein binding and
`elimination rate.
`
`ance. Clearance (Cl) is related to both Vy and
`half-life. In infants, drug half-life may be pro-
`longed (vide infra), and this may offset the
`increase in V, (recall that Cl = 0.7 X Vu/ty).
`The net effect is frequently a reduced dosage
`requirementor an increased loading dose, fol-
`lowed by a reduced maintenance dose.
`
`DISEASE-RELATED CHANGES
`Several processes affect V, by altering body
`water content, body fat content, or the degree
`of protein binding. Notable examples are ure-
`mia, chronic liver disease, and congestive heart
`failure.
`In uremia, the water content of the body is
`frequently greater than normal. This factor, to-
`gether with disturbed protein binding, causes
`the volumeofdistribution (V,) of several drugs
`to increase (e.g., gentamicin) or decrease(e.g.,
`digoxin). As in newborns, the larger V,is fre-
`quently accompanied by prolongation of drug
`half-life.
`Chronic liver disease is associated with de-
`creasedlevels of plasma proteins and with fluid
`accumulation. Thus,it is not surprising that the
`V, of several drugs increases in the presence of
`cirrhosis. Other conditions associated with ex-
`tracellular fluid expansionalso increasethedis-
`tribution of certain drugs; for example, ami-
`noglycoside antibiotics distribute into ascitic
`fluid. Thus, in the presence of ascites, the V,
`of these agents may be substantially increased.
`In cystic fibrosis, the V, of several of the ami-
`noglycosides seems to be higher than average.
`Unless daily dosageis increased, this may lead
`to subtherapeutic drug concentrations.
`Abnormalities of regional or global blood
`flow may reduce distribution by limiting per-
`fusion to sites of uptake. In patients with low-
`output states (congestive heart failure, CHF;
`shock), the rafe of distribution is likely to be
`
`_
`
`Page 7
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket