throbber
The Effect of Chronic Renal Failure on Hepatic Drug
`Metabolism and Drug Disposition
`
`Albert W. Dreisbach*,† and Juan J. L. Lertora†
`
`*Division of Nephrology, Department of Medicine, and †Division of Clinical Pharmacology, Department
`of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
`
`ABSTRACT
`
`There is abundant evidence that chronic renal failure (CRF)
`and end-stage renal disease (ESRD) alter drug disposition by
`affecting protein and tissue binding and reducing systemic
`clearance of renally cleared drugs. What is not fully appreciated
`is that CRF can significantly reduce nonrenal clearance and
`alter the bioavailability of drugs predominantly metabolized by
`the liver. Animal studies in CRF have shown a major down-
`regulation (40–85%) of hepatic cytochrome P-450 metabolism
`involving specific isozymes. Phase II reactions such as acety-
`lation and glucuronidation are also involved, with some
`isozymes showing induction and others inhibition. Hepatic
`
`enzymes exhibiting genetic polymorphisms such as N-acetyl-
`transferase-2 (NAT-2), which is responsible for the rapid and
`slow acetylator phenotypes, have been shown to be inhibited by
`ESRD and reversed by transplantation. There is some evidence
`pointing to the possibility of inhibitory factors circulating in the
`serum in ESRD patients which may be dialyzable. This review
`includes all significant animal and clinical studies using the
`search terms ‘‘chronic renal failure,’’ ‘‘cytochrome P-450,’’ and
`‘‘liver metabolism’’ over the past 10 years obtained from the
`National Library of Medicine MEDLINE database, including
`relevant articles back to 1969.
`
`There is a tacit assumption that the clearance and
`distribution of drugs that are removed from plasma
`exclusively by hepatic metabolism are not affected by
`chronic renal failure (CRF). Therefore no dose adjust-
`ments are necessary in hepatically cleared drugs in this
`population. This is a widespread misconception, primar-
`ily due to a lack of information, which is not supported
`by investigations spanning several decades. The purpose
`of this review is to outline the different ways in which
`drug disposition is altered by CRF, cite specific drugs of
`clinical concern, and discuss proposed mechanisms.
`Citations were derived from MEDLINE (1990–2001)
`using the search terms ‘‘liver metabolism,’’ ‘‘chronic
`renal failure,’’ and ‘‘cytochrome P-450’’ including other
`relevant articles dating back to 1969.
`
`Background Pharmacokinetics
`
`In order to understand the various effects of CRF on
`drug disposition it may be helpful to review some basic
`pharmacokinetic
`concepts
`(1). Systemic
`clearance
`(CLSYS) is equal to the sum of renal (CLR) and nonrenal
`(CLNR) clearances:
`CLSYS ¼ CLR þ CLNR
`
`ð1Þ
`
`Address correspondence to: Albert W. Dreisbach, MD,
`Division of Nephrology, SL45, Department of Medicine,
`Tulane University School of Medicine, 1430 Tulane Ave.,
`New Orleans, LA 70112, or email: adreisb@tulane.edu.
`Seminars in Dialysis—Vol 16, No 1 (January–February) 2003
`pp. 45–50
`
`45
`
`Renal clearance is defined as the amount of drug
`eliminated per unit time in the final urine normalized to
`plasma concentration:
`ð2Þ
`CLR ¼ UV=P;
`where U is the urine concentration, P is the plasma
`concentration, and V is the urine flow rate.
`Renal drug metabolism according to this definition is
`not a component of CLR, but rather is incorporated into
`the nonrenal clearance term CLNR. The renal cyto-
`chrome P-450 activity is 20% of the hepatic cytochrome
`P-450 activity per gram of tissue and the kidney
`participates in conjugation reactions as well
`(2,3).
`However, the total renal mass is one-fifth of the hepatic
`mass and therefore renal cytochrome P-450 may contri-
`butes a minor fraction of the total activity. Nevertheless,
`for drugs which are highly extracted by the kidney and
`whose metabolism is therefore blood flow limited, the
`kidney may play a more significant role, since renal blood
`flow is three- to fivefold greater than hepatic blood flow
`per gram of tissue (4). Of course, this renal drug
`metabolizing capacity deteriorates with the progression
`of CRF, leading to a reduction in nonrenal clearance
`independent of any effects on the liver or intestinal
`metabolism.
`Nonrenal clearance and the distribution volume of
`drugs is altered by CRF via changes in hepatic clearance,
`plasma protein binding, and tissue binding. CRF
`has been shown to decrease plasma protein binding of
`acidic drugs, increasing the free (unbound) fraction (fu),
`as shown in Table 1 (5–18). The mechanism is presum-
`ably the reduction of the apparent binding affinity to
`albumin due to conformational changes in albumin or
`
`Boehringer Ex. 2018
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 1
`
`

`
`46
`
`Dreisbach and Lertora
`
`competition with unmeasured small molecules and
`accumulated metabolites that are not entirely removed
`by dialysis (19,20).
`CRF increases plasma levels of a1-acid glycoprotein
`which is induced in acute and chronic inflammation. The
`a1-acid glycoprotein binds basic drugs and one would
`predict increased binding of these drugs in CRF.
`However, in most cases drug binding is reduced or
`remains unchanged and is only occasionally increased, as
`shown in Table 1 (6). The reduced binding of basic drugs
`to a1-acid glycoprotein may result from mechanisms
`similar to those that impair the binding of acidic drugs to
`albumin, such as reduced binding affinity and competing
`ligands circulating in the plasma of end-stage renal
`disease (ESRD) patients.
`CRF has also been shown to decrease the distribution
`volume (Vd) of drugs such as digoxin, which has its Vd
`reduced by 50% in ESRD due to reduced tissue binding
`(2). The opposite effect is observed with phenytoin, which
`shows a twofold increase in Vd in ESRD due to reduced
`plasma protein binding (2). Furthermore, CRF reduces
`hepatic clearance of drugs by mechanisms which will be
`discussed later.
`The determinants of hepatic clearance will be better
`understood after a further discussion of clearance
`concepts (1,21). The rate of extraction of a drug across
`an organ of elimination is described by the blood flow
`through the organ (Q) and the concentrations of the drug
`at the arterial side (input) (CA) and at the venous side
`(output) (CV) as shown in the equation 3. Extraction
`ratio (E) is the rate of extraction divided by the rate of
`presentation (equation 4). Clearance (CL) is the rate of
`extraction normalized to CA (equation 5). Hepatic
`clearance (CLH) is equal to hepatic blood flow multiplied
`by EH (equation 6).
`Rate of extraction ¼ QðCA CVÞ
`E ¼ rate of extraction=rate of presentation
`¼ CA CV=CA
`CL ¼ rate of extraction=CA
`¼ QðCA CVÞ=CA
`
`ð3Þ
`
`ð4Þ
`
`ð5Þ
`
`TABLE 1. Drugs with changes in protein binding in CRF
`
`Drug type
`
`Change in binding
`
`Acidic
`Desmethyldiazepam
`Phenytoin
`Valproic acid
`Salicylate
`Phenylbutazone
`Furosemide
`Warfarin
`Sulfamethoxazole
`
`Basic
`Disopyramide
`Vancomycin
`Morphine
`Oxazepam
`
`Adapted from Refs. 5 and 6.
`
`Decreased
`Decreased
`Decreased
`Decreased
`Decreased
`Decreased
`Decreased
`Decreased
`
`Increased
`Decreased
`Decreased
`Decreased
`
`ð6Þ
`CLH ¼ QEH
`If a drug has a high hepatic extraction ratio, E
`approaches one, then the total hepatic clearance (CLH) is
`rate limited by hepatic blood flow. With a low-extraction
`drug, E approaches zero, then total hepatic clearance is
`rate limited by intrinsic (unbound) clearance (CLINT)
`and free fraction (fu) of drug. The relationship between
`these parameters is expressed in the well-stirred model of
`total hepatic clearance (CLHTOT) (equation 7) (21):
`ð7Þ
`CLHTOT ¼ QCLINTfu=Q þ CLINTfu:
`Intrinsic hepatic clearance relates the rate of metabo-
`lism at steady state to the free unbound fraction in blood.
`If the drug is highly extracted, we assume CLINTfu>>Q,
`then CLHTOT ¼ Q and total hepatic clearance is blood
`flow limited and relatively independent of changes in
`intrinsic hepatic clearance which may be produced by a
`down-regulation or induction of hepatic cytochrome
`P-450. If the drug is a low-extraction agent then we
`assume Q>>CLINTfu, then CLHTOT ¼ CLINTfu and
`would be highly influenced by changes in plasma protein
`binding and intrinsic hepatic clearance. These various
`permutations have been aptly simulated using empiric-
`ally derived models (22). Hepatic blood plasma flow has
`been shown to be unaffected by CRF (23). However,
`concomitant diseases such as congestive heart failure and
`chronic liver disease are frequently associated with ESRD
`and are known to reduce hepatic blood flow and CLHTOT.
`For a highly extracted drug, orally administered, with
`a large first-pass effect (presystemic clearance) and low
`systemic bioavailability (F), the magnitude of the first-
`pass effect can be estimated by equation 8 (1,21):
`F ¼ QH=QH þ CLINTfu:
`ð8Þ
`For highly extracted drugs, CLINTfu>>QH, then
`F ¼ QH/CLINTfu. Therefore changes in intrinsic hepatic
`clearance and plasma protein binding induced by CRF
`would be predicted to produce significant changes in
`systemic bioavailability. For example, a reduction in
`intrinsic hepatic clearance would reduce first-pass meta-
`bolism, causing a sizable increase in bioavailability of a
`highly extracted drug, leading to a clinically significant
`increase in steady-state plasma concentration.
`In summary, CRF can significantly affect the dispo-
`sition of both low and high hepatic extraction drugs
`which are cleared predominantly by the liver (Table 2).
`Hepatic clearance of low-extraction drugs is limited by
`intrinsic hepatic clearance and plasma protein binding.
`A reduction in intrinsic hepatic clearance will lead to
`reduced systemic clearance and produce an increase in
`steady-state plasma levels for both free and total drug
`concentrations of low-extraction drugs. Reduced plasma
`protein binding leading to increased free fraction (fu)
`will also produce an elevated total hepatic clearance
`that leads to a reduction in total steady-state drug
`concentrations but no change in free concentrations as
`long as intrinsic hepatic clearance is unchanged. Since
`free concentrations are presumably the biologically
`active fraction, then the result may have little clinical
`impact.
`
`Boehringer Ex. 2018
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 2
`
`

`
`CRF AND DRUG METABOLISM
`
`47
`
`TABLE 2. Summary of the effects of CRF on pharmacokinetic parameters
`
`Low hepatic extraction drugs (EH < 0.20)
`Cytochrome P-450 down-regulation, circulating inhibitors fi CLINTfl fi CLTOTHEPflCT›Cu›
`(Acidic drugs) fi PPBfl fi CLTOTHEP›CTflCu«
`High hepatic extraction drugs (EH > 0.80)
`Cytochrome P-450 down-regulation, circulating inhibitors fi CLINTfl fi F›
`(Acidic drugs) fi PPBfl fi Ffl
`
`Hepatic extraction, EH; total hepatic clearance, CLTOTHEP; total steady-state plasma concentration, CT; free steady-state plasma
`concentration, Cu; intrinsic hepatic clearance, CLINT; plasma protein binding, PPB; bioavailability, F.
`
`For high-extraction drugs, total hepatic clearance is
`blood flow limited. Since hepatic blood flow is not altered
`in renal failure, total systemic clearance should not be
`affected by changes in intrinsic hepatic clearance or
`plasma protein binding. However, first-pass metabolism
`(presystemic clearance) is a function of CLINT and fu, so
`changes in these parameters will result in significant
`changes in systemic bioavailability, as much as several
`fold for high-extraction drugs.
`
`Animal Studies: Effect of Renal Failure
`on Cytochrome P-450 Metabolism
`
`Acute Renal Failure Models
`
`Animal studies spanning several decades have shown
`reduced hepatic microsomal enzyme activity in experi-
`mental models of acute and chronic renal failure. In rats
`with acute renal failure (ARF) (produced by subtotal
`nephrectomy), hepatic microsomal protein content (per
`gram of liver) was reduced (24). Demethylation of
`aminopyrine and p-nitroanisole, and para-hydroxyla-
`tion of acetanilide also dropped by 30–50%. There was
`stimulation of O-demethylation of p-nitroanisole by
`injections of d-aminolevulinic
`125%. Intraperitoneal
`acid normalized total microsomal protein cytochrome
`P-450 content but did not reverse the inhibition of
`cytochrome P-450 activity. In other rat models of ARF
`(induced by bilateral ureteral ligation or uranylnitrate
`injection), a reduction of aminopyrine N-demethylation
`was seen (25). Total cytochrome P-450 and aniline
`hydroxylation was reduced as well, but did not reach
`statistical significance.
`An example of how the pharmacokinetics of a highly
`extracted, extensively metabolized drug can be altered by
`experimental renal failure in the intact rat is illustrated by
`the following study using a rat model in which ARF is
`induced by uranyl nitrate (26). Following injection of
`ARF rats and controls with intravenous levoproprano-
`lol, no differences in the area under the plasma drug
`concentration versus time curve (AUC) or plasma
`clearance were seen. Using the same protocol with oral
`levopropranolol,
`the ARF rats showed a 2.5-fold
`increase in systemic bioavailability from 7 to 18%. The
`same investigators then studied levopropranolol first-
`pass metabolism in isolated perfused rat liver (27). Liver
`from normal rats perfused with normal blood yielded a
`97.4% extraction (EH) of levopropranolol. Livers from
`ARF rats perfused with uremic blood yielded a levopro-
`pranolol extraction of 90.6%. This resulted in a greater
`than threefold increase in bioavailability (F) (2.6–9.4%)
`consistent with the data from intact animals and
`
`corresponding to a 50% drop in CLINT. When livers
`from normal rats were cross-perfused with uremic blood,
`EH was reduced to 92.7%, not significantly different
`from the uremic livers perfused with uremic blood.
`Surprisingly, uremic livers cross-perfused with normal
`blood showed an EH of 97.0%, identical to the control
`normal livers perfused with normal blood. These data
`suggest that cytochrome P-450 metabolism of propran-
`olol in uremic rats is not down-regulated but that a
`rapidly acting inhibitory factor exists in uremic serum.
`
`Chronic Renal Failure Models
`
`A study utilizing a CRF rat model induced by subtotal
`nephrectomy showed a 24–32% decrease in hepatic
`N- and O-demethylation activities, whereas S-deme-
`thylase, esterase, UDP-glucuronyl
`transferase, and
`monoamine oxidase were not altered (28). Alcohol
`dehydrogenase activity increased by 71% and cyto-
`chrome P-450 levels decreased by 26%. CRF also
`increased hexobarbital sleeping time. In each case
`alterations correlated with extent of CRF.
`Over the past 15 years the cytochrome P-450 gene
`superfamily has been sequenced and various isozymes
`and isozyme-specific probe drugs have been identified. In
`a CRF model in rats using subtotal nephrectomy, the
`effect of CRF on the activity of various cytochrome
`P-450 isozymes, in vivo, was investigated using 14C-labe-
`led caffeine (CYP1A2), aminopyrine (CYP2C11), and
`erythromycin (CYP3A2) breath tests (29). There was a
`35% reduction in CYP2C11 and CYP3A2 activities and
`no difference in CYP1A2 as measured by their respective
`breath tests. Total cytochrome P-450 was reduced by
`40% and protein expression of CYP2C11, CYP3A1, and
`CYP3A2 in CRF rats was reduced by 45%, 85%, and
`45%, respectively; CYP1A2 did not change. There was
`also a significant correlation between cytochrome P-450
`activity measured by breath tests and protein expression
`of the various cytochrome P-450 isozymes.
`A later study by the same investigators also confirmed
`these findings (30). Protein expression of CYP2C6,
`CYP2D, and CYPE1 also remained constant. Northern
`blot analysis showed a down-regulation in gene expres-
`sion in CYP2C11, CYP3A1, and CYP3A2 with no
`change in the expression of the remaining cytochrome
`P-450 isozymes tested. Phenobarbital and dexametha-
`sone maintained their ability to induce CYP3A1 and
`CYP3A2 isozymes. Demethylation of erythromycin
`(CYP3A2) was reduced by CRF, which was reversed
`by treatment with phenobarbital and dexamethasone.
`In a CRF model (45 days of severe uremia in rats
`caused by subtotal nephrectomy), no induction of
`
`Boehringer Ex. 2018
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 3
`
`

`
`48
`
`Dreisbach and Lertora
`
`microsomal cytochrome P-450 enzymes occurred (31).
`However, induction of the phase II reaction, glucuron-
`idation, did occur (77% increase in activity). Total
`cytochrome P-450 content (in nmol/mg of protein) was
`reduced by 45%. P-nitroanisole activity was reduced by
`40%, aminopyrine by 35%, and acetanilide by 40%.
`Treatment with the plasticizer di(2-ethylhexyl)phthalate
`caused a 54% increase in liver wet weight and 65%
`increase in microsomal protein content, a 23% increase
`of demethylation of aminopyrine and a threefold
`reduction in hexobarbital sleeping time in CRF rats but
`not in sham-operated controls. These results and those of
`the previous study (30) suggest that known enzyme
`inducers can partially reverse the effects of CRF on
`hepatic cytochrome P-450 metabolism and that phase II
`reactions such as glucuronidation are relatively preserved
`and in some cases stimulated by CRF.
`In summary, various animal models of ARF and CRF
`have shown reduced activity of hepatic cytochrome
`P-450 in which certain pathways appear to be affected
`and others remain intact. Later studies have shown that
`certain cytochrome P-450 isozymes are down-regulated
`and that reduced protein expression correlates with
`reduced cytochrome P-450 activity measured by specific
`probes, while the expression of other isozymes and their
`activity remains unchanged. Surprisingly levoproprano-
`lol hepatic extraction is inhibited by uremic blood in
`perfused normal rat liver. Uremic livers showed no
`impairment in hepatic first pass when perfused with
`normal blood, suggesting a rapidly acting circulating
`competitive inhibitor of cytochrome P-450 activity
`without down-regulation of cytochrome P-450 expres-
`sion in the uremic liver. This is consistent with the finding
`that CYP2D6, the isozyme that metabolizes levopro-
`pranolol, is not down-regulated in rat CRF models and
`suggests that two mechanisms may explain the reduced
`cytochrome P-450 activity: circulating factors and
`decreased protein expression. Some phase II reactions
`such as glucuronidation appear to be stimulated in CRF.
`The mechanisms for these phenomena have not been
`elucidated.
`
`Clinical Investigations: Effect of CRF on Drug
`Disposition
`
`A number of reviews on the effect of CRF on drug
`disposition exist in the literature (2,4,5,32–34). One of the
`earliest clinical studies showed a reduction in the
`nonrenal elimination (acetylation) of sulfisoxazole given
`intravenously to patients with CRF and questioned the
`concept of therapeutics prevalent in the 1960s that
`azotemic patients should receive the usual doses of drugs
`eliminated by hepatic metabolism (34,35). A later review
`showed decreased reduction, acetylation, and hydrolysis
`reactions but preserved sulfation, glucuronidation, and
`oxidation reactions (34).
`Clinical data exist showing significant alterations
`in nonrenal clearance in CRF for a number of drugs
`(Table 3) (36–61). The majority of these studies involve
`ESRD patients; a few included subjects with moderate to
`severe CRF. These alterations include both substantial
`
`TABLE 3. Effect of CRF on nonrenal clearance
`
`Drug
`
`Increased
`Phenytoin
`Fosinopril
`Bumetanide
`
`Decreased
`Acyclovir
`Aztreonam
`Cefotaxime
`Cilastin
`Imipenem
`Moxalactam
`Captopril
`Procainamide
`Nimodipine
`Verapamil
`Metoclopramide
`Desmethyldiazepam
`Warfarin
`
`NA, not available.
`
`Change in
`nonrenal clearance (%)
`
`Metabolism
`
`57
`70
`57
`
`50
`33
`40
`92
`58
`63
`50
`60
`87
`54
`66
`63
`50
`
`CYP2C9, 2C19
`NA
`NA
`
`NA
`NA
`NA
`NA
`NA
`NA
`Sulfoxidation
`NAT-2
`CYP3A4
`CYP3A4
`CYP2D6
`CYP2C9
`CYP2C9
`
`TABLE 4. Effect of CRF on bioavailability
`
`Propranolol
`Erythromycin
`Tacrolimus
`Propoxyphene
`
`Increased
`Increased
`Increased
`Increased
`
`reductions and increases (36,39) in nonrenal clearance
`ranging from 30 to 90%. In the case of phenytoin, the
`50% increase in total hepatic clearance is produced by a
`two- to threefold increase in free fraction (fu ¼ 20–30%)
`due to reduced plasma protein binding (normal
`fu ¼ 10%) (10,36). This results in a reduction in steady-
`state total phenytoin concentration to 5–10 lg/ml with
`no change in free concentration (1–2 lg/ml) because
`intrinsic hepatic clearance is not altered. Increasing the
`dose of phenytoin to achieve a target concentration of
`10–20 lg/ml for total phenytoin may raise free concen-
`trations to toxic levels.
`For high-extraction drugs, the effect of CRF is
`manifest as an increase in bioavailability (F), as shown
`in Table 4 (36,61–64). The plasma AUC of propoxy-
`phene was increased twofold in ESRD (61). For
`erythromycin, CRF results in a fourfold increase in
`plasma AUC after an oral dose (62,63). Half-life is
`unchanged and Vd is increased in renal failure, suggesting
`that the increase in AUC was due to an increase in F
`because of reduced hepatic uptake.
`In patients with CRF approaching ESRD, oral
`propranolol showed a bioavailability of 62%, compared
`with 32% in ESRD patients on hemodialysis and 19% in
`healthy volunteers (64). This is in agreement with the
`previous animal data showing an increase in F for
`propranolol
`in uremic rats and consistent with the
`pharmacokinetic principle that the bioavailability of high
`hepatic extraction drugs is increased with reductions in
`intrinsic hepatic clearance. The same investigators also
`found that the bioavailability of propranolol was greater
`on the day of dialysis (43%) compared to the day after
`dialysis (34%). These findings are in agreement with the
`
`Boehringer Ex. 2018
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 4
`
`

`
`CRF AND DRUG METABOLISM
`
`49
`
`animal data in isolated perfused livers which suggest that
`a circulating dialyzable inhibitory factor is present in
`uremic serum. However, another investigator found no
`significant difference in apparent oral clearance, total
`clearance, or bioavailability of propranolol in stable
`renal
`failure patients with creatinine clearance of
`approximately 15 ml/min (65). It is possible that in these
`patients there is enough residual renal function to
`prevent the inhibition of hepatic metabolism.
`Zidovudine is eliminated primarily by glucuronidation
`and 25% is excreted unchanged by the kidneys. The
`plasma AUC was doubled in the CRF group receiving a
`single 200 mg oral dose, indicating a decrease in intrinsic
`hepatic clearance, suggesting that hepatic conjugation
`reactions can be reduced in CRF (66).
`is also
`Another conjugation reaction, acetylation,
`altered in ESRD. N-acetyltransferase (NAT-2) exhibits
`genetic polymorphisms, with approximately half of the
`African American and Caucasian populations exhibit-
`ing either the rapid or the slow acetylator phenotype.
`The effect of ESRD on acetylator phenotype was
`addressed in the following study (67). First, isoniazid
`and acetylisoniazid pharmacokinetics were determined
`after a single 400 mg oral dose of isoniazid and the
`same protocol repeated after renal transplantation.
`Based on the half-life of isoniazid and using the
`criteria of Reidenberg et al. (68), patients were divided
`into rapid and slow acetylator groups. The pharma-
`cokinetic parameters were compared pre- and post-
`transplantation within each phenotypic group. There
`was a 50% reduction in CLNR in CRF patient in the
`rapid acetylators which was reversed by transplanta-
`tion and there was a threefold reduction in CLNR in
`slow acetylators which was also reversed by transplan-
`tation. This demonstrated that ESRD affected the
`NAT-2 phenotype, reducing the activity of both rapid
`and slow acetylators, and the slow acetylator pheno-
`type was inhibited to a much greater extent.
`These data suggest that drugs which are cleared
`extensively by the liver and exhibit polymorphic meta-
`bolism may be at much higher risk of adverse drug
`reactions in CRF, since the poor metabolizer phenotype
`may be more susceptible to the inhibitory effects of CRF
`on metabolism (67). We have preliminary data from our
`hemodialysis unit to show that ESRD reduces hepatic
`CYP2C9 activity by 50% as measured by the S/R
`warfarin ratio as a phenotypic probe (69). (S-warfarin is
`metabolized exclusively by CYP2C9, while R-warfarin is
`metabolized by multiple pathways.)
`
`Conclusion
`
`There is abundant evidence that nonrenal clearance,
`protein binding, and distribution volume of drugs are
`altered in CRF increasing the risk of adverse drug
`reactions. The majority of the data are in patients with
`ESRD. Even drugs cleared predominantly by the liver
`are significantly affected, especially those which exhibit
`genetic polymorphisms (67,69). Caution should be
`exercised in dosing the drugs in Tables 3 and 4, which
`show significant reductions in nonrenal clearance and
`
`increased bioavailability. Careful titration from the
`lowest dose should be attempted.
`One mechanism of the reduced nonrenal clearance
`appears to be down-regulation of protein expression of
`specific hepatic cytochrome P-450 isozymes. Phase II
`reactions such as glucuronidation and acetylation are
`affected, showing inhibition or induction. This appears
`to be isozyme specific, as well. There is also evidence that,
`at least in the case of propranolol (CYP2D6), there may
`be circulating inhibitors in the serum of ESRD patients
`which may be dialyzable. Otherwise there are scant data
`available concerning the effect of dialysis on these
`changes in nonrenal clearance. More pharmacokinetic
`studies investigating the effect of CRF on nonrenal
`clearance, bioavailability, and plasma protein binding of
`extensively metabolized drugs and mechanisms of these
`phenomena and the effect of dialysis are needed.
`
`Acknowledgment
`
`is a recipient of the
`Albert W. Dreisbach, MD,
`Pharmaceutical Research and Manufacturers of Amer-
`ica Foundation Faculty Development Award in Clinical
`Pharmacology. This work is also supported by the
`GCRC NIH Program Grant #R5M01RR05096-09.
`
`References
`
`1. Rowland M, Tozer TN: Elimination. In: Clinical Pharmacokinetics, 2nd ed.
`Malvern, PA: Lea & Febiger, 1989:148–176
`2. Lam YW, Banerji S, Hatfield C, Talbert RL: Principles of drug administra-
`tion in renal insufficiency. Clin Pharmacokinet 32:30–57, 1997
`3. Anders MW: Metabolism of drugs by the kidney. Kidney Int 18:636–647,
`1980
`4. Gibson TP: Renal disease and drug metabolism: an overview. Am J Kidney
`Dis 8:7–17, 1986
`5. Elston AC, Bayliss MK, Park GR: Effect of renal failure on drug metabolism
`by the liver. Br J Anaesth 71:282–290, 1993
`6. Reidenberg MM, Drayer DE: Alterations of drug protein binding in renal
`disease. Clin Pharmacokinet 9(suppl 1):18–26, 1984
`7. Ochs HR, Rauh HW, Greenblatt DJ, Kaschell HJ: Chlorazepate dipotas-
`sium and diazepam in renal insufficiency: serum concentrations and protein
`binding of diazepam and desmethyldiazepam. Nephron 37:100–104, 1984
`8. Andreasen F: Protein binding of drugs in plasma from patients with acute
`renal failure. Acta Pharmacol Toxicol 32:417–429, 1973
`9. Hooper WD, Bochner F, Eadie MJ: Plasma protein binding of diphenylhy-
`dantion: effects of sex hormones, renal and hepatic disease. Clin Pharmacol
`Ther 49:457–467, 1991
`10. Reidenberg MM, Odar-Cederlof I, Von Bahr C, Borga O, Sjoqvist F: Protein
`binding of diphenylhydantoin and desmethylimipramine in plasma from
`patients with poor renal function. N Engl J Med 285:264–267, 1971
`11. Farrell PC, Grib NL, Fry DL, Popovich RP, Broviac JW, Babb AL: A
`comparison of in vitro and in vivo solute-protein binding interactions in
`normal and uraemic subjects. Trans Am Soc Artif Intern Organs 18:268–276,
`1972
`12. Lowenthal DT, Briggs WA, Lev G: Kinetics of salicylate elimination in
`anephric patients. J Clin Invest 54:1221–1226, 1974
`13. Andreasen F, Jakobsen P: Determination of frusemide in blood plasma and
`its binding to proteins in normal plasma and in plasma of patients with acute
`renal failure. Acta Pharmacol Toxicol 35:49–57, 1974
`14. Rane A, Villeneuve JP, Stone WJ, Nies AS, Wilkinson GR, Branch RA:
`Plasma binding and distribution of frusemide in the nephrotic syndrome and
`uraemia. Clin Pharmacol Ther 24:199–207, 1978
`15. Bachmann K, Shapiro R, Mackiewicz J: Influence of renal dysfunction on
`warfarin plasma protein binding. J Clin Pharmacol 16:168–172, 1976
`16. Belpaire FM, Bogaert MG, Mussche MM: Influence of renal failure on the
`protein binding of drugs in animals and man. Eur J Clin Pharmacol 11:27–32,
`1977
`17. Haughey DB, Kraft CJ, Matzke GR, Keane WF, Halstenson CE: Protein
`binding of disopyramide and elevated alpha-1-acid glycoprotein concentra-
`tions in serum obtained from dialysis patients and renal transplant recipients.
`Am J Nephrol 5:35–39, 1985
`
`Boehringer Ex. 2018
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 5
`
`

`
`50
`
`Dreisbach and Lertora
`
`18. Tan CC, Lee HS, Ti TY, Lee EJ: Pharmacokinetics of intravenous
`vancomycin in patients with end-stage renal failure. Ther Drug Monit
`12:29–34, 1990
`19. Koch-Weser J, Sellers EM: Binding of drugs to serum albumin. N Engl J Med
`294:311–316, 526–531, 1976 (2 parts)
`20. Dromgoole SH: The binding capacity of albumin and renal disease.
`J Pharmacol Exp Ther 191:318–323, 1974
`21. Wilkinson GR, Shand DG: Commentary: a physiological approach to
`hepatic drug clearance. Clin Pharmacol Ther 18:377–390, 1975
`22. Yuan R, Venitz J: Effect of chronic renal failure on the disposition of highly
`hepatically metabolized drugs. Int J Clin Pharmacol Ther 38:245–253, 2000
`23. Leblanc M, Roy LF, Villeneuve JP, Malo B, Pomier-Layrargues G, Legault
`L: Liver blood flow in chronic hemodialysis patients. Nephron 73:396–402,
`1996
`24. Leber HW, Schutterle G: Oxidative drug metabolism in liver microsomes
`from uremic rats. Kidney Int 2:152–158, 1972
`25. VanPeer AP, Belpaire FM: Hepatic oxidative drug metabolism in rats
`with experimental renal failure. Arch Int Pharmacodyn Ther 228:180–183,
`1977
`26. Terao N, Shen DD: Effect of experimental renal failure on the disposition
`kinetics of l-propranolol in rats. J Pharmacol Exp Ther 227:295–301, 1983
`27. Terao N, She DD: Reduced extraction of l-propranolol by perfused rat liver
`in the presence of uremic blood. J Pharmacol Exp Ther 233:277–284, 1985
`28. Patterson SE, Cohn VH: Hepatic drug metabolism in rats with experimental
`chronic renal failure. Biochem Pharmacol 33:711–716, 1984
`29. Leblond FA, Groux L, Villeneuve JP, Pichette V: Decreased in vivo
`metabolism of drugs in chronic renal failure. Drug Metab Dispos 28:1317–
`1320, 2000
`30. Leblond F, Geurin C, Demers C, Pellerin I, Gascon-Barre M, Pichette V:
`Down regulation of hepatic cytochrome P450 in chronic renal failure. J Am
`Soc Nephrol 12:326–332, 2001
`31. Leber HW, Gleumes L, Schutterle G: Enzyme induction in the uremic liver.
`Kidney Int 13(suppl 8):S43–S48, 1978
`32. Reidenberg MM: Drug metabolism in uremia. In: Renal Function and Drug
`Action. Philadelphia: WB Saunders, 1971:19–32
`33. Touchette MA, Slaughter RL: The effect of renal failure on hepatic drug
`clearance. DICP Ann Pharmacother 25:1214–1224, 1991
`34. Talbert RL: Drug dosing in renal insufficiency. J Clin Pharmacol 34:99–110,
`1994
`35. Reidenberg MM, Kostenbauder H, Adams WP: Rate of drug metabolism in
`obese volunteers before and during starvation and azotemic patients.
`Metabolism 18:209–213, 1969
`36. Letteri JM, Mellk H, Louis S, Kutt H, Durante P, Glazko A: Diphenylhy-
`dantoin metabolism in uraemia. N Engl J Med 285:648–652, 1971
`37. Odar-Cederlof I, Borga O: Kinetics of diphenylhydantoin in uraemic
`patients: consequences of decreased plasma protein binding. Eur J Clin
`Pharmacol 7:31–37, 1974
`38. Hui KK, Duchin KL, Kripalani KL, Chan D, Kramer PK, Yanagawa N:
`Pharmacokinetics of fosinopril in patients with various degrees of renal
`function. Clin Pharmacol Ther 49:457–467, 1991
`39. Marcantonio LA, Auld WHR, Murdoch WR, Purohit R, Skellerin GG,
`Howes CA: The pharmacokinetics and pharmacodynamics of the diuretic
`bumetanide in hepatic and renal disease. Br J Clin Pharmacol 15:245–252,
`1983
`40. Laskin OL, Longsteth JA, Saral R, deMiranda P, Keeney R, Lietman PS:
`Pharmacokinetics and tolerance of acyclovir, a new antiherpes agent, in
`humans. Antimicrob Agents Chemother 21:393–398, 1982
`41. Laskin OL, Longsteth JA, Whelton A, Rocco L, Lietman PS, Krasny HC,
`Keeney RE: Acyclovir kinetics in end-stage renal disease. Clin Pharmacol
`Ther 31:594–601, 1982
`42. deMiranda P, Good SS, Laskin OL, Krasny HC, Connor JD, Lietman PS:
`Disposition of intravenous radioactive acyclovir. Clin Pharmacol Ther
`30:662–672, 1981
`43. Mihindu JCL, Krasny HC, Page DA, Scheld WM, Bolton ND, Spyker DA,
`Swabb EA, Boltion WK: Pharmacokinetics of aztreonam in patients with
`various degrees of renal dysfunction. Antimicrob Agents Chemother 24:252–
`261, 1983
`44. Watson AJS, Stout RL, Whelton A: The intrarenal distribution of aztreonam
`in healthy and diseased kidneys: Clinical therapeutic implications. J Infect Dis
`150:631–635, 1984
`45. Ings RMJ, Fillastre J-P, Godin M, Leroy A, Humbert G: Pharmacokinetics
`of cefotaxime in subjects with normal and impaired renal function. Rev Infect
`Dis 6:809–815, 1980
`
`46. Fillastre JP, Leroy A, Humbert G, Godin M: Pharmacokinetics of
`cefo

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