throbber
Current Drug Metabolism, 2003, 4, 91-103
`
`91
`
`Drug Metabolism in Chronic Renal Failure
`
`Vincent Pichette* and François A. Leblond
`
`Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine,
`Université de Montréal, Québec, Canada
`
`Abstract: Pharmacokinetic studies conducted in patients with CRF demonstrate that the nonrenal clearance of
`multiple drugs is reduced. Although the mechanism by which this occurs is unclear, several studies have shown
`that CRF affects the metabolism of drugs by inhibiting key enzymatic systems in the liver, intestine and kidney.
`The down-regulation of selected isoforms of the hepatic cytochrome P450 (CYP450) has been reported secondary to
`a decrease in gene expression. This is associated with major reductions in metabolism of drugs mediated by
`CYP450. The main hypothesis to explain the decrease in liver CYP450 activity in CRF appears to be the
`accumulation of circulating factors which can modulate CYP450 activity. Liver phase II metabolic reactions are also
`reduced in CRF. On the other hand, intestinal drug disposition is affected in CRF. Increased bioavailability of
`several drugs has been reported in CRF, reflecting decrease in either intestinal first-pass metabolism or extrusion of
`drugs (mediated by P-glycoprotein). Indeed, intestinal CYP450 is also down-regulated secondary to reduced gene
`expression, whereas, decreased intestinal P-glycoprotein activity has been described. Finally, although the kidneys
`play a major role in the excretion of drugs, it has the capacity to metabolize endogenous and exogenous
`compounds. CRF will lead to a decrease in the ability of the kidney to metabolize drugs, but the repercussions on
`the systemic clearance of drugs is still poorly defined, except for selected xenobiotics. In conclusion, reduced drug
`metabolism should be taken into account when evaluating the pharmacokinetics of drugs in patients with CRF.
`
`Key Words: Chronic renal failure, cytochrome P450, gene expression, drug metabolism, intestine, liver, serum mediators,
`P-glycoprotein.
`
`1. INTRODUCTION
`
`(CRF),
`failure
`renal
`The prevalence of chronic
`predominantly due to aging, diabetes, hypertension and renal
`vascular disease, has increased steadily during the last
`decades [1, 2]. The cost of care for these patients has also
`risen progressively
`in part because of excessive or
`inappropriate drug use [3, 4]. Some studies have revealed
`that patients with CRF require an average of more than seven
`drugs to manage the underlying renal disease and their
`comorbid states [3]. Furthermore 40% of patients with
`creatinine clearance of less than 40 mL/min receive drug
`dosages higher (range 1.07-6.45) than required [5]. Both,
`excessive use of drugs and especially inappropriate dosage
`increase the risk of adverse effects, including the risk of
`nephrotoxicity, and increase the cost of care [3, 4, 6]. For
`instance, Johnson and Bootman have estimated
`that
`$US76.6 billion are spent every year to manage drug-related
`morbidity and mortality [7]. Thus, appropriate dosage of
`drugs in CRF is a crucial consideration in avoiding adverse
`effects, minimizing the time and cost of management of
`adverse events, and ensuring optimal patient outcome.
`
`CRF interferes with the elimination of many drugs
`because of the reduction in glomerular filtration rate (GFR),
`
`*Address correspondence to this author at the Centre de recherche Guy-
`Bernier, Hôpital Maisonneuve-Rosemont, 5415 boul. de l’Assomption,
`Montréal, Québec, Canada H1T 2M4; Tel: (514) 252-3489; Fax: (514 255-
`3026; E-mail: vpichette.hmr@ssss.gouv.qc.ca
`
`and tubular secretion [8]. Dose adjustment of drugs excreted
`by the kidney is made according to the GFR. However
`despite dosage adjustment, patients with CRF still present a
`great number of adverse effects [6, 8]. Part of
`this
`phenomenon is related to the fact that CRF also affects the
`nonrenal route of elimination of drugs, i.e. hepatic, intestinal
`and renal metabolism of drugs is decreased in CRF [6, 9-
`14]. It could also be explained by alteration in non-metabolic
`elimination of drugs by the liver and the intestine, i.e. drug
`elimination mediated by P-glycoprotein.
`
`The aim of this article is to review drug metabolic
`disturbances
`in
`renal
`failure, emphasizing on drug
`metabolizing enzymes, particularly cytochrome P450
`(CYP450). The repercussions of renal failure on intestinal
`and liver P-glycoprotein will also be reviewed.
`
`2. LIVER DRUG METABOLISM
`
`2.1. Human Studies
`
`Many investigators have shown, that in patients with
`renal failure there is a decrease in the metabolic clearance of
`numerous drugs. Several reviews have been published on the
`subject [3, 4, 6, 8, 9, 13]. Some of these drugs are presented
`in table 1. The vast majority is metabolized in the liver,
`suggesting that renal failure impedes hepatic biotransfor-
`mation of drugs. Several of the substrates in table 1 are
`eliminated catalyzed by the CYP450, suggesting that hepatic
`CYP450 is altered in CRF [9, 15, 16].
`
` 1389-2002/03 $41.00+.00
`
`© 2003 Bentham Science Publishers Ltd.
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 1
`
`

`
`92 Current Drug Metabolism, 2003, Vol. 4, No. 2
`
`Pichette and Leblond
`
`Table 1. Effect of Chronic Renal Failure on Nonrenal Clearance (% Change from Normal Clearance)
`
`Acyclovir (62)
`
`Aztreonam (33)
`
`Captopril (50)
`
`Cefmenoxime (45)
`
`Cefonicid (60)
`
`Cefsulodin (52)
`
`Cimetidine (62)
`
`Ciprofloxacin (33)
`
`Codeine (17)
`
`Fluconazole (50)
`
`Imipenem (85)
`
`Metclopramide (66)
`
`Moxalactam (63)
`
`Minoxidil (46)
`
`Nicardipine (37)
`
`Procainamide (61)
`
`Verapamil (54)
`
`Adapted from Lam, Y, W.; Banerji, S.; Hatfield, C. and Talbert, R.L. (1997) Principles of drug administration in renal insufficiency. Clin Pharmacokinet., 32 (1), 30-57. with
`permission from publisher Adis International Ltd.
`
`We are not aware of human studies demonstrating a
`reduction in liver cytochrome P450 isoforms in CRF.
`However, several methods have been described to assess, in
`vivo, the CYP activity in humans and the most widely used
`is the administration of probe drugs that are selectively
`metabolized by specific CYP450 isoforms [17]. Kevorkian
`et al. reported that the use of dextromethorphan or sparteine,
`to assess CYP2D6 activity, was possible in patients with
`CRF [18]. The results of their study revealed that there was a
`decrease in the metabolic clearance of sparteine, suggesting a
`decrease in CYP2D6 activity in CRF patients [18]. Other
`authors have also suggested that CYP2D6 is compromised
`in parallel with the deterioration of renal function in CRF
`patients [19].
`
`There are few studies on liver phase II reactions in CRF.
`Sulfation appears
`to be normal, whereas acetylation
`(isoniazide) and glucuronidation (fluconazole) have been
`reported to be reduced [20-22] (see section 2.2.2).
`
`Although the studies cited above strongly suggest that
`renal failure reduces the metabolism of drugs in CRF
`patients, it is still difficult to predict exactly how renal
`failure will influence the disposition of a specific drug. Part
`of this problem comes from the fact that most clinical
`studies were made
`in conditions where concurrent
`medications, age and smoking habits (all factors known to
`strongly influence CYP450) were not necessarily controlled.
`Furthermore, the severity of the renal failure should be taken
`into account in interpreting pharmacokinetics studies in renal
`failure because some investigators have reported a correlation
`between the decrease in GFR and the decrease in nonrenal
`clearance [6, 23-25]. In addition to the severity, the duration
`of
`the renal failure seems
`to
`influence
`the hepatic
`metabolism. For
`instance,
`the nonrenal clearance of
`imipenem was 50 ml/min in patients with CRF, 95 ml/min
`in patients with acute renal failure (ARF) and 130 ml/min in
`control patients [26].
`
`In summary, renal failure in human is associated with a
`decrease in
`the metabolism of several drugs that are
`preferentially biotransformed by the liver. This decrease in
`biotransformation appears to be related to a reduction in the
`activity of hepatic CYP450, and the importance of this
`phenomenon seems to be related to the severity and the
`duration of renal failure.
`
`2.2. Animal Studies
`
`2.2.1. Cytochrome P450
`
`2.2.1.1. In Vitro Metabolism
`Several studies have looked at the repercussions of
`experimental renal failure on microsomial and cytosolic
`enzyme content of the liver [27-34]. Most of the studies have
`focused on the CYP450 since it is the major enzymatic
`system involved in drug metabolism. The results of these
`studies show that in male rats with CRF, total hepatic
`CYP450 content decreases between 19 to 47%. Moreover,
`significant reductions in enzymatic reactions normally carried
`by the CYP450 have been reported in vitro: N-demethylation
`of erythromycin, aminopyrine and ethylmorphine, O-
`demethylation of codeine and hydroxylation of aniline. There
`is a correlation between the decrease in total CYP450
`activity and the severity of renal failure in rats [30-31, 33,
`34]. We also reported a correlation between the reduction in
`creatinine clearance and the decrease in in vitro metabolism
`of erythromycin [33, 34].
`
`2.2.1.2. CYP450 Protein Expression
`is
`In male adult rat, hepatic cytochrome CYP450
`composed of several isoforms and those involved in drug
`metabolism processes
`include CYP1A2, CYP2C11,
`CYP2D, CYP2E1 and CYP3A1/3A2 [35]. Knowledge of
`which isoforms are decreased in CRF is critical in order to
`predict which drugs are at risk for accumulation. Although
`several studies have focused on the repercussions of CRF on
`total hepatic CYP450 content, only few investigators have
`studied specific CYP450 isoforms. Uchida et al. reported a
`reduction in the levels of hepatic CYP2C6, CYP2C11 and
`CYP3A2 and a slight increase in CYP1A2 in male adult rats
`with CRF [31]. In our laboratory, we described a significant
`reduction in CYP2C11, CYP3A1 and CYP3A2 in male
`adult rats with CRF, while no isoform induction was noted
`(Fig. (1)) [33, 34]. Furthermore, the levels of CYP450
`protein were inversely correlated with the degree of renal
`failure, assessed by the creatinine clearance [33, 34].
`Interestingly, CYP3A1 and 3A2 in the rat correspond to
`CYP3A4 in humans. Since this isoform is responsible for
`the metabolism of multiple drugs commonly used in CRF
`patients, patients with CRF could be at risk for drug
`accumulation and toxicity.
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 2
`
`

`
`Drug Metabolism in Chronic Renal Failure
`
`Current Drug Metabolism, 2003, Vol. 4, No. 2 93
`
`Fig. (1). Protein expression of liver CYP450 isoforms in control paired-fed (c) and CRF rats (g). Protein bands are expressed in
`densitometry units (%). The densitometry units of control paired-fed rats were arbitrarily defined as 100%. Data are the mean –
` S.E. of
`six rats in each group.
`
`*P < 0.001 compared with control paired-fed rats.
`
`Reproduced from Leblond, F.A.; Giroux, L.; Villeneuve, J.P. and Pichette, V. (2000) Decreased in vivo metabolism of drugs in chronic
`renal failure. Drug Metab. Dispos., 28(11), 1317-1320 with permission from the American Society for Pharmacology and
`Experimental Therapeutics .
`
`2.2.1.3. In Vivo Metabolism
`is
`In vitro studies clearly demonstrated that CRF
`associated with a down-regulation of
`liver CYP450.
`However, the consequences of
`liver CYP450 decrease
`induced by CRF on the in vivo drug metabolizing capacity
`remain poorly documented. In animals, very few data are
`available on the repercussions of CRF on systemic drug
`metabolism by the liver. A major problem with drug
`disposition studies in rats is that the blood samples used for
`pharmacokinetic analysis can lead to significant blood loss
`and hypovolemia. Uchida et al. studied the changes in
`trimethadione (TMO) metabolism using the microdialysis
`method (avoiding excessive blood samples) in CRF rats
`[31]. They found that the N-demethylation of TMO was
`reduced by 25 % in CRF. However, since TMO N-
`demethylation is catalyzed by several CYP450 isoforms, a
`reduction in its metabolism does not indicate which specific
`isoform is reduced [36]. On the other hand, Tvedegaard et al.
`found no modification in the antipyrine clearance in rabbits
`with CRF [37].
`
`We used breath tests as probes for evaluating in vivo liver
`metabolism in CRF rats. Breath tests have been developed
`as methods to evaluate the catalytic activity of CYP450
`isoenzymes by measuring the rate of demethylation of a drug
`[38]. The formaldehyde generated by CYP450 mediated
`demethylation reactions is rapidly oxidized and excreted as
`carbon dioxide in the breath. The rate of production of 14CO2
`from a suitable radiolabelled substrate reflects the in vivo rate
`of its demethylation and thus the catalytic activity of either a
`subset or a specific cytochrome P450 depending on the
`studied substrate. Various substrates have been used in
`breath tests to evaluate CYP450 activity in vivo in rats and
`humans. The aminopyrine breath test has often been used to
`evaluate liver metabolic function [39, 40]. In vitro and in
`vivo studies suggest that aminopyrine breath tests can be
`used to evaluate the activity of CYP2C11 in the rat [41, 42]
`although other CYP450 isoenzymes also contribute to its
`demethylation, including CYP1A2, 2A2, 2B, and 2D1.
`Caffeine and erythromycin breath tests have also been used
`to measure the liver catalytic activity of CYP1A2 and
`CYP3A2 isoenzymes [42-45].
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 3
`
`

`
`94 Current Drug Metabolism, 2003, Vol. 4, No. 2
`
`Pichette and Leblond
`
`In our study, the aminopyrine breath test was used to
`evaluate the activity of CYP2C11; caffeine and erythromycin
`breath tests were used to measure the catalytic activity of
`CYP1A2 and CYP3A2
`isoenzymes,
`respectively. We
`reported that aminopyrine and erythromycin breath tests were
`reduced by 35 % in CRF rats, while the caffeine breath test
`remained unchanged (Fig. (2)) [32]. We also found a
`correlation between the reduction in creatinine clearance and
`the decrease in the in vivo metabolism of drugs. These
`results demonstrate that in rats, CRF is associated with a
`reduction in the metabolism of drugs in vivo, secondary to a
`decrease in selective liver CYP450
`isoforms, namely
`CYP2C11 and CYP3A1/3A2. Whether
`reduction
`in
`extrahepatic metabolism of drugs (e.g. intestinal), that could
`further participate in the reduction of drug metabolism found
`in this study, remains to be defined.
`
`2.2.1.4. CYP450 Gene Expression
`The reduction in liver CYP450 in CRF could be secon-
`dary to a decrease in synthesis or an increase in degradation.
`Our group studied, in the male adult rat, the repercussions of
`CRF on the CYP450 gene expression in the liver [33, 34].
`The results demonstrated that there was an association
`between lower levels of mRNAs and protein expression for
`several isoforms of hepatic cytochrome P450, namely
`CYP2C11, 3A1 and 3A2 [33, 34]. These results strongly
`suggest that CRF leads to a reduction in gene expression of
`liver CYP450
`isoforms. However,
`the mechanisms
`underlying the diminution of liver CYP2C11, 3A1 and 3A2
`gene expression in CRF are not known. Caloric restriction,
`as seen in CRF rats, down-regulates hepatic genes of drug
`metabolizing enzymes in the mouse and in the rat [46, 47].
`However, in our control paired-fed rats we did not observe
`
`Fig. (2). Breath tests with erythromycin (EBT), aminopyrine (ABT), and caffeine (CBT) in control paired-fed (c) and CRF rats (g).
`Values represent the 2-h cumulative 14CO2 output expressed as a percentage of the injected dose. Data are the mean –
` S.E. of six rats in
`each group.
`
`*P < 0.001 compared with control paired-fed rats.
`
`Reproduced from Leblond, F.A.; Giroux, L.; Villeneuve, J.P. and Pichette, V. (2000) Decreased in vivo metabolism of drugs in chronic
`renal failure. Drug Metab. Dispos., 28(11), 1317-1320 with permission from the American Society for Pharmacology and
`Experimental Therapeutics .
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 4
`
`

`
`Drug Metabolism in Chronic Renal Failure
`
`Current Drug Metabolism, 2003, Vol. 4, No. 2 95
`
`any modification in cytochrome CYP450 levels despite a
`similar weight loss in controls and CRF rats [33, 34].
`
`2.2.1.5. Mechanisms Implicated in Liver CYP450 Activity
`and Expression Down-Regulation
`The mechanism underlying the down-regulation of liver
`CYP450 in CRF remains poorly understood. Leber and
`Schutterle have shown that in CRF, the reduction of liver
`CYP450 induced by CRF could be reversed in part by l-
`aminolevulinic acid but
`these results have not been
`confirmed [27]. It appears excluded that chronic protein
`malnutrition, as seen in CRF, could affect CYP450
`synthesis in the liver [33, 34].
`
`An attractive hypothesis to explain CYP450 activity and
`expression down regulation is the presence of endogenous
`inhibitors in the blood of uremic animals that may modulate
`the activity of CYP450. Terao and Chen have shown, in
`single-pass rat liver perfusion studies, that the extraction of
`L-propranolol was significantly lower in rats with acute renal
`failure compared to control rats [48]. When livers of rats
`with acute renal failure were perfused with blood from
`control rats, the extraction of L-propranolol was similar to
`that of control rats. However, when livers of control rats
`were perfused with blood
`from animals with
`renal
`insufficiency,
`there was a significant decrease
`in L-
`propranolol extraction. Although this study was done in rats
`using an acute renal failure model and reported no data on
`drug metabolizing enzyme, it provided the first evidence for
`the presence of an inhibitory factor in the uremic blood that
`could modify the biotransformation of drugs [48].
`
`The presence of a circulating factor in uremia that inhibits
`liver metabolism has also been implicated in CRF patients.
`Ahmed et al. studied the pharmacokinetics of nicardipine in
`three groups, control subjects, patients with CRF and
`patients undergoing hemodialysis [49]. Plasma clearance of
`nicardipine was reduced in patients with CRF (6.5 –
` 2.6
`ml/min/kg) compared with control subjects (10.4 –
` 3.1
`ml/min/kg) and patients undergoing hemodialysis (12.5 –
`4.6 ml/min/kg). The fact that the clearance of nicardipine
`was identical in control subjects and patients undergoing
`hemodialysis suggest that the inhibitors can be removed by
`dialysis. Further supporting the presence of serum inhibitors,
`it has been shown that incubation of microsomes, prepared
`from healthy human livers, with serum of patients with CRF
`is associated with a decrease
`in
`the metabolism of
`midazolam and
`tolbutamide,
`reflecting CYP3A4 and
`CYP2C9 activities [50].
`
`In order to confirm the presence of serum mediators
`affecting activity and/or expression of the isoforms of hepatic
`CYP450, we recently conducted a study where we incubated
`normal rat hepatocytes with serum from CRF and control
`male adult rats [51]. This study revealed, that in normal
`hepatocytes incubated for 24 hours with serum (concentration
`of 10%) from rats with CRF, total CYP450 level decreased
`by 35% compared to serum from control animals [51]. We
`also showed that protein expression of several CYP450
`isoforms (CYP2C6, 2C11, 3A1 and 3A2) were decreased by
`more than 35% in normal hepatocytes incubated with serum
`from CRF rats (Fig. (3)). The decrease in protein expression
`of CYP450 isoforms mediated by serum from rats with CRF
`
`was secondary to reduced gene expression (Fig. (4)).
`Although the mechanisms responsible for the diminished
`hepatic gene expression in CRF are not known, the present
`study suggests that uremic mediator(s) may affect CYP450
`promoters.
`
`A number of studies indicate that animals with CRF
`display
`impaired protein synthesis, by
`reduced gene
`expression in
`the liver,
`the skeletal muscle, and
`the
`cardiomyocytes [52-54]. For instance, the mRNA of hepatic
`lipase and insulin-like growth factor 1 receptor are decreased
`in hepatocytes and skeletal muscle of rats with CRF,
`respectively [52-54]. CRF is associated with sustained
`elevations in calcium in many cell types, including the
`hepatocytes. This high intracellular calcium seems to be a
`major factor underlying cell reduced protein synthesis [55,
`56]. It has been suggested that the increase in basal [Ca2+]i of
`hepatocytes was secondary to parathyroid hormone (PTH)
`elevation that accompanies CRF [57]. The predominant
`pathway for the PTH-induced increase in [Ca2+]i is the
`stimulation of a G protein-adenylate cyclase-cAMP system,
`which leads to stimulation of a calcium transport system
`[57]. On the other hand, cAMP has been shown to down-
`regulate CYP450 [58]. We recently demonstrated that the
`molecular weight of the mediator(s) present in uremic serum
`is between 10 and 30 kDa. Since rat PTH molecular weight
`is around 10 kDa , it could be a potential mediator of hepatic
`CYP450 down-regulation in CRF [51].
`
`Other potential uremic serum mediators implicated in the
`down-regulation of liver CYP450 are cytokines, since their
`molecular weight averages 20 kDa. Several studies have
`demonstrated that CRF
`is associated with a chronic
`activation of inflammatory response and patients with CRF
`present an increase in plasma levels of many cytokines [59-
`63]. On the other hand, cytokines are able to down-regulate
`hepatic CYP450 in vitro and in vivo [64-66].
`
`In summary, animal studies demonstrate that CRF is
`associated with a decrease in the expression of liver CYP450
`isoforms secondary
`to
`reduced mRNA
`levels. Drug
`metabolism activity, assessed by several oxidative reactions,
`normally carried out by the CYP450 is also depressed in rats
`with CRF. Hepatic CYP450 down-regulation is correlated
`with the degree of renal failure. We may speculate that this
`down-regulation could explain
`the reduction
`in drug
`metabolism observed in patients with CRF, since rat
`CYP3A1 and 3A2 correspond to CYP3A4 in humans which
`is responsible for the metabolism of many drugs commonly
`used in patients with CRF. The main hypothesis for
`decreased CYP450 activity and expression appears to be the
`presence of uremic factors that accumulate in CRF.
`
`2.2.2. Phase II Reactions
`Phase II reactions in CRF have not been studied as
`extensively as phase I reactions. Several human studies have
`shown that conjugation reactions can be altered in CRF.
`Singlas et al. studied the disposition of zidovudine in
`patients with CRF, drug eliminated by glucuronidation
`(75%), and renal excretion (25%), and demonstrated that
`zidovudine AUC was significantly higher in patients with
`CRF than in patients without renal failure [20]. Since the
`increase in AUC could not be explained solely by the
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 5
`
`

`
`96 Current Drug Metabolism, 2003, Vol. 4, No. 2
`
`Pichette and Leblond
`
`Fig. (3). Protein expression of hepatic cytochrome P450 isoforms in hepatocytes incubated with serum from control (c) and CRF
`rats (g). Protein bands are expressed in densitometry units (%). The densitometry units of control rats were arbitrarily defined as
`100%. Data are the mean –
` SEM of 8 experiments in each group.
` *P < 0.01 as compared to serum from control.
`
`Reproduced from Guevin, C.; Michaud, J.; Naud, J.; Leblond, F.A. and Pichette, V. (2002) Down-regulation of hepatic cytochrome
`P450 in chronic renal failure: role of uremic mediators. Br. J. Pharmacol., 137(7), 1039-1046. with permission from publisher Nature
`Publishing group .
`
`decrease in the urinary clearance, it has been suggested that
`there was a reduction in the metabolism of zidovudine.
`Several studies have examined the effect of CRF on
`metoclopramide metabolism, which is primarily mediated
`through
`conjugation
`reactions
`(glucuronidation
`and
`sulfation). Baterman et al. found a 30% reduction in the
`clearance of metoclopramide in patients with renal failure
`compared with healthy volunteers [67]. Similar results have
`been shown by Lehmann et al. [68]. Acetylation also appears
`to be reduced in patients with CRF; several reports have
`demonstrated that the metabolic clearance of procainamide is
`reduced by approximately 60% in renal failure [4, 69, 70].
`
`Moreover, a decrease in the acetylation of isoniazide has also
`been reported in patients with CRF [22].
`
`The mechanism leading to a decrease in phase II
`enzymatic reactions is unknown. Very few in vitro studies
`have been done. Paterson and Cohn studied the uridine
`diphosphate
`(UDP)-glucuronyl
`transferase activity
`in
`microsomes from control and rats with CRF and reported a
`19% reduction
`in UDP-glucuronyl
`transferase activity,
`although this difference was not significant (p=0.06) [30].
`On the other hand, Taburet et al. reported that incubation of
`microsomes, prepared from healthy human livers, with
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 6
`
`

`
`Drug Metabolism in Chronic Renal Failure
`
`Current Drug Metabolism, 2003, Vol. 4, No. 2 97
`
`Fig. (4). mRNA encoding hepatic cytochrome P450 isoforms (CYP2C11, CYP2D and 3A2) in hepatocytes incubated with serum from
`control (c) and CRF rats (g). mRNA bands are expressed in standardised densitometry units (%). The densitometry values for
`cytochrome P450 isoforms were standardised by dividing these values by the values for 18S ribosomal RNA. The standardised
`densitometry units of control rats were arbitrarily defined as 100%. Data are the mean –
` SEM of 8 experiments in each group.
`*P < 0.005 as compared to serum from control rats.
`
`Reproduced from Guevin, C.; Michaud, J.; Naud, J.; Leblond, F.A. and Pichette, V. (2002) Down-regulation of hepatic cytochrome
`P450 in chronic renal failure: role of uremic mediators. Br. J. Pharmacol., 137(7), 1039-1046. with permission from publisher Nature
`Publishing group .
`
`serum of patients with CRF was associated with a decrease
`in the metabolism of zidovudine, reflecting an inhibition of
`UDP-glucuronyl
`transferase [50]. As demonstrated for
`hepatic CYP450, these data strongly suggest the presence of
`serum factors in the blood of patients with CRF that could
`not only down-regulate liver CYP450, but also other
`enzymatic reactions implicated in the metabolism of drugs,
`such as glucuronidation and acetylation.
`
`3. INTESTINAL DRUG METABOLISM
`
`in drug
`role
`liver plays a major
`the
`Although
`metabolism, enzymes that contribute to the metabolism of
`
`drugs are also present at other sites, particularly in the
`intestine [71, 72]. The predominant biotransformation
`system in the intestine is the cytochrome P450 [73]. The
`intestine contains several isoforms of the families CYP1A,
`CYP2B, CYP2C, CYP2D and CYP3A [72-74]. The ability
`of these CYP450 isoforms to metabolize drugs is as
`important as that of the liver. More importantly, small
`intestinal CYP450 provides the principal, initial source of
`biotransformation for
`ingested oral xenobiotics. Small
`intestinal CYP450 is implicated in the phenomenon of first-
`pass metabolism which prevents the absorption of drugs
`[75]. Thus, any modification in intestinal CYP450 could
`have important repercussions on the bioavailability of
`xenobiotics [75].
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 7
`
`

`
`98 Current Drug Metabolism, 2003, Vol. 4, No. 2
`
`Pichette and Leblond
`
`Several studies have been conducted to assess the effect of
`CRF on the bioavailability of drugs, and have documented
`that
`the bioavailability
`of
`propranolol,
`tolamolol,
`dihydrocodeine,
`dextropropoxyphene,
`oxprenolol,
`erythromycin, and tacrolimus is increased in patients with
`CRF [3, 6, 76-79]. These observations indicate that in
`patients with CRF, presystemic or first-pass metabolism of
`selected drugs is reduced. Since some of these drugs (e.g.
`propranolol,
`dextropropoxyphene,
`tacrolimus,
`and
`erythromycin) are subject to intestinal first-pass metabolism,
`it is likely that CRF also diminishes intestinal metabolism
`[6, 11]. In the case of tolamolol, CRF increases its
`availability without affecting its half-life, strongly suggesting
`that CRF reduces its intestinal first-pass [80].
`
`that CRF could modify
`Supporting the hypothesis
`intestinal metabolism of drugs, a number of studies indicate
`
`that other intestinal functions could be altered in CRF.
`Several enzymatic reactions carried out by the intestinal
`mucosa have been shown to be decreased in rats with CRF.
`For instance, sucrase and maltase activities are significantly
`reduced, while the activities of other dipeptidases remain
`unchanged [81]. More recently, Vaziri et al. reported that
`enteric xanthine oxidase activity was diminished in animals
`with CRF [82].
`
`Recently, we have studied the repercussions of CRF on
`intestinal CYP450 [83]. Our results revealed a major
`reduction in total intestinal CYP450 in CRF male adult rats,
`with significant decrease in protein expression of selective
`isoforms, namely CYP1A1 and 3A2 (Fig. (5)) [83]. Our
`results also demonstrated that there was an association
`between lower levels of mRNAs and protein for CYP1A1
`and CYP3A2 isoform suggesting that there is reduced gene
`
`Fig. (5). Protein expression of intestinal cytochrome P450 isoforms in control (c) and CRF rats (g). Protein bands are expressed in
`densitometry units (%). The densitometry units of control paired-fed rats were arbitrarily defined as 100%. Data are the mean –
` SD of
`20 rats in each group.
`*P = 8x10-6 and ** P = 2x10-8 as compared to control rats.
`
`Reproduced from Leblond, F.A.; Petrucci, M.; Dube. P.; Bernier, G.; Bonnardeaux, A. and Pichette, V. (2002) Down-regulation of
`intestinal cytochrome P450 in chronic renal failure. J. Am. Soc. Nephrol., 13(6), 1579-1585. with permission from publisher
`Lippincott Williams & Wilkins.
`
`Boehringer Ex. 2019
`Mylan v. Boehringer Ingelheim
`IPR2016-01565
`Page 8
`
`

`
`Drug Metabolism in Chronic Renal Failure
`
`Current Drug Metabolism, 2003, Vol. 4, No. 2 99
`
`expression. The metabolic consequences are important since
`we observed a 30 % decrease in the in vitro metabolism of
`substrate mediated by these two isoforms [83]. Interestingly,
`CYP1A1 and CYP3A1/3A2
`in
`the rat correspond to
`CYP1A1 and CYP3A4 in humans. Since these isoforms are
`responsible for the metabolism of several drugs commonly
`used in CRF, patients could be at risk for drug accumulation
`and toxicity. Indeed, these results could explain why the
`bioavailability of erythromycin and
`tacrolimus
`(both
`metabolized by CYP3A family in the intestine) are increased
`by more than 30 % in CRF patients [78, 79].
`
`In summary, several pharmacokinetic studies conducted
`in human demonstrate that the bioavailability of selected
`drugs is increased in patients with CRF. Although the
`mechanism is still unclear, it appears that this phenomenon
`can be related to decreased intestinal first-pass metabolism.
`Indeed, as happens in the liver, CRF decreases intestinal
`CYP450. Whether uremic circulating factors are also
`implicated in the down-regulation of intestinal CYP450
`remains to be defined.
`
`4. RENAL DRUG METABOLISM
`
`Enzymes involved in the metabolism of drugs are also
`present in the kidneys [13, 72, 84]. Several CYP450
`isoforms have been identified in the kidney from studies of
`enzyme activities and also by molecular biology techniques
`[84]. Although the ability of these isoforms to metabolize
`xenobiotics has been confirmed in vitro [84], the role of
`renal CYP450 in the systemic metabolism of drugs has not
`been documented. However, a reduction in functional renal
`mass, as seen in CRF, should theoretically lead to a decrease
`in the metabolic capacity of the kidney mediated by CYP450
`isoforms.
`
`Enzymes involved in phase II reactions are also present in
`the kidney, particularly those implicated in conjugation
`reactions, such as UDP-glucuronyl-transferases, glutathione
`S-transferases, and N-sulphotransferases [13, 72, 84]. Some
`of these enzymes, particularly UDP-glucuronyl-transferases,
`have a high activity in the kidney, predominantly in the
`proximal tubule [72]. Interestingly, such a location allows
`the kidney to be exposed to the greatest quantity of drugs
`since the proximal tubule is subjected to the bulk of
`glomerular ultrafiltrate and also the highest concentration of
`drugs on the contraluminal side. In vitro studies using either
`isolated perfused
`tubules or perfused kidneys have
`demonstrated that the kidney has the ability to biotransform
`several drugs: paracetamol, morphine,
`sulindac, and
`furosemide. However, few in vivo studies have been
`performed to compare the ability of the kidney to metabolize
`xenobiotics with that of other organs, e.g. the liver or the
`intestine [85]. Furthermore, the contribution of the kidney to
`the systemic metabolism has been poorly documented [85].
`
`The contribution of the kidney to the metabolism of
`furosemide, was studied

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