10
TOXICOLOGICAL SCIENCES 106(1), 64–73 (2008) doi:10.1093/toxsci/kfn163 Advance Access publication August 21, 2008 Buprenorphine Alters Desmethylflunitrazepam Disposition and Flunitrazepam Toxicity in Rats Ste ´phane Pirnay,* , Bruno Megarbane,* , ,1 Xavier Decle `ves,* Patricia Rise `de,* Stephen W. Borron,§ Ste ´phane Bouchonnet,{ Be ´range `re Perrin,Marcel Debray,* , k Nathalie Milan,Tania Duarte,* Ivan Ricordel,and Fre ´de ´ric J. Baud* , *INSERM U705, CNRS, UMR7157, Universite ´ Paris-Descartes, and Universite ´ Paris-Diderot, Paris F-75018, France; Laboratoire de Toxicologie de l’Institut National de Police Scientifique, Paris F-75012, France; Re ´animation Me ´dicale et Toxicologique, Ho ˆ pital Lariboisie `re, Paris F-75010, France; §Department of Surgery, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7736, San Antonio, TX 78229, USA; {DCMR, Ecole Polytechnique, Route de Saclay, 91128 Palaiseau, France; and kLaboratoire de Biomathe ´matiques, Universite ´ Paris-Descartes, 75006 Paris, France Received June 5, 2008; accepted August 4, 2008 High-dosage buprenorphine (BUP) consumed concomitantly with benzodiazepines (BZDs) including flunitrazepam (FZ) may cause life-threatening respiratory depression despite a BUP ceiling effect and BZDs’ limited effects on ventilation. However, the mechanism of BUP/FZ interaction remains unknown. We hypoth- esized that BUP may alter the disposition of FZ active metabolites in vivo, contributing to respiratory toxicity. Plasma FZ, desmethyl- flunitrazepam (DMFZ), and 7-aminoflunitrazepam (7-AFZ) con- centrations were measured using gas chromatography–mass spectrometry. Intravenous BUP 30 mg/kg pretreatment did not alter plasma FZ and 7-AFZ kinetics in Sprague-Dawley rats infused with 40 mg/kg FZ over 30 min, whereas resulting in a three-fold increase in the area under the curve (AUC) of DMFZ concentrations compared with control (p < 0.01). In contrast, BUP did not significantly modify plasma DMFZ concentrations after intravenous infusion of 7 mg/kg DMFZ, whereas resulting in a similar peak concentration to that generated from 40 mg/kg FZ administration. Regarding the effects on ventilation, BUP (30 mg/kg) as well as its combination with FZ (0.3 mg/kg) significantly increased PaCO 2 , whereas only BUP/FZ combination decreased PaO 2 (p < 0.001). Interestingly, FZ (40 mg/kg) but not DMFZ (40 mg/kg) significantly increased PaCO 2 (p < 0.05), whereas DMFZ but not FZ decreased PaO 2 (p < 0.05). Thus, decrease in PaO 2 appears related to BUP- mediated effects on DMFZ disposition, although increases in PaCO 2 relate to direct BUP/FZ additive or synergistic dynamic interactions. We conclude that combined high-dosage BUP and FZ is responsible for increased respiratory toxicity in which BUP-mediated alteration in DMFZ disposition may play a significant role. Key Words: buprenorphine; desmethylflunitrazepam; flunitrazepam; interaction; metabolism; pharmacokinetics. Buprenorphine (BUP), an hemisynthetic opioid, exerts agonist properties on l opioid-receptors and antagonist properties on j opioid-receptors, with an elevated affinity for both types of receptors (Cowan, 1995, 2003). BUP demon- strates a very slow dissociation from opioid-receptors resulting in a long duration of action. Interestingly, in contrast with morphine and methadone (Cowan et al., 1977; McCormick et al., 1984), a plateau of respiratory depressive effects has been consistently reported both in animals and humans (Cowan et al., 1977; Dahan et al., 2005, 2006; Nielsen and Taylor, 2005; Walsh et al., 1994). Due to its duration of action and ceiling effects supporting its safety, high-dosage BUP has been marketed as a maintenance therapy for heroin addiction. Consequently, a reduction in lethal overdoses and improvement in patients’ social function- ing has been observed (Emmanuelli and Desenclos, 2005). However, forensic studies have reported several fatalities involving BUP (Kintz, 2001; Pirnay et al., 2004a; Reynaud et al., 1998; Tracqui et al., 1998). The observed asphyxic deaths were attributed to either BUP misuse consisting of i.v. self-injection of crushed tablets or concomitant intake of psychotropic drugs including benzodiazepines (BZDs) (Kintz, 2001; Lai et al., 2006; Pirnay et al., 2004a; Reynaud et al., 1998; Tracqui et al., 1998). Consistent with the latter, BZDs have been detected in 91 among 117 fatalities with BUP detection in body fluids reported in France (Kintz, 2001), suggesting a major role for BZDs in the occurrence of BUP- induced respiratory toxicity. Widespread prescription of BZDs has been attributed to their wide therapeutic index and minimal adverse side effects. However, these drugs have been subject to increasing misuse and abuse (Garretty et al., 1997; Hojer et al., 1989; Verwey et al., 2000). While deaths involving BZD alone appear uncommon in the absence of underlying pathology (Buckley and McManus, 2004; Drummer et al., 1993), BUP/BZD deleterious interactions were hypothesized in BUP-associated fatalities. The likelihood of such a drug-drug interaction was further supported by blood BUP concentrations within the therapeutic range in the majority of deaths (Kintz, 2001; Lai et al., 2006; Pirnay et al., 2004a; Tracqui et al., 1998). Although rarely detected due to rapid in vitro degradation, 1 To whom correspondence should be addressed at Re ´animation Me ´dicale et Toxicologique, Ho ˆ pital Lariboisie `re, 2 Rue Ambroise Pare ´, 75010 Paris, France. E-mail: [email protected]. Ó The Author 2008. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For permissions, please email: [email protected] by guest on June 3, 2013 http://toxsci.oxfordjournals.org/ Downloaded from

Buprenorphine Alters Desmethylflunitrazepam Disposition and Flunitrazepam Toxicity in Rats

Embed Size (px)

Citation preview

TOXICOLOGICAL SCIENCES 106(1), 64–73 (2008)

doi:10.1093/toxsci/kfn163

Advance Access publication August 21, 2008

Buprenorphine Alters Desmethylflunitrazepam Dispositionand Flunitrazepam Toxicity in Rats

Stephane Pirnay,*,† Bruno Megarbane,*,‡,1 Xavier Decleves,* Patricia Risede,* Stephen W. Borron,§ Stephane Bouchonnet,{Berangere Perrin,† Marcel Debray,*,k Nathalie Milan,† Tania Duarte,* Ivan Ricordel,† and Frederic J. Baud*,‡

*INSERM U705, CNRS, UMR7157, Universite Paris-Descartes, and Universite Paris-Diderot, Paris F-75018, France; †Laboratoire de Toxicologie de l’Institut

National de Police Scientifique, Paris F-75012, France; ‡Reanimation Medicale et Toxicologique, Hopital Lariboisiere, Paris F-75010, France; §Department of

Surgery, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7736, San Antonio, TX 78229, USA; {DCMR, EcolePolytechnique, Route de Saclay, 91128 Palaiseau, France; and kLaboratoire de Biomathematiques, Universite Paris-Descartes, 75006 Paris, France

Received June 5, 2008; accepted August 4, 2008

High-dosage buprenorphine (BUP) consumed concomitantly

with benzodiazepines (BZDs) including flunitrazepam (FZ) may

cause life-threatening respiratory depression despite a BUP ceiling

effect and BZDs’ limited effects on ventilation. However, the

mechanism of BUP/FZ interaction remains unknown. We hypoth-

esized that BUP may alter the disposition of FZ active metabolites

in vivo, contributing to respiratory toxicity. Plasma FZ, desmethyl-

flunitrazepam (DMFZ), and 7-aminoflunitrazepam (7-AFZ) con-

centrations were measured using gas chromatography–mass

spectrometry. Intravenous BUP 30 mg/kg pretreatment did not alter

plasma FZ and 7-AFZ kinetics in Sprague-Dawley rats infused with

40 mg/kg FZ over 30 min, whereas resulting in a three-fold increase

in the area under the curve (AUC) of DMFZ concentrations

compared with control (p < 0.01). In contrast, BUP did not

significantly modify plasma DMFZ concentrations after intravenous

infusion of 7 mg/kg DMFZ, whereas resulting in a similar peak

concentration to that generated from 40 mg/kg FZ administration.

Regarding the effects on ventilation, BUP (30 mg/kg) as well as its

combination with FZ (0.3 mg/kg) significantly increased PaCO2,

whereas only BUP/FZ combination decreased PaO2 (p < 0.001).

Interestingly, FZ (40 mg/kg) but not DMFZ (40 mg/kg) significantly

increased PaCO2 (p < 0.05), whereas DMFZ but not FZ decreased

PaO2 (p < 0.05). Thus, decrease in PaO2 appears related to BUP-

mediated effects on DMFZ disposition, although increases in PaCO2

relate to direct BUP/FZ additive or synergistic dynamic interactions.

We conclude that combined high-dosage BUP and FZ is responsible

for increased respiratory toxicity in which BUP-mediated alteration

in DMFZ disposition may play a significant role.

Key Words: buprenorphine; desmethylflunitrazepam;

flunitrazepam; interaction; metabolism; pharmacokinetics.

Buprenorphine (BUP), an hemisynthetic opioid, exerts

agonist properties on l opioid-receptors and antagonist

properties on j opioid-receptors, with an elevated affinity for

both types of receptors (Cowan, 1995, 2003). BUP demon-

strates a very slow dissociation from opioid-receptors resulting

in a long duration of action. Interestingly, in contrast with

morphine and methadone (Cowan et al., 1977; McCormick

et al., 1984), a plateau of respiratory depressive effects has

been consistently reported both in animals and humans (Cowan

et al., 1977; Dahan et al., 2005, 2006; Nielsen and Taylor,

2005; Walsh et al., 1994).

Due to its duration of action and ceiling effects supporting its

safety, high-dosage BUP has been marketed as a maintenance

therapy for heroin addiction. Consequently, a reduction in

lethal overdoses and improvement in patients’ social function-

ing has been observed (Emmanuelli and Desenclos, 2005).

However, forensic studies have reported several fatalities

involving BUP (Kintz, 2001; Pirnay et al., 2004a; Reynaud

et al., 1998; Tracqui et al., 1998). The observed asphyxic

deaths were attributed to either BUP misuse consisting of i.v.

self-injection of crushed tablets or concomitant intake of

psychotropic drugs including benzodiazepines (BZDs) (Kintz,

2001; Lai et al., 2006; Pirnay et al., 2004a; Reynaud et al.,1998; Tracqui et al., 1998). Consistent with the latter, BZDs

have been detected in 91 among 117 fatalities with BUP

detection in body fluids reported in France (Kintz, 2001),

suggesting a major role for BZDs in the occurrence of BUP-

induced respiratory toxicity.

Widespread prescription of BZDs has been attributed to their

wide therapeutic index and minimal adverse side effects.

However, these drugs have been subject to increasing misuse

and abuse (Garretty et al., 1997; Hojer et al., 1989; Verwey

et al., 2000). While deaths involving BZD alone appear

uncommon in the absence of underlying pathology (Buckley

and McManus, 2004; Drummer et al., 1993), BUP/BZD

deleterious interactions were hypothesized in BUP-associated

fatalities. The likelihood of such a drug-drug interaction was

further supported by blood BUP concentrations within the

therapeutic range in the majority of deaths (Kintz, 2001; Lai

et al., 2006; Pirnay et al., 2004a; Tracqui et al., 1998).

Although rarely detected due to rapid in vitro degradation,

1 To whom correspondence should be addressed at Reanimation Medicale et

Toxicologique, Hopital Lariboisiere, 2 Rue Ambroise Pare, 75010 Paris,

France. E-mail: [email protected].

� The Author 2008. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved.For permissions, please email: [email protected]

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

Flunitrazepam (FZ) was suspected to be involved in a large

number of both BUP nonfatal (Gueye et al., 2002b) as well as

fatal intoxications (Druid et al., 2001; Kintz, 2001; Pirnay

et al., 2004a). However, the mechanism of BUP/FZ toxicity

including any interaction regarding FZ disposition has not been

worked out in humans.

In rats, we previously showed that the combination of 40

mg/kg FZ and 30 mg/kg BUP induced the rapid onset of

respiratory depression (Megarbane et al., 2005a). However, the

mechanism of BUP/FZ interaction remained poorly under-

stood. We demonstrated that rat pretreatment with FZ altered

neither plasma nor striatal BUP kinetics (Megarbane et al.,2005a). Moreover, in vitro studies using rat as well as human

liver microsomes predicted no significant metabolic BUP/FZ

interaction in vivo (Ibrahim et al., 2000; Kilicarslan and Sellers,

2000; Umehara et al., 2002). However, to our knowledge, BUP

effects on FZ pharmacokinetics have not been previously

addressed in vivo. Because many BZDs have active metabo-

lites, we hypothesized that BUP may alter in vivo the

disposition of active FZ metabolites, thus increasing FZ

toxicity. Therefore, we studied in the rat the respiratory effects

of a BUP/FZ combination, investigating BUP effects on the

kinetics of FZ and its main active metabolites, desmethyl-

flunitrazepam (DMFZ), and 7-aminoflunitrazepam (7-AFZ).

We choose to study elevated doses of both drugs in rats in

order to mimic clinical exposures in drug addicts, as reported in

severe or fatal poisonings (Kintz, 2001; Pirnay et al., 2004a)

and help appropriate extrapolation from rats to humans.

MATERIALS AND METHODS

All experiments were carried out within the ethical guidelines established by

the National Institute of Health and the French Minister of Agriculture.

Drugs

BUP was generously supplied by Schering-Plough, SA. It was subsequently

dissolved in a mixture of sterile water (4.7 ml), absolute ethanol (400 ll), and

hydrochloric acid 0.1 N (300 ll) at a concentration of 18.5 mg/ml. d4-BUP was

purchased from Radian Inc. FZ was generously provided by Hoffman-La

Roche, Inc. A FZ solution was prepared in propylene glycol at a concentration

of 10 mg/ml. Both FZ metabolites, DMFZ and 7-AFZ, as well as d4-DMFZ

were purchased from Radian Inc. A mixture of 99% N,O-bis(trimethylsilyl)-

trifluoroacetamide (BSTFA) with 1% trimethylchlorosilane (TMCS) was

purchased from Sigma Chemical Co.

Animals

Male Sprague-Dawley rats (Iffa-Credo, France) weighing 250–300 g at the

time of experimentation were used. They were housed for 8 days before

experimentation in a temperature- and light-controlled animal care unit. They

were allowed food and water ad libitum until one day prior to experimentation.

The day before the study, animals were anesthetized with i.p. 70 mg/kg

ketamine (Ketalar, Parke Davis) and 10 mg/kg xylazine (Rompun, Bayer), then

placed on a warming blanket with a regulating thermostat. A rectal probe

permitted feedback temperature control. The femoral vein and artery were

catheterized with 30-cm-long silastic tubes (Dow Corning Co, Midland, MI)

with external and internal diameters of 0.94 and 0.51 mm, respectively (Gueye

et al., 2001, 2002a). Arterial and venous catheters were then tunneled

subcutaneously and fixed at the back of the neck. Heparinized saline was

injected into each catheter to avoid thrombosis and catheter obstruction. Rats

were then returned to their individual cages for a minimum 24-h recovery

period to allow complete washout of anesthesia. On the experiment day, rats

were placed in a restraining chamber (Harvard Apparatus). Drugs were

administered into the venous catheter via an infusion pump (Harvard

Instruments-PHD 2000). The final injected volumes were constant and adjusted

with 0.9% NaCl to 1.3 ml. The arterial catheter allowed blood sampling for the

kinetic and arterial blood-gases studies. Blood samples were obtained in

restrained rats and collected in heparinized syringes.

Gas Chromatography–Mass Spectrometry

A general gas chromatography–mass spectrometry (GC-MS) method for

detection and quantification of FZ, BUP, and their main metabolites was

validated according to internationally accepted criteria (Shah et al., 2000). The

method was set up and adapted for analysis of small rat plasma samples as

previously reported (Pirnay et al., 2004b). Extraction involved a clean-up

procedure using liquid-liquid Toxi-tubes A cartridges followed by derivatiza-

tion with N,O-bis-(trimethylsilyl)trifluoroacetamide, according to previously

reported methods (Libong et al., 2003; Pirnay et al., 2002). The percentages of

recovery were 71, 67, 51, and 81%, for BUP, FZ, 7-AFZ, and DMFZ,

respectively, with coefficient of variation (CV) ranging from 5.4 to 13.9%.

They were concentration independent.

All analyses were performed on a Hewlett Packard 5973 apparatus including

a gas chromatograph coupled with a quadruple mass spectrometer and fitted

with an autosampler. Chromatographic separation was carried out on a HP-5

MS capillary column (30 m 3 0.25 mm I.D., 0.25 lm film thickness). All

chromatographic and spectrometric parameters were established according to

a previous reported validation (Pirnay et al., 2004b). Detection was operated

under selected ion monitoring mode. The ions: m/z 285.1 for FZ, m/z 355.2 for

7-AFZ, m/z 370.1 for DMFZ, m/z 450.2 for BUP, m/z 375.1 for DMFZ-d4, m/z454.3 for BUP-d4 were the most abundant and used for quantification (Pirnay

et al., 2004b).

Limits of Quantification. The limits of quantification in plasma were 25 ng/

ml for 7-AFZ and 125 ng/ml for DMFZ, FZ, and BUP. The CVs varied between

3.85 (BUP) and 13.45% (FZ); accuracies between 10.09 (BUP) and 14.54% (7-

AFZ) (Pirnay et al., 2004b).

Linearity Range. Linearity was found in plasma between 125 and 25,000

ng/ml for BUP, 125 and 12,500 ng/ml for DMFZ, 125 and 5000 ng/ml for FZ,

and between 25 and 50,000 ng/ml for 7-AFZ. Coefficients of correlation (R2)

were 0.999 for DMFZ, FZ, and BUP and 0.984 for 7-AFZ (Pirnay et al.,

2004b). For all plasma concentrations above the upper limit of linearity range,

plasma was diluted as required before quantification.

Intra- and Interassay Precision and Accuracy. Good reproducibility

(intra-assay CV ¼ 0.32–11.69%; interassay CV ¼ 0.63–9.55%) and accuracy

(intra-assay error ¼ 2.58–12.73%; interassay error ¼ 0.83–11.07%) were

obtained (Pirnay et al., 2004b).

Sample Preparation. Eighty microliters of DMFZ-d4 and 80 ll of BUP-

d4 at 1 lg/ml in acetonitrile and methanol, respectively, were added to 40 ll of

plasma. The final volume was adjusted to 1 ml by addition of deionized water.

The mixture was vortexed and transferred into a toxi-tube A (Ansys

Diagnostics, Inc.) which had been previously conditioned by 2 ml of deionized

water. The tube was mechanically agitated before centrifugation for 5 min at

3000 rpm. The organic phase was transferred into a clean tube and evaporated

to dryness under a nitrogen stream at 25�C. After complete evaporation of the

solvent, the residue was heated at 80�C for 5 min in order to eliminate any trace

of water. Forty microliters of BSTFA/TMCS (99/1%) were added to the residue

for trimethylsilylation. The sample was then heated at 80�C for 20 min and

cooled down to an ambient temperature prior to GC-MS analysis.

To separate free from bound plasma proteins by ultrafiltration, 200 ll of

plasma were added on a Centifree Micropartition Device (Millipore, Inc), with

a cut of 30,000 Da. The cartridges were centrifuged for 30 min at 2000 rpm.

BUPRENORPHINE ALTERS FLUNITRAZEPAM TOXICITY 65

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

Eighty microliters of d4-DMFZ and 80 ll of d4-BUP at 1 lg/ml in acetonitrile

and methanol, respectively, were added to 40 ll of the ultrafiltrate. The mixture

was vortexed and transferred into a toxi-tube A (Ansys Diagnostics, Inc.) which

had been previously conditioned with 2 ml of deionized water. The same

protocol as described above was used for the extraction and silylation prior to

GC-MS analysis.

Study Designs and Dosage Justification

Study 1. Study of plasma FZ kinetics at four dosages (3, 10, 30, and 40

mg/kg) in order to test its linearity. Catheterized rats were randomized into

four groups of five animals. Rats received i.v. infusion of either 3, 10, 30, or

40 mg/kg FZ over 30 min. Plasma FZ kinetics were studied over 240 min. One-

hundred microliters of blood samples was collected at �30, �25, �20, �15,

�10, and at 0, 5, 10, 20, 60, 120, 180, and 240 min following FZ injection.

Study 2. Effect of 30 mg/kg BUP pretreatment on the time course of

plasma FZ, DMFZ, and 7-AFZ concentrations after 40 mg/kg FZ

infusion. Dosages were chosen in accordance with those previously used to

describe the respiratory depressive effects of BUP/FZ combinations

(Megarbane et al., 2005a). These high doses of both molecules were reported

to be deprived of significant effects on rat arterial blood gases in comparison to

solvent. Catheterized rats were randomized into two groups of five animals.

Rats in the control group received i.v. infusion of BUP solvent over 3 min

followed by 40 mg/kg FZ i.v. over 30 min. Rats in the study group received

30 mg/kg BUP over 3 min followed by 40 mg/kg FZ i.v. over 30 min. Plasma

kinetics of FZ and its metabolites DMFZ and 7-AFZ were studied over

180 min. One-hundred microliters of blood samples was collected before and

after BUP (or solvent) infusion at �33 and �30 min, and during FZ infusion

at �25, �20, �15, �10, and at 0, 5, 10, 20, 60, 120, and 180 min following

FZ injection.

Study 3. Effect of 40 mg/kg FZ on the time course of plasma BUP

concentrations after 30 mg/kg BUP administration. Catheterized rats were

randomized into two groups of five animals. Rats in the control group received

i.v. infusion of 30 mg/kg BUP over 3 min followed by FZ solvent i.v. over

30 min. Rats in the study group received 30 mg/kg BUP over 3 min followed

by 40 mg/kg FZ i.v. over 30 min. Plasma kinetics of BUP were studied over

180 min. One-hundred microliters of blood samples was collected before and

after BUP infusion at �33 and �30 min, and during FZ (or solvent) infusion

at �25, �20, �15, �10, and at 0, 5, 10, 20, 60, 120, and 180 min following

FZ (or solvent) injection.

Study 4. Effect of 30 mg/kg BUP pretreatment on the time course of

plasma DMFZ after 7 mg/kg DMFZ infusion. The DMFZ dosage was

chosen as it was estimated to give a similar peak concentration (Cmax) to the

one observed with 40 mg/kg FZ. Catheterized rats were randomized into two

groups of five animals. Rats in the control group received i.v. infusion of BUP

solvent over 3 min followed by 7 mg/kg DMFZ i.v. over 30 min. Rats in the

study group received 30 mg/kg BUP over 3 min followed by 7 mg/kg DMFZ

i.v. over 30 min. Plasma kinetics of total DMFZ were studied over 240 min.

One-hundred microliters of blood samples was collected before and after

BUP (or solvent) infusion at �33 and �30 min, and during DMFZ infusion

at �25, �20, �15, �10, and at 0, 5, 10, 20, 30, 60, 120, 180, and 240 min

following DMFZ injection.

Study 5. Effect of 30 mg/kg BUP pretreatment on the total and free

plasma concentrations of FZ and DMFZ. Catheterized rats were random-

ized into two groups of five animals. Rats in the control group received i.v.

infusion of BUP solvent over 3 min followed by 40 mg/kg FZ i.v. over 30 min.

Rats in the study group received 30 mg/kg BUP over 3 min followed by

40 mg/kg FZ i.v. over 30 min. Plasma concentrations of total FZ and DMFZ as

well as free FZ and DMFZ were studied during FZ infusion at �20 min and at

the end of FZ infusion. Five-hundred microliters of blood samples was

collected at each sampling point.

Study 6. Effect of 40 mg/kg FZ and 40 mg/kg DMFZ on arterial blood

gases. We chose to test elevated dosages of both FZ and DMF in order to

maximize their potential effects on ventilation. We used 40 mg/kg FZ, based on

previous data showing that this dose administered i.p. induced a long-lasting

coma without mortality (Borron et al., 2002). Catheterized rats were

randomized into three groups of four animals. Rats in the control group

received BUP i.v. solvent over 3 min, followed by FZ solvent i.v. over 30 min.

Rats in the FZ group received BUP solvent i.v. over 3 min followed by 40 mg/

kg FZ i.v. over 30 min. Rats in the DMFZ group received BUP solvent i.v. over

3 min, followed by 40 mg/kg DMFZ i.v. over 30 min. Three-hundred

microliters of arterial blood samples was collected at 5, 20, 40, 60, 90, 120, and

180 min after FZ, DMFZ or solvent infusion. Blood gases were immediately

measured using a Rapid lab 248 (Bayer corporation, East Walpole, MA) blood-

gas analyzer.

Study 7: Effect of 30 mg/kg BUP, 0.3 mg/kg FZ, and their combination on

arterial blood gases. We chose a pharmacological range FZ dosage in the

BUP/FZ combination in order to better substantiate the effects of kinetic drug-

drug interaction. We chose 0.3 mg/kg FZ as previously shown to represent the

median effective dose to significantly modify rat behavior in continuous

conditioned avoidance tests without incapacitation (Randall and Kappell, 1973;

Zbinden and Randall, 1967). Nevertheless, pharmacokinetics of FZ were not

determined at this dosage, because plasma concentrations were anticipated to

be lower than the limit of quantification of our GC-MS method. Catheterized

rats were randomized into four groups of seven animals. Rats in the control

group received BUP solvent i.v. over 3 min, followed by FZ solvent i.v. over

30 min. Rats in the BUP group received 30 mg/kg BUP i.v. over 3 min

followed by FZ solvent i.v. over 30 min. Rats in the FZ group received BUP

solvent i.v. over 3 min followed by 0.3 mg/kg FZ i.v. over 30 min. Rats in the

BUP/FZ combination group received 30 mg/kg BUP i.v. over 3 min followed

by 0.3 mg/kg FZ i.v. over 30 min. Three-hundred microliters of blood samples

was collected at 5, 20, 40, 60, 90, 120, and 180 min after FZ or solvent

infusion. Blood gases were immediately determined.

Pharmacokinetic Analysis

Noncompartmental analysis (WinNonlin v 4.1, Pharsight Corp, Mountain

View, CA) was used to calculate the Cmax, the terminal elimination half-lives in

plasma, the AUC from 0 to infinity (AUC0/N), the total plasma clearance

TABLE 1

Plasma Kinetic Parameters of FZ at Different Dosages i.v. Infused over 30 min in the Rat

Dose (mg/kg) Half-life (min) Cmax (ng/ml) AUC0/N (ng/min/ml) Clt (ml/min/kg) Vss (ml/kg)

3 44 ± 11 2771 ± 980 66,362 ± 15,994 48 ± 12 1370 ± 288

10 36 ± 14 6366 ± 1604 238,916 ± 77,750 51 ± 21 1444 ± 540

30 70 ± 17 20,163 ± 1980 799,860 ± 49,144 38 ± 3 1574 ± 144

40 50 ± 3 28,091 ± 3676 1,137,756 ± 197,847 40 ± 6 1697 ± 295

Note. Values represent mean ± SEM.

66 PIRNAY ET AL.

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

(Clt), and the volume of distribution at the steady state (VSS). The mean Cmax

was determined as the mean of the peak concentrations observed in each rat. To

test the linearity of plasma FZ kinetics, AUC0/infinity/dosage ratios were

calculated for each dosage and each rat.

Statistical Analysis

Results are presented as mean ± SEM. Regarding kinetics, data were

compared between groups with Mann-Whitney tests. To test the linearity of FZ

kinetics, mean AUC0/N/dosage values were compared between each dosage

using one-way ANOVA. Regarding arterial blood gases, baseline values were

compared using one-way ANOVA, followed by multiple comparison tests

using Bonferroni correction. To study the time-course effect of each drug on

blood gases, we calculated the AUC0/93 using the trapezoid method between

the values obtained before injection and at 93 min after injection. Comparisons

of AUC0/93 were performed using one-way ANOVA followed by multiple

comparison tests using Bonferroni correction. All tests were two-tailed and

performed using Prism version 3.0 (GraphPad Software, Inc, San Diego, CA).

p values of less than 0.05 were considered significant.

RESULTS

Pharmacokinetic Studies (Studies 1–5)

We first demonstrated that FZ kinetics were linear in the

3–40 mg/kg dosage range as AUC/dosage did not significantly

differ with respect to FZ dosage (Study 1, Table 1). This study

showed that FZ kinetics were of first-order in this range,

allowing an adequate assessment of BUP/FZ drug-drug

interaction.

Then, we studied 40 mg/kg BUP effects on the time course

of FZ, DMFZ, and 7-AFZ concentrations in plasma after

30 mg/kg i.v. FZ administration (Study 2, Fig. 1). None of the

parameters of plasma FZ kinetics, including the Cmax, the peak

time (Tmax), the AUC0/infinity, the Clt, and the VSS were

significantly altered by BUP pretreatment (Fig. 1A; Table 2A).

By contrast, significant differences were observed in the time

course of plasma DMFZ concentrations (Fig. 1B; Table 2B).

DMFZ Cmax was 5592 ± 1312 ng/ml in the BUP þ FZ group

versus 2708 ± 588 ng/ml in the FZ group (p ¼ 0.03). Similarly,

the AUC0/infinity values were significantly different (p ¼0.009), whereas semilog representation of the terminal phases

of DMFZ and FZ kinetics showed parallel apparent declines

irrespective of BUP pretreatment (Fig. 2). In contrast to DMFZ,

no significant alteration was observed in the time course of

7-AFZ concentrations in plasma following BUP pretreatment

(Fig. 1C; Table 2B). In addition, FZ infusion did not alter BUP

kinetics in plasma (Study 3, Fig. 3).

FIG. 1. Kinetics of FZ (A), DMFZ (B), and 7-AFZ (C) in plasma after rat

treatment with either BUP solvent þ 40 mg/kg FZ (open squares) or 30 mg/kg

BUP þ 40 mg/kg FZ (filled squares). In each group, five rats were used. Values

represent mean ± SEM at each time. Significant differences were shown using

Mann-Whitney tests between both DMFZ kinetics (B) for time �20, �15, �10,

þ20, and þ120 min (*p < 0.05). In contrast, no significant differences were

shown between both FZ (A) and 7-AFZ (C) kinetics.

BUPRENORPHINE ALTERS FLUNITRAZEPAM TOXICITY 67

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

To test the hypothesis of a direct interaction of BUP on

DMFZ distribution in plasma, we studied the effects of 40 mg/kg

BUP i.v. on DMFZ kinetics after 7 mg/kg DMFZ infusion

(Study 4). The Cmax generated by 7 mg/kg DMFZ (2024 ± 204

ng/ml) was similar to that observed when 30 mg/kg FZ was

given (2708 ± 588 ng/ml). The time course of DMFZ

concentrations in plasma as well as the various parameters of

plasma DMFZ kinetics were not significantly altered by BUP

pretreatment (Fig. 4, Table 3). Semilog representation of the

terminal phases of DMFZ kinetics showed parallel apparent

declines whether DMFZ was directly administered or resulted

from FZ administration (Fig. 2). DMFZ elimination half-lives

were not significantly different in either situations, that is, in the

absence (62 ± 14 vs. 41 ± 7 min) or presence of BUP (56 ± 9 vs.

64 ± 22 min). In addition, to test the hypothesis of BUP effect on

FZ or DMFZ protein binding in plasma, we measured the free/

total ratios of FZ and DMFZ in rats treated with 40 mg/kg FZ i.v.

(Study 5). There was no significant alteration of these ratios in

relation to 30 mg/kg BUP i.v. pretreatment (Table 4).

Pharmacodynamic Studies (Studies 6 and 7)

We studied the effects on arterial blood gases of 40 mg/kg

FZ and 40 mg/kg DMFZ in comparison to FZ solvent (Study

6). Arterial pH did not significantly differ between the three

groups (Fig. 5A). PaCO2 was significantly more elevated in the

FZ than in DMFZ or solvent treated rats (p ¼ 0.03, Fig. 5B).

TABLE 2

Plasma Kinetic Parameters of FZ (A) and its Metabolites DMFZ and 7-AFZ (B) in Rats Treated with either 30 mg/kg BUP 1 40

mg/kg FZ (BUP 1 FZ Group) or BUP Solvent 1 40 mg/kg FZ (FZ Group)

(A) FZ Half-life (min) Cmax (ng/ml) AUC0/N (ng/min/ml) Clt (ml/min/kg) Vss (ml/kg)

FZ group 48 ± 6 26,188 ± 6117 1,053,796 ± 327,025 43 ± 10 1802 ± 496

BUP þ FZ group 45 ± 6 34,824 ± 13,739 1,279,224 ± 517,192 37 ± 13 1539 ± 824

FIG. 2. Log y axis scale representation of the time courses of plasma FZ

(open circles) and DMFZ (open squares) concentrations after rat treatment with

either BUP solvent þ 40 mg/kg FZ (A) or 30 mg/kg BUP þ 40 mg/kg FZ (B).

Time courses of plasma DMFZ (filled squares) after treatment with either BUP

solvent þ 7 mg/kg DMFZ (A) or 30 mg/kg BUP þ 40 mg/kg DMFZ (B) were

added. In each group, five rats were used. Values represent mean ± SEM at

each time.

FIG. 3. Kinetics of BUP in plasma after rat treatment with either 30 mg/kg

BUP þ 40 mg/kg FZ (filled squares) or 30 mg/kg BUP þ FZ solvent (open

squares). In each group, five rats were used. Values represent mean ± SEM at

each time. No significant differences were shown using Mann-Whitney tests.

68 PIRNAY ET AL.

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

Interestingly, there was no significant difference in PaO2

between the FZ and solvent treated rats, whereas PaO2 was

significantly lower with DMFZ treatment in comparison to the

solvent (p ¼ 0.03, Fig. 5C). The lowest PaO2 value was

observed in the DMFZ group at 20 min and significantly differed

from the solvent (9.36 ± 0.23 vs. 12.58 ± 0.28 kPa, p ¼ 0.03).

Then, we investigated the effects on arterial blood gases of

30 mg/kg BUP þ 0.3 mg/kg FZ combination (Study 7). There

were no significant differences regarding arterial pH (Fig. 6A).

Interestingly, 0.3 mg/kg FZ did not have any significant effect

on PaCO2 in comparison to the solvents (Fig. 6B). In contrast,

30 mg/kg BUP as well as the combination of 30 mg/kg BUP þ0.3 mg/kg FZ significantly increased PaCO2 in comparison

with the solvents (p ¼ 0.03 and 0.001, respectively). The

highest PaCO2 value was observed in the combination group at

the end of FZ infusion and significantly differed from the

solvents (7.17 ± 0.14 vs. 5.56 ± 0.19 kPa, p ¼ 0.001).

PaO2 did not significantly differ between 30 mg/kg BUP, 0.3

mg/kg FZ, and the solvent groups, whereas PaO2 was

significantly lower in the BUP/FZ combination group (p ¼0.002) (Fig. 6C). The lowest PaO2 value was observed in the

combination group at 20 min and significantly differed from

the solvents (8.68 ± 0.29 vs. 10.38 ± 0.45 kPa, p ¼ 0.004).

DISCUSSION

In this study, we investigated the respiratory effects of a BUP/

FZ combination and demonstrated the existence of a significant

BUP-mediated alteration of DMFZ disposition that may have

contributed to increase FZ toxicity. To our knowledge, we report

here for the first time plasma kinetics of FZ and its major

metabolites in rat at a toxic dosage (40 mg/kg). Previous studies

described kinetics after intravenous administration of 1 mg/kg

(Becherucci et al., 1985) and 2.5 mg/kg (Mandema et al., 1991)

FZ. We demonstrated that FZ kinetics were linear in the 3–40

mg/kg dosage range obviating any saturation in distribution or

elimination processes. We found comparable kinetic parameters

with those published at 2.5 mg/kg (half-life: 76 ± 10 min; Vss:

3800 ± 300 ml/kg; Clt: 80 ± 5 ml/min/kg) (Mandema et al.,1991). Thus, our experimental conditions were considered as

favorable to allow pertinent analysis of FZ pharmacokinetic/

pharmacodynamic relationships.

We first studied the respiratory effects of an elevated BUP

dosage (30 mg/kg). As previously demonstrated (Gueye et al.,2001, 2002a; Ohtani et al., 1997), we found only a mild and

transient effect on PaCO2 with no significant consequence on pH

or PaO2 in comparison with the solvent. Similarly, we showed

that 40 but not 0.3 mg/kg FZ increased PaCO2, whereas neither

dosage resulted in significant changes in arterial pH and PaO2.

BZD effects on ventilation are known to be rather limited.

Consistently, a massive dose of i.p. midalozam (160 mg/kg)

reproducibly induced deep coma and moderate respiratory

acidosis, without significant change in PaO2 (Gueye et al.,2002a; Megarbane et al., 2005b). Moreover, even an increase in

PaO2 has been reported following i.p. administration of 20 mg/kg

diazepam (McCormick et al., 1984) or subcutaneous adminis-

tration of 10 mg/kg chlordiazepoxide (Verborgh et al., 1998).

In stark contrast to the limited effects on resting ventilation

resulting from separate administration of BUP or BZDs, the

combination of both drugs has been reported to induce

significant toxicity. Pretreatment with 40 mg/kg FZ induced

a sixfold decrease in the BUP median lethal dose and

significantly prolonged time to death in BUP-treated animals

(Borron et al., 2002). In this study, BUP/FZ combination

FIG. 4. Kinetics of DMFZ in plasma after rat treatment with either BUP

solvent þ 7 mg/kg DMFZ (open squares) or 30 mg/kg BUP þ 7 mg/kg DMFZ

(filled squares). In each group, five rats were used. Values represent mean ± SEM

at each time. No significant differences were shown using Mann-Whitney tests.

TABLE 3

Plasma Kinetic Parameters of DMFZ i.v. Infused Over 30 min in Rats Treated with either 30 mg/kg BUP 1 7 mg/kg DMFZ (BUP 1

DMFZ Group) or BUP Solvent 1 7 mg/kg DMFZ (DMFZ Group)

Group Half-life (min) Cmax (ng/ml) AUC0/N (ng/min/ml) Clt (ml/min/kg) Vss (ml/kg)

DMFZ group 62 ± 14 2024 ± 204 53,085 ± 9844 144 ± 23 3995 ± 1315

BUP þ DMFZ group 56 ± 9 2431 ± 197 66,099 ± 5036 108 ± 8 4053 ± 451

Note. Values represent mean ± SEM.

BUPRENORPHINE ALTERS FLUNITRAZEPAM TOXICITY 69

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

induced hypoxia and hypercapnia, whereas 0.3 mg/kg FZ alone

induced neither hypercapnia nor hypoxia and 40 mg/kg FZ

induced only significant hypercapnia.

Interestingly, we observed that 40 mg/kg DMFZ, the major FZ

metabolite induced hypoxia like the BUP/FZ combination,

whereas FZ did not. Thus, whereas the BZD relationship to

opioids in augmenting respiratory depression has been qualified

as simply additive (Henry, 1999), experimental data suggest that

the BUP/BZD interaction appears more complex than previously

believed, enhancing the interest of mechanistic studies.

Drug-drug interactions may result from either a pharmacoki-

netic or a pharmacodynamic process. Regarding BUP/FZ

kinetic interactions, previous in vitro studies performed on rat

as well as human microsome preparations predicted the absence

of in vivo metabolic interactions between both drugs at

therapeutic concentrations (Ibrahim et al., 2000; Kilicarslan

and Sellers, 2000; Umehara et al., 2002). In addition, we

previously demonstrated that rat pretreatment with FZ alter

neither plasma nor striatal BUP distribution (Megarbane et al.,2005a). Here we confirmed that 30 mg/kg BUP had no

significant impact on plasma FZ kinetics nor did 40 mg/kg FZ

have an impact on plasma BUP kinetics. In contrast, we clearly

showed that BUP pretreatment had a significant impact on the

time course of DMFZ concentrations, tripling its AUC0/N in

comparison to control (p ¼ 0.009). Moreover, although DMFZ

Cmax values were equivalent whether 40 mg/kg FZ (2708 ± 588

ng/ml) or 7 mg/kg DMFZ (2024 ± 204 ng/ml) was administered

to rats in the absence of BUP, DMFZ Cmax was significantly

higher in the presence of 30 mg/kg BUP, when 40 mg/kg FZ

(5592 ± 1312 ng/ml) rather than 7 mg/kg DMFZ (2431 ± 197

ng/ml) was administered (p ¼ 0.009, Tables 2B and 3).

The significant increase in PaCO2 with BUP/FZ combination

in comparison to FZ did not appear to be related to BUP-

mediated increase in DMFZ AUC0/N as DMFZ was not

responsible of any alteration in PaCO2. This observation thus

suggested that effect of BUP/FZ combination on PaCO2 may

be the consequence of additive or synergic direct pharmaco-

dynamic interactions of high dosages of both drugs. In contrast,

as DMFZ induced a significant decrease in PaO2, the decrease

in PaO2 with BUP/FZ combination in comparison to FZ or

BUP may be attributed at least in part to a BUP-mediated

increase in DMFZ AUC0/N. This effect was more clearly

established in this study at pharmacological (0.3 mg/kg) than in

our previous study at toxic dosage (40 mg/kg) of FZ

(Megarbane et al., 2005a), supporting the hypothesis of

a possible BUP/FZ kinetic interaction evidenced at a lower

dosage.

We considered various possible mechanisms of BUP/FZ

kinetic interaction that may result in the increase of DMFZ

concentrations. From a theoretical point of view, BUP effects

TABLE 4

Comparison of the Free/Total Ratios of Plasma FZ (A) and

DMFZ (B) in Rats Treated with either 30 mg/kg BUP 1 40 mg/

kg FZ (BUP 1 FZ Group) or BUP Solvent 1 40 mg/kg FZ (FZ

Group)

Treatment BUP þ FZ group FZ group p

(A) Free/total ratios of plasma FZ concentrations (%)

T-20 22.5 ± 1.5 16.9 ± 2.8 0.13

T0 20.5 ± 1.4 26.9 ± 5.6 0.94

(B) Free/total ratios of plasma DMFZ concentrations (%)

T-20 29.4 ± 4.2 29.7 ± 2.9 0.66

T0 21.4 ± 1.6 21.5 ± 4.0 1

Note. Measurements were performed at two times: 20 min after the initiation of

FZ infusion (T-20) and at the end of FZ infusion (T0). Values represent mean ±

SEM. No significant differences were shown using Mann-Whitney tests.

FIG. 5. Effects of i.v. 40 mg/kg FZ and 40 mg/kg DMFZ in comparison

with FZ solvent on arterial pH (A), PaCO2 (B), and PaO2 (C). In each group,

four rats were used. Values represent mean ± SEM of the AUC0/93 of each

parameter between the time before injection and time 93-min postinjection.

Comparisons were performed using one-way ANOVA followed by multiple

comparison tests using Bonferroni’s correction. PaCO2 was significantly more

elevated in the FZ group (*p < 0.05). PaO2 was significantly decreased in the

DMFZ group (*p < 0.05).

70 PIRNAY ET AL.

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

on DMFZ disposition may be the consequence of either

a formation-dependent or an elimination-dependent process.

BUP did not appear to interact with DMFZ elimination process

as there was no significant increase in DMFZ elimination half-

life when DMFZ was directly administered to the rats.

Interestingly, we observed parallel terminal slopes of both FZ

and DMFZ in plasma, irrespective of BUP administration

(Fig. 2). Moreover, we did not observe any significant BUP

effect on FZ or on DMFZ free concentrations in plasma (Table

4). FZ binds to human serum albumin and to alpha 1-acid

glycoprotein (Maruyama et al., 1992). Heel et al. (1979)

reported unpublished human data from Reckitt & Colman,

indicating that BUP is highly protein-bound (95–98%),

primarily to alpha and beta globulin fractions. However, to

our knowledge, FZ and BUP protein binding has never been

described in rats.

On the other hand, as we used a single BUP dose and as

BUP effect on FZ kinetics was rapidly observed as soon as 10

min after the start of FZ infusion, we assumed that we were

unable to assess a formation-dependent mechanism such as an

enzymatic induction. However, using the same calculation

previously performed in humans (Drouet-Coassolo et al.,1990), we showed that the DMFZ AUC0/N/FZ AUC0/N

ratio named ‘‘metabolism index’’ was significantly increased in

BUP-pretreated rats (41%) in comparison to controls (15%).

In humans, both cytochrome (CYP)3A4 and 2C19 are

involved in the metabolic pathways of FZ to DMFZ and to the

third FZ metabolite, 3-OHFZ (Coller et al., 1998; Hesse et al.,2001). Although not firmly established, CYP2C19 appears to

be the primary route for DMFZ formation, whereas CYP3A4 is

the primary route for 3-OHFZ formation (Kilicarslan et al.,2001). In contrast in rats, the metabolic pathways of FZ are

unknown, rendering it impossible to establish any definitive

hypothesis. However, we showed that administration of

massive doses of BUP resulted in significant DMFZ production

while not altering FZ and 7-AF concentrations, thus raising the

question of a possible substrate competition for FZ metabolism,

downshifting the route producing DMFZ relative to the routes

producing other metabolites, including 3-OHFZ. This hypoth-

esis may be supported by in vitro studies on human

microsomes showing significant inhibition of CYP3A4-medi-

ated pathways of 3-OHFZ (Ibrahim et al., 2000; Kilicarslan

and Sellers, 2000; Umehara et al., 2002). However, apparent Ki

values of CYP3A4-mediated 3-OHFZ formation were shown

to be at least 120 times more elevated than the expected plasma

BUP concentrations in treated patients (Umehara et al., 2002).

Thus, this kinetic mechanism of BUP/FZ interaction was

considered as clinically nonsignificant. Projected in vivoinhibition of CYP3A4-mediated FZ metabolism by BUP was

even estimated at 0.1–2.5% under pharmacological conditions

(Kilicarslan and Sellers, 2000). Our results were consistent

with these conclusions, as plasma BUP concentrations (Cmax:

8360 ± 2553 ng/ml ¼ 17.9 ± 5.5 lmol/l) was significantly

lower than the reported concentration (>150 lmol/l) required

for a 50% reduction in CYP3A activity in rat liver microsomes

(Ibrahim et al., 2000). Furthermore, under the hypothesis of

BUP inhibition of CYP3A-mediated pathway of FZ metabo-

lism, an increase in FZ concentrations would be expected,

contrasting with our observations. Thus, the exact mechanism

of BUP-mediated increase of DMFZ concentrations in vivoremains to be determined. However, based on our results, we

may clearly question the supposition that FZ/BUP interaction is

unrelated to a pharmacokinetic mechanism in the situation of

drug abusers with combination of elevated doses of both drugs,

resulting in severe or even fatal poisonings (Kintz, 2001;

FIG. 6. Effects of i.v. 30 mg/kg BUP þ 0.3 mg/kg FZ combination in

comparison to 30 mg/kg BUP alone, 0.3 mg/kg FZ alone, and solvents on

arterial pH (A), PaCO2 (B), and PaO2 (C). In each group, seven rats were used.

Values represent mean ± SEM of the AUC0/93 of each parameter between the

time before injection and time 93-min postinjection. Comparisons were

performed using one-way ANOVA followed by multiple comparison tests

using Bonferroni’s correction. PaCO2 was significantly more elevated in the

BUP (*p < 0.05) and BUP/FZ combination (**p < 0.01) groups. PaO2 was

significantly decreased in the BUP/FZ combination group (**p < 0.01).

BUPRENORPHINE ALTERS FLUNITRAZEPAM TOXICITY 71

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

Pirnay et al., 2004a; Reynaud et al., 1998; Tracqui et al.,1998). To our best knowledge, no study including FZ and

DMFZ kinetics was performed in such situation in humans.

Acute and prolonged multidrug exposures in drug addicts make

drug-drug kinetic interactions highly plausible. It would be of

interest, based on our data, to investigate BUP alteration of

DMFZ disposition in patients with acute respiratory depression

resulting from self coadministration of BUP and FZ.

One important limitation of this work was that 3-OHFZ,

including its possible contribution to toxicity, was not studied.

However, it is generally believed that the parent drug FZ is

principally responsible for the sleep-inducing effect, whereas

the relative activities of metabolites are unclear (Kilicarslan

et al., 2001). The pharmacological activity of 3-OHFZ has not

been established to our knowledge, whereas it has been

suggested that DMFZ is active (Berthault et al., 1996) and that

7-AFZ can have anesthetic activity in animals (Korttila and

Linnoila, 1976). Thus we chose to limit our study to the main

active metabolites of FZ.

In conclusion, our study showed that BUP/FZ combination

induced significant toxic effects on rat ventilation. Pretreatment

with BUP had no effects on FZ disposition while inducing

a three-fold increase in plasma DMFZ concentrations. When

analyzing the respiratory effects of this BUP/FZ combination,

the decrease in PaO2 appears to be rather related to BUP-

mediated effects on DMFZ disposition, whereas increase in

PaCO2 to a direct BUP/FZ additive or synergic dynamic

interaction. However, the exact mechanisms of each of these

drug/drug interactions remain to be determined.

FUNDING

MILDT (‘‘Mission Inter-ministerielle pour la Lutte contreles Drogues et la Toxicomanie’’); and Schering-Plough,

Levallois-Perret, France.

ACKNOWLEDGMENTS

We thank the Doctor Jerome Barre of the laboratory of

Pharmacology (Hopital intercommunal de Creteil) for his

helpful advice regarding the plasma filtration technique.

REFERENCES

Becherucci, C., Palmi, M., and Segre, G. (1985). Pharmacokinetics of

flunitrazepam in rats studied by a radioreceptor assay. Pharmacol. Res.

Commun. 17, 733–747.

Berthault, F., Kintz, P., and Mangin, P. (1996). Simultaneous high-performance

liquid chromatographic analysis of flunitrazepam and four metabolites in

serum. J. Chromatogr. B Biomed. Appl. 685, 383–387.

Borron, S. W., Monier, C., Risede, P., and Baud, F. J. (2002). Flunitrazepam

variably alters morphine, buprenorphine, and methadone lethality in the rat.

Hum. Exp. Toxicol. 21, 599–605.

Buckley, N. A., and McManus, P. R. (2004). Changes in fatalities due to

overdose of anxiolytic and sedative drugs in the UK (1983-1999). Drug Saf.

27, 135–141.

Coller, J. K., Somogyi, A. A., and Bochner, F. (1998). Quantification of

flunitrazepam’s oxidative metabolites, 3-hydroxyflunitrazepam and desme-

thylflunitrazepam, in hepatic microsomal incubations by high-performance

liquid chromatography. J. Chromatogr. B Biomed. Sci. Appl. 719, 87–92.

Cowan, A. (1995). Update on the general pharmacology of buprenorphine. In

Buprenorphine: Combating Drug Abuse With a Unique Opioid (A. Cowan

and J. W. Lewis, Eds.), pp. 31–47. Wiley-Liss, New York.

Cowan, A. (2003). Buprenorphine: New pharmacological aspects. Int. J. Clin.

Pract. Suppl. 3–8.

Cowan, A., Doxey, J., and Harry, E. (1977). The animal pharmacology of

buprenorphine, an oripavine analgesic agent. Br. J. Pharmacol. 60,

547–554.

Dahan, A., Yassen, A., Bijl, H., Romberg, R., Sarton, E., Teppema, L.,

Olofsen, E., and Danhof, M. (2005). Comparison of the respiratory effects of

intravenous buprenorphine and fentanyl in humans and rats. Br. J. Anaesth

94, 825–834.

Dahan, A., Yassen, A., Romberg, R., Sarton, E., Teppema, L., Olofsen, E., and

Danhof, M. (2006). Buprenorphine induces ceiling in respiratory depression

but not in analgesia. Br. J. Anaesth. 96, 627–632.

Drouet-Coassolo, C., Iliadis, A., Coassolo, P., Antoni, M., and Cano, J. P.

(1990). Pharmacokinetics of flunitrazepam following single dose oral

administration in liver disease patients compared with healthy volunteers.

Fundam. Clin. Pharmacol. 4, 643–651.

Druid, H., Holmgren, P., and Ahlner, J. (2001). Flunitrazepam: An evaluation

of use, abuse and toxicity. Forensic Sci. Int. 122, 136–141.

Drummer, O. H., Syrjanen, M. L., and Cordner, S. M. (1993). Deaths involving

the benzodiazepine flunitrazepam. Am. J. Forensic Med. Pathol. 14, 238–243.

Emmanuelli, J., and Desenclos, J. C. (2005). Harm reduction interventions,

behaviors and associated health outcomes in France, 1996-2003. Addiction

100, 1690–1700.

Garretty, D. J., Wolff, K., Hay, A. W., and Raistrick, D. (1997).

Benzodiazepine misuse by drug addicts. Ann. Clin. Biochem. 34, 68–73.

Gueye, P. N., Borron, S. W., Risede, P., Monier, C., Buneaux, F., Debray, M.,

and Baud, F. J. (2002a). Buprenorphine and midazolam act in combination to

depress respiration in rats. Toxicol. Sci. 65, 107–114.

Gueye, P. N., Borron, S. W., Risede, P., Monier, C., Buneaux, F., Debray, M.,

and Baud, F. J. (2001). Lack of effect of single high doses of buprenorphine

on arterial blood gases in the rat. Toxicol. Sci. 62, 148–154.

Gueye, P. N., Megarbane, B., Borron, S. W., Adnet, F., Galliot-Guilley, M.,

Ricordel, I., Tourneau, J., Goldgran-Toledano, D., and Baud, F. J. (2002b).

Trends in opiate and opioid poisonings in addicts in north-east Paris and

suburbs, 1995-99. Addiction 97, 1295–1304.

Heel, R. C., Brogden, R. N., Speight, T. M., and Avery, G. S. (1979).

Buprenorphine: A review of its pharmacological properties and therapeutic

efficacy. Drugs 17, 81–110.

Henry, J. (1999). Fatal opioid toxicity—A clinical perspective. Addiction 94,

974–975.

Hesse, L. M., Venkatakrishnan, K., von Moltke, L. L., Shader, R. I., and

Greenblatt, D. J. (2001). CYP3A4 is the major CYP isoform mediating the in

vitro hydroxylation and demethylation of flunitrazepam. Drug Metab.

Dispos. 29, 133–140.

Hojer, J., Baehrendtz, S., and Gustafsson, L. (1989). Benzodiazepine

poisoning: Experience of 702 admissions to an intensive care unit during

a 14-year period. J. Intern. Med. 226, 117–122.

Ibrahim, R. B., Wilson, J. G., Thorsby, M. E., and Edwards, D. J. (2000). Effect

of buprenorphine on CYP3A activity in rat and human liver microsomes.

Life Sci. 66, 1293–1298.

72 PIRNAY ET AL.

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from

Kilicarslan, T., Haining, R. L., Rettie, A. E., Busto, U., Tyndale, R. F., and

Sellers, E. M. (2001). Flunitrazepam metabolism by cytochrome P450S

2C19 and 3A4. Drug Metab. Dispos. 29, 460–465.

Kilicarslan, T., and Sellers, E. M. (2000). Lack of interaction of buprenorphine

with flunitrazepam metabolism. Am. J. Psychiatry 157, 1164–1166.

Kintz, P. (2001). Deaths involving buprenorphine: A compendium of French

cases. Forensic Sci. Int. 121, 65–69.

Korttila, K., and Linnoila, M. (1976). Amnesic action of and skills related to

driving after intravenous flunitrazepam. Acta Anaesthesiol. Scand. 20, 160–168.

Lai, S. H., Yao, Y. J., and Lo, D. S. (2006). A survey of buprenorphine related

deaths in Singapore. Forensic Sci. Int. 162, 80–86.

Libong, D., Pirnay, S., Bruneau, C., Rogalewicz, F., Ricordel, I., and

Bouchonnet, S. (2003). Adsorption-desorption effects in ion trap mass

spectrometry using in situ ionization. J. Chromatogr. A 1010, 123–128.

Mandema, J. W., Sansom, L. N., Dios-Vieitez, M. C., Hollander-Jansen, M.,

and Danhof, M. (1991). Pharmacokinetic-pharmacodynamic modeling of the

electroencephalographic effects of benzodiazepines. Correlation with receptor

binding and anticonvulsant activity. J. Pharmacol. Exp. Ther. 257, 472–478.

Maruyama, T., Furuie, M. A., Hibino, S., and Otagiri, M. (1992). Comparative

study of interaction mode of diazepines with human serum albumin and

alpha 1-acid glycoprotein. J. Pharm. Sci. 81, 16–20.

McCormick, G. Y., White, W. J., Zagon, I. S., and Lang, C. M. (1984). Effects

of diazepam on arterial blood gas concentrations and pH of adult rats acutely

and chronically exposed to methadone. J. Pharmacol. Exp. Ther. 230,

353–359.

Megarbane, B., Lesguillons, N., Galliot-Guilley, M., Borron, S. W., Trout, H.,

Decleves, X., Risede, P., Monier, C., Boschi, G., and Baud, F. J. (2005b).

Cerebral and plasma kinetics of a high dose of midazolam and correlations

with its respiratory effects in rats. Toxicol. Lett. 159, 22–31.

Megarbane, B., Pirnay, S., Borron, S. W., Trout, H., Monier, C., Risede, P.,

Boschi, G., and Baud, F. J. (2005a). Flunitrazepam does not alter cerebral

distribution of buprenorphine in the rat. Toxicol. Lett. 157, 211–219.

Nielsen, S., and Taylor, D. A. (2005). The effect of buprenorphine and

benzodiazepines on respiration in the rat. Drug Alcohol Depend 79, 95–101.

Ohtani, M., Kotaki, H., Nishitateno, K., Sawada, Y., and Iga, T. (1997).

Kinetics of respiratory depression in rats induced by buprenorphine and its

metabolite, norbuprenorphine. J. Pharmacol. Exp. Ther. 281, 428–433.

Pirnay, S., Borron, S., Giudicelli, C., Tourneau, J., Baud, F. J., and Ricordel, I.

(2004a). A critical review of the causes of death among post-mortem

toxicological investigations: Analysis of 34 buprenorphine-associated and 35

methadone-associated deaths. Addiction 99, 978–988.

Pirnay, S., Bouchonnet, S., Herve, F., Libong, D., Milan, N., D’Athis, P.,

Baud, F. J., and Ricordel, I. (2004b). Development and validation of a gas

chromatography-mass spectrometry method for the simultaneous determi-

nation of buprenorphine, flunitrazepam and their metabolites in rat plasma:

Application to the pharmacokinetic study. J. Chromatogr. B Anal. Technol.

Biomed. Life Sci. 807, 335–342.

Pirnay, S., Ricordel, I., Libong, D., and Bouchonnet, S. (2002). Sensitive

method for the detection of 22 benzodiazepines by gas chromatography-ion

trap tandem mass spectrometry. J. Chromatogr. A 954, 235–245.

Randall, L. O., and Kappell, B. (1973). Pharmacological activity of some

benzodiazepines and their metabolites. In The Benzodiazepines (S. Garattini,

L. O. Randall, and E. Mussini, Eds.), pp. 27–51. Raven Press, New York.

Reynaud, M., Tracqui, A., Petit, G., Potard, D., and Courty, P. (1998). Six

deaths linked to misuse of buprenorphine-benzodiazepine combinations. Am.

J. Psychiatry 155, 448–449.

Shah, V. P., Midha, K. K., Findlay, J. W., Hill, H. M., Hulse, J. D.,

McGilveray, I. J., McKay, G., Miller, K. J., Patnaik, R. N., Powell, M. L.,

et al. (2000). Bioanalytical method validation—A revisit with a decade of

progress. Pharm. Res. 17, 1551–1557.

Tracqui, A., Kintz, P., and Ludes, B. (1998). Buprenorphine-related deaths

among drug addicts in France: A report on 20 fatalities. J. Anal. Toxicol. 22,

430–434.

Umehara, K., Shimokawa, Y., and Miyamoto, G. (2002). Inhibition of human

drug metabolizing cytochrome P450 by buprenorphine. Biol. Pharm. Bull.

25, 682–685.

Verborgh, C., De Coster, R., D’Haese, J., Camu, F., and Meert, T. F. (1998).

Effects of chlordiazepoxide on opioid-induced antinociception and re-

spiratory depression depression in restrained rats. Pain 37, 103–110.

Verwey, B., Eling, P., Wientjes, H., and Zitman, F. G. (2000). Memory

impairment in those who attempted suicide by benzodiazepine overdose. J.

Clin. Psychiatry 61, 456–459.

Walsh, S., Preston, K., Stitzer, M., Cone, E., and Bigelow, G. (1994). Clinical

pharmacology of buprenorphine: Ceiling effect at high doses. Clin.

Pharmacol. Ther. 65, 569–580.

Zbinden, G., and Randall, L. O. (1967). Pharmacology of benzodiazepines:

Laboratory and clinical correlations. In Advances in Pharmacology (S. Garattini

and P. A. Shore, Eds.), pp. 213–291. Academic Press, New York.

BUPRENORPHINE ALTERS FLUNITRAZEPAM TOXICITY 73

by guest on June 3, 2013http://toxsci.oxfordjournals.org/

Dow

nloaded from