throbber
Introduction
`
`
`
`SCIENTIFIC DISCUSSION
`
`1.
`
`Drug addiction is a worldwide problem of which opioid dependence, notably heroin addiction, is a
`major component. Most addicts inject drugs, quite often with dirty or shared syringes and needles and
`this behaviour is linked directly with the transmission of human immunodeficiency virus (HIV) and
`the hepatitis viruses. A key aim of treatment programs for opioid drug dependence is to stop the
`subjects from injecting drugs. Substitution is the treatment approach for opioid dependence in which
`street heroin of unknown strength and purity is replaced with a pharmaceutical grade opioid with a
`longer duration of action, such as buprenorphine.
`
`Buprenorphine is a well-known substance available in several European countries for the treatment of
`severe pain. For the treatment of opioid dependence it was first approved in 1995 (France) and is
`currently available in most European countries. Buprenorphine has lower intrinsic activity than
`methadone and other full agonists, produces less sedation and cognitive impairment, and has a ceiling
`on potential depressant effects, even if injected, particularly on cardiac and respiratory functions.
`Sublingual buprenorphine (marketed as Buprenorphine alone) is an established substitution treatment
`for opiate abuse, but there has been some diversion to the intravenous route because buprenorphine
`produces a moderate opiate agonist effect. Thus, in the opinion of the applicant, there is a need for a
`formulation of buprenorphine that has low potential for intravenous misuse.
`
`SUBOXONE is a fixed combination product for chronic substitution therapy in opiate dependence
`consisting of buprenorphine and naloxone formulated
`into a sublingual
`tablet containing
`buprenorphine and naloxone in the ratio 4:1 of the bases.
`The claimed indication for SUBOXONE is substitution treatment for opioid drug dependence,
`within a framework of medical, social and psychological treatment. The intention of the
`naloxone component is to deter intravenous misuse. As requested by the CHMP, treatment is
`intended for use in adults and adolescents over 15 years of age who have agreed to be treated for
`addiction.
`The product is intended as a “take home” medication presented in two strengths:
`1. Buprenorphine 8 mg + naloxone 2 mg sublingual tablet
`2. Buprenorphine 2 mg + naloxone 0.5 mg sublingual tablet.
`
`The combination of an opioid antagonist with a potent µ-opioid analgesic is an established strategy for
`reducing the potential for intravenous misuse. Naloxone is a well-known opioid antagonist. As a
`mono-substance it is indicated for the treatment of opioid-overdosage or –intoxication. When
`administered in usual doses to patients who have not recently received opioids, naloxone exerts little
`or no pharmacologic effect. In patients who have received large doses of opioids, naloxone
`antagonises most of the effects of the opioid. The addition of naloxone to buprenorphine is intended to
`render the product less abusable by deterring intravenous injection.
`
`2. Quality aspects
`
`Introduction
`
`Suboxone is presented as sublingual tablets containing a fixed dose combination of buprenorphine
`hydrochloride and naloxone hydrochloride dihydrate, at a ratio of 4:1, with respect to the free bases.
`Suboxone is available in two strengths:
`• 2 mg / 0.5 mg tablets containing 2.16 mg buprenorphine hydrochloride (equivalent to 2 mg
`buprenorphine base) and 0.61 mg naloxone hydrochloride dihydrate (equivalent to 0.5 mg
`naloxone base).
`
`
`
`©EMEA 2006
`
`1/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`• 8 mg / 2 mg containing 8.64 mg buprenorphine hydrochloride (equivalent to 8 mg
`buprenorphine base) and 2.44 mg naloxone hydrochloride dihydrate (equivalent to 2 mg
`naloxone base).
`The excipients used in this formulation are lactose monohydrate, mannitol, maize starch, povidone
`K30, citric acid anhydrous granular, sodium citrate, natural lemon and lime flavour, acesulfame
`potassium and magnesium stearate. Suboxone is administered via the sublingual route and is packed in
`nylon/aluminium/PVC blister packs containing either 7 or 28 tablets.
`
`Active Substance 1. (Buprenorphine hydrochloride)
`
`Buprenorphine hydrochloride is an established active substance and the subject of a monograph in the
`Ph. Eur.
`Buprenorphine hydrochloride is designated chemically as (2S)-2-[17-Cyclopropylmethyl-4,5α-epoxy-
`3-hydroxy-6-methoxy-6α,14-ethano-14α-morphinan-7α-yl]-3,3-dimethylbutan-2-ol
`hydrochloride
`and its chemical structure is as follows:
`
`
`
`
`Buprenorphine hydrochloride is a white or almost white, crystalline powder, sparingly soluble in
`water, freely soluble in methanol, soluble in alcohol, practically insoluble in cyclohexane.
`Buprenorphine has several chiral centres and it is therefore optically active.
`The potential for polymorphism was investigated using powder X-Ray diffraction and Differential
`Scanning Calorimetry (DSC) techniques. The results showed that there is no evidence for
`polymorphism.
`
` •
`
` Manufacture
`Buprenorphine hydrochloride is synthesized from thebaine. The structure has been confirmed by
`elemental analysis, spectroscopic analysis (UV, IR, NMR and MS) and X-Ray crystallography. The
`stereochemistry of the intermediates and the final active substance was investigated using X-ray
`crystallography and NMR spectroscopy. The absolute configuration was confirmed at different stages
`during the synthesis.
`
` •
`
` Specification
`The specification of the active substance includes physical description, visual inspection of the
`appearance in solution, assay by titration and by HPLC, specific optical rotation, acidity or alkalinity
`and related substances (HPLC). Additional tests performed are as follows: control for water content
`using the Karl Fisher method and residue on ignition, residual solvent, ionic chloride and particle size.
`
`The analytical methods used were those described in the PhEur. Monograph, one major exception
`being the determination of related substances. The HPLC method for determination of related
`impurities uses specific impurity markers. It allows detection and quantitation of the five major
`impurities specified, whereas using the method described in the Ph. Eur. only two of the impurities can
`be detected. In addition, the acceptance criteria set for each specified impurity is more stringent than
`the limits mentioned in Ph. Eur. monograph. The maximum limit for total related impurities is also
`
`
`
`©EMEA 2006
`
`2/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`more stringent than that mentioned in the current Ph. Eur. Monograph. All analytical methods have
`been validated according to the ICH guideline on “Validation of Analytical methods”.
`
`Batch analysis data was provided for 23 batches of buprenorphine hydrochloride manufactured
`following the proposed synthetic method. The results showed that the active substance can be
`reproducibly manufactured.
`
`
` Stability
`The stability of buprenorphine hydrochloride was investigated in 3-production scale batches stored in
`the proposed packaging according to the ICH guideline. Stability studies were performed under long
`term and intermediate ICH conditions for 156 weeks, and accelerated ICH conditions for 52 weeks.
`An additional study was performed using a larger scale production batch. In this study the stability
`studies were performed under long-term and intermediate conditions for 52 weeks and accelerated
`conditions for 39 weeks. The results obtained demonstrate that buprenorphine hydrochloride remains
`physically and chemically stable for 52 week at long-term and intermediate conditions and 26 weeks
`at accelerated conditions.
`
`The data provided is sufficient to confirm the proposed re-test period.
`
`Active Substance 2. (Naloxone hydrochloride dihydrate)
`
`Naloxone hydrochloride dihydrate is an established active substance and the subject of a monograph
`in the Ph. Eur. Naloxone hydrochloride dihydrate is designated chemically as Morphinan-6-one, 4,5-
`epoxy-3, 14-dihydroxy-17-(2-propenyl)-, hydrochloride, (5α)-dihydrate. Its chemical structure is as
`follows:
`
` •
`
`2
`
`HO
`
`3
`
`
`
`4
`
`O
`
`5
`
`12
`
`15
`
`13
`
`14
`
`1
`
`11
`
`10
`16
`
`
`19
`
`18
`
`N
`
`9
`OH
`
`17
`
`6
`
`O
`
`7
`
`8
`
`
`
`Naloxone hydrochloride dihydrate is a white or almost white crystalline powder, hygroscopic, soluble
`in water and alcohol, practically insoluble in ether. The pKa of Naloxone hydrochloride dihydrate is
`7.94 at 20oC and the melting point is 200-205oC.
`
` •
`
` Manufacture
`Naloxone hydrochloride is synthesised from noroxymorphone. The assigned structure of naloxone
`hydrochloride dihydrate is supported by the evidence of IR spectrophotometry, and by 1H-NMR and
`13C-NMR spectrometry.
`
`Naloxone hydrochloride dihydrate contains four chiral centres, all of which are already present in the
`starting material of the synthesis, noroxymorphone, which is derived from natural opiates.
`
`The possibility of polymorphism was investigated by standard techniques. The results showed that all
`batches exhibited the same morphic form.
`
` •
`
` Specification
`Naloxone hydrochloride dihydrate is tested for compliance with both PhEur. and USP monographs by
`the active substance manufacturer. These tests include physical description, identification by IR, TLC
`and chloride, specific optical rotation, loss on drying, Noroxymorphone hydrochloride and other
`impurities by TLC, chloride content, appearance of solution, acidity or alkalinity, related substances
`by HPLC, water content, sulphated ash and assay by titration. The specification also includes some
`additional non-pharmacopoeial tests (a stability-indicating HPLC method for assay and related
`
`
`
`©EMEA 2006
`
`3/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`substances, UV identification, sieve analysis and the melting range of naloxone base). The stability-
`indicating HPLC assay has been fully validated and was shown to have satisfactory linearity,
`precision, accuracy and ruggedness. The peak area of naloxone decreases and degradation products are
`observed in samples exposed to stress conditions, confirming that the assay is stability-indicating.
`
`Five batches of naloxone hydrochloride were manufactured using the proposed synthetic method. The
`results indicate that every batch complied with the limits for related substances.
`
` •
`
` Stability
`The stability of naloxone hydrochloride was investigated in 12 batches stored in the proposed
`packaging according to the ICH guideline. Stability studies were performed under long term,
`intermediate and accelerated ICH conditions for up to 60 months. No marked evidence of instability
`was revealed under any of the storage conditions and the proposed re-test period appears to be
`justified on the basis of the stability data presented.
`
`Medicinal Product
`
` •
`
` Pharmaceutical Development
`Suboxone was developed in order to deliver a similar dose of buprenorphine compared to
`buprenorphine alone tablets (medicinal product containing buprenorphine that is authorised in the EU
`for the treatment of opioid addiction), but reducing the potential for intravenous abuse. Naloxone, an
`opiate antagonist, has poor bioavailability when administered by the sublingual route and
`consequently when Suboxone is taken sublingually it shows only the required effects of
`buprenorphine and delivers the same performance as an equivalent dose of buprenorphine alone
`tablets. However, if abused intravenously by an opiate-dependent subject, the antagonist effects of
`naloxone become apparent first as intense withdrawal symptoms followed by the attenuated agonist
`effects of buprenorphine.
`
`Therefore the Suboxone formulation is closely based on the formulation of buprenorphine alone
`sublingual tablets but with naloxone added to reduce the potential for abuse by the intravenous route.
`A buprenorphine to naloxone ratio of 4:1 contains sufficient naloxone to produce opiate antagonist
`effects following intra-venous administration, but does not impair the effectiveness of buprenorphine
`when the mixture is taken by the sublingual route.
`
`The excipients used in Suboxone are qualitatively and quantitatively identical to those used in the
`existing buprenorphine alone sublingual tablets, i.e., lactose monohydrate, mannitol, maize starch,
`povidone K30, citric acid anhydrous, sodium citrate and magnesium stearate. Acesulfame potassium
`and natural lemon and lime flavour (sweetener and flavouring agents, respectively) were included to
`disguise the bitter taste of naloxone. The content of lactose monohydrate was reduced slightly in order
`to maintain identical compression weights. All excipients have been widely used in commercial
`pharmaceutical dosage forms or as food additives. Except for the natural lemon and lime flavour all
`excipients comply with the specification of the Ph. Eur. Natural lemon and lime flavour is a natural
`flavouring, which complies the requirements of directive 88/388/EEC (as amended) on flavourings for
`use in food. Certificates of analysis have been provided for all excipients.
`
` •
`
` •
`
` Adventitious Agents
`Lactose monohydrate is the only excipient of animal origin. However, it is prepared from bovine milk
`suitable for human consumption, which is sourced from healthy animals. Magnesium stearate is of
`vegetable origin.
`
` Manufacture of the Product
`The manufacturing process of the finished product comprises standard mixing, wet granulation and
`compression techniques. Process parameter ranges (sieve sizes, mixing times and speed, drying time
`and temperature) were described for each step of the manufacturing process. Validation studies
`involved the preparation of 3 full-scale batches of the tablet blend. Each of the batches was then sub-
`divided into two sub-lots for the preparation of tablets of both strengths, i.e., 2 mg/0.5 mg tablets and
`8 mg/2 mg tablets. An additional full-scale batch of each tablet strength was manufactured. All eight
`batches complied with final product specification. From the evidence of the process validation studies
`
`
`
`©EMEA 2006
`
`4/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`provided, it can be concluded that the process is capable of consistently producing batches of the
`required quality.
`
`
` Product Specification
`The product specifications include methods for appearance, identification (buprenorphine and
`naloxone) by HPLC and TLC, assay and content uniformity (buprenorphine and naloxone) by HPLC,
`dissolution of buprenorphine and dissolution of naloxone, disintegration time, buprenorphine
`degradation products, naloxone degradation products, water content and microbiological integrity.
`
`The drug product specifications have been justified and all methods of analysis have been described
`and adequately validated.
`
` •
`
` •
`
` Stability of the Product
`Stability data on three batches of each strength of Suboxone sublingual tablets (8 mg / 2 mg and 2 mg
`/ 0.5 mg) packaged under a nitrogen atmosphere was provided. The studies were performed under
`long-term, intermediate, and accelerated conditions. The parameters evaluated during these studies
`were those mentioned in the shelf-life specification, except for two minor deviations. Analytical
`results up to 156 weeks were presented. All tests remained within specification for 156 weeks at
`25°C/60% RH. The key shelf-life limiting parameter appeared to be disintegration time. There is
`evidence of a time-dependent increase but all samples stored at complied with the specification for
`156 weeks.
`
`
`Based on the available stability data, the proposed shelf life and storage conditions, as stated in the
`SPC, are acceptable.
`
`Discussion on chemical, pharmaceutical and biological aspects
`
`Information on development, manufacture and control of the drug substances and drug product has
`been presented in a satisfactory manner. The results of test carried out indicate satisfactory consistency
`and uniformity of important product quality characteristics, and these in turn lead to the conclusion
`that the product should have a satisfactory and uniform performance in the clinic.
`
`
`
`
`Introduction
`Most of the preclinical studies were conducted in accordance with good laboratory practice
`regulations. Some studies have been performed prior to the introduction of GLP regulation and are not
`GLP-compliant. Since both of the active ingredients are established substances the documentation for
`pharmacology consists of published literature plus study reports with the combination of the active
`ingredients.
`
` •
`
` Pharmacology
`Buprenorphine is a semisynthetic, highly lipophilic opioid derived from thebaine with a 25 -30 fold
`higher analgesic potency as compared to morphine and a longer lasting effect. It is a partial agonist at
`the µ- and an antagonist at the κ-opioid receptor subtype. It dissociates very slowly from opioid
`receptors (t½ 166 min vs. 7 min for fentanyl) and is, once bound, hardly displaced by naloxone,
`however, prior treatment with naloxone can prevent e.g. respiratory depression. It is able to substitute
`for other opioids such as heroin but provides only moderate opiate agonist effects and a low degree of
`physical dependence. Being a partial µ-receptor agonist it may cause symptoms of abstinence in
`patients treated with µ-receptor agonists (e.g. morphine) and restricts its own analgesic effects once a
`maximum is reached, resulting in a bell-shaped dose response curve. When treatment with
`buprenorphine is discontinued withdrawal signs are generally mild due to slow dissociation from the
`µ-receptor and concomitant adaptive processes.
`
`Naloxone is the N-allyl derivative of oxymorphone. It has antagonistic effects at µ, δ- and κ-opioid
`receptors and is currently marketed in injectable form for the complete or partial reversal of opiate
`effects or for the suspected acute opiate overdose. When given alone, hardly any effect is observed
`
`
`
`©EMEA 2006
`
`5/42
`
`
`
`Non-clinical aspects
`
`3.
`
` •
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`hinting at a low endogenous opioidergic tonus. Because of its almost complete first pass metabolism,
`naloxone administered orally or sublingually is not expected to exert antagonistic activity. No
`documents dealing with the pharmacodynamics of naloxone were submitted.
`
`Receptor Binding:
`In the review presented in association with this application the receptor binding affinities of
`buprenorphine and naloxone for narcotic receptors are outline in the following table.
`
`Table: In vitro receptor-binding affinities for Buprenorphine and Naloxone.
`Drug
`Ki (nM)
`Kappa (κ)
`
`Buprenorphine
`1.1
`Naloxone
`12
`
`Mu (µ)
`0.77
`1.2
`
`Delta (δ)
`2.2
`19
`
`Sigma (σ)
`>100000
`>1000000
`
`Ratio (σ/κ)
`>91000
`>83000
`
` •
`
` Primary pharmacodynamics
`In vivo studies have been carried out in order to examine the interaction of both substances as regards
`effects (antinociception), precipitation of withdrawal in morphine dependent rats, drug discrimination
`in rats and avoidance behaviour in monkeys.
`
`Antinociceptive effects of buprenorphine and morphine given alone or in combination with naloxone
`(subcutaneous route) were measured in the rat tail pressure test: the results provided some evidence
`that buprenorphine is antagonised by naloxone, although less readily than morphine.
`
`Buprenorphine and naloxone administered intraperitoneally to precipitate withdrawal signs in
`morphine-dependent rats suggest that buprenorphine did not affect the ability of naloxone to
`precipitate signs of opiate withdrawal.
`
`To test whether buprenorphine plus naloxone mixtures are still perceived like buprenorphine alone, a
`drug discrimination study was performed. Male rats with about 300 g b.w. were trained to discriminate
`between subcutaneous (s.c.) injection of buprenorphine 0.03 mg/kg in an operant chamber with two
`levers, where repeated pressure of the correct lever (10x) resulted in the delivery of a food pellet. 30
`min after application the pattern of lever pressing was recorded. No food reward was given during
`generalisation trials. Generalisation was considered to have occurred, if the percent responding on the
`relevant lever was 70 % or more. Responding to the buprenorphine-appropriate lever was 97 %
`following buprenorphine, 2 % following saline and 8 % following naloxone. Addition of naloxone
`0.002, 0.01 and 0.02 mg/kg to buprenorphine resulted in 93 %, 59 % and 23 % buprenorphine lever
`pressing, respectively. It is concluded that buprenorphine combined with naloxone in a ratio 3:2 is not
`probable to be a narcotic cue to support opiate misuse.
`
`Negative reinforcing properties of naloxone were studied in the non-dependent rhesus monkey:
`scheduled infusions of naloxone (1-100 µg/kg/inf.) and of buprenorphine (250 µg/kg/inf.) generated
`drug avoidance behavior in the non-dependent rhesus monkey under a continuous avoidance-escape
`paradigm. Pentazocine (1-100 µg/kg/inf.) codeine, (1-100 µg/kg/inf. and tilidine (1-250 µg/kg/inf.)
`were ineffective. Addition of varying doses of naloxone to scheduled infusions of codeine, tilidine,
`and pentazocine generated avoidance behavior not present with scheduled infusions of these opioids
`alone. The naloxone doses necessary for generation of avoidance behavior were low with the agonists
`codeine and tilidine, higher with the weak antagonist pentazocine, and highest with the strong
`antagonist buprenorphine.
`Monkeys were trained to avoid conditioned noxious stimuli. Subsequently, avoidance behaviour was
`extinguished and scheduled intravenous infusions of drugs were tested for their ability to re-introduce
`avoidance behaviour. Comparable to nalorphine, the opioid antagonist naloxone exerted negative
`reinforcing properties. The agonists codeine and tilidine had no effects on the behaviour of their own,
`as had the mixed agonist/antagonist pentazocine, while buprenorphine had a weak negative reinforcing
`effect only in the highest dose tested. Naloxone was added to equi-analgesic doses of the test
`compounds. Low doses were needed to induce negative reinforcement in the presence of codeine and
`tilidine, somewhat higher doses in the presence of pentazocine and high doses in the presence of
`buprenorphine.
`
`
`
`©EMEA 2006
`
`6/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`
`Overall, where interaction between buprenorphine and naloxone was shown, the routes of
`administration used in the pharmacology studies may have limited relevance to the proposed clinical
`route of administration (sublingual) and the ratios chosen in these studies (3:1) do not reflect the
`proposed clinical ratio of 4:1. Thus ‘proof of principle’ that co-administration of naloxone will prevent
`intravenous misuse of buprenorphine has further to be derived from clinical studies and clinical
`evidence.
`
`Buprenorphine and norbuprenorphine did not displace peripheral [3H]PK 11195 binding and central
`[3H]flunitrazepam binding, indicating lack of interaction with the GABAA-receptors.
`
`Diazepam and flunitrazepam have no significant affinity to human µ- and δ- opioid receptors and poor
`affinity to human κ-opioid receptors.
`
` •
`
` Secondary pharmacodynamics
`In order to look for blood compatibility (protein precipitating and haemolytic effects) of the
`combination of buprenorphine and naloxone 3: 2, the powdered product was dissolved in 5 % aqueous
`glucose or was diluted from a pre-prepared solution and added to citrated blood withdrawn from
`beagle dogs. The slight haemolysis observed with therapeutic concentration is not likely to be of
`clinical significance in the case that intravenous abuse of the product should occur.
`
` •
`
` •
`
` Safety pharmacology programme
`No new studies were submitted. Due to the well-known pharmacology of the single substances and
`since no adverse interactions between these to substances are expected this procedure is acceptable.
`
` Pharmacodynamic drug interactions
`No unexpected pharmacodynamic drug interactions have been identified.
`
`Pharmacokinetics
`Pharmacokinetics of both buprenorphine and naloxone are well known. Buprenorphine is sufficiently
`absorbed following sublingual administration and is eliminated with a long half-life. Naloxone has a
`low oral bioavailability and and is rapidly eliminated when given parenterally.
`In a preclinical study it is shown, that co-administration of buprenorphine and naloxone had little or
`no effect on their individual pharmacokinetic profiles following administration by i.v. or i.m or by oral
`dosing.
`
`
`
`©EMEA 2006
`
`7/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`
`Table. Results of single dose pharmacokinetic studies in rats and dogs with 3H-buprenorphine and 3H-naloxone
`alone or in combination with non-radiolabelled naloxone or buprenorphine respectively.
`3H-Buprenorphine
`3H Buprenorphine +
`3H-Naloxone
`R
`S
`O
`Naloxone
`P
`U
`E
`AUC (0-8
`T
`C
`hr)
`E
`I
`ng/g h
`
`E
`S
`Rat
`
`3H-Naloxone +
`Buprenorphine
`AUC
`Cmax
`Cmax
`Plasm
`Brain
`(0-8 hr)
`a ng/g
`ng/
`ng/g h
`equiv
`
`690
`760
`26
`Auc (0-
`24 hr)
`ng/g h
`20
`Auc
`(0-96
`hr)
`ng/g h
`25.5
`5.5
`
`1736
`1850
`5853
`
`
`*
`
`
`*
`*
`
`Dog
`
`
`
` •
`
`Cmax
`Plasm
`a ng/g
`
`AUC (0-8
`hr)
`ng/g h
`
`i.v.
`i.m.
`p.o.
`
`
`1442
`892
`571
`
`
`i.v.
`
`
`788
`
`
`1799
`2248
`188
`Auc (0-24
`hr) ng/g
`h
`65
`Auc (0-
`96 hr)
`ng/g h
`
`i.m
`p.o.
`
`103
`ND
`
`68.5
`ND
`
`Cmax
`Brain
`ng/
`equiv
`
`4763
`1051
`1456
`
`
`Cmax
`Plas
`ma
`ng/g
`
`1306
`839
`915
`
`
`*
`
`
`*
`*
`
`739
`
`
`129
`ND
`
`Cmax
`Brain
`ng/
`equivs
`
`3626
`760
`1564
`
`
`Cmax
`Plasm
`a ng/g
`
`761
`865
`313
`
`
`*
`
`
`*
`*
`
`349
`
`
`258
`ND
`
`AUC
`(0-8 hr)
`Ng/g
`h
`
`726
`908
`48
`Auc (0-
`24 hr)
`ng/g h
`18
`Auc
`(0-96
`hr)
`ng/g h
`26
`8
`
`1653
`1453
`243
`Auc (0-24
`hr) ng/g
`h
`67
`Auc (0-
`96 hr)
`ng/g h
`
`64
`ND
`
`Cmax
`Brain
`ng/
`equiv
`
`2141
`1756
`8518
`
`
`574
`833
`134
`
`
`*
`
`
`*
`*
`
`357
`
`
`258
`ND
`
`The only clinically relevant pharmacokinetic interaction detected is confined to inhibitors of CYP3A
`resulting in enhanced bioavailability of buprenorphine. Based on these studies inhibitors of CYP 3A
`enzyme can potentially increase the hepatic toxicity of buprenorphine. Patients concomitantly
`administered inhibitors of CYP 3A should be closely monitored for markers of liver toxicity.
`This is reflected in the clinical pharmacokinetics and is taken into account in the SPC.
`
`Toxicology
`
` Single dose toxicity
`Extensive acute toxicity studies employing different routes of administration (i.v., s.c. i.m. and oral)
`are presented in which the toxicity of buprenorphine and naloxone are compared with the toxicity of
`mixtures of these components. These studies demonstrate that there is no synergistic enhancement of
`toxicity when buprenorphine and naloxone are co-administered.
`
` •
`
` Repeat dose toxicity (with toxicokinetics)
`Dietary toxicity studies of 28 days and 13 weeks duration in rats were conducted using buprenorphine:
`naloxone at a ratio of 4:1 in terms of the bases (i.e. the ratio intended for human use).
`The dietary route was selected for repeated dose toxicity evaluation of Suboxone since this route was
`relevant to the proposed sublingual route of human exposure. In addition to data on the dietary
`toxicity of Suboxone in rats, data on the toxicity of development formulations containing ratios of
`buprenorphine and naloxone of 1:1 and 3:2 for periods of up to 28 days in both the rat and the dog by
`a variety of enteral and parenteral routes of administration are presented.
`
`Clinical observations were aggressive behaviour and excessive grooming. An analgesic effect
`(prolonged time to tail flick) has been observed in females of the 2000 ppm group. Bodyweight was
`significant lower throughout the study for males only. Food consumption and food utilisation was
`decreased in males also. A treatment-related increase in adrenal weight adjusted for body weight was
`recorded for males receiving Suboxone. The adrenals were histologically normal and this apparent
`weight change was not considered to be of toxicological importance. No other organ weight changes
`were apparent that were considered to be related to treatment and no treatment-related abnormalities
`were observed at autopsy. Histological examination revealed an increased incidence and severity of
`mononuclear cell infiltration of the Harderian gland in females of all groups receiving Suboxone and
`in males receiving ≥500 ppm. However, as there is no known clinical effect of insult to the Harderian
`gland, the toxicological importance of these findings remains unclear. No toxicological relevant target
`
`
`
`©EMEA 2006
`
`8/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`organ toxicity was apparent apart from weight changes of the liver and histological effects on the
`kidneys, probably adaptive effects due to the high doses administered.
`
`Safety margins based on AUC were calculated based on human data from Clinical Study CR97/007:
`
`Rat : human buprenorphine ratios based on AUC (ng·h/ml)
`Table 7
`Dietary concentration
`Buprenorphine dose (mg)
`of Suboxone (ppm)
`
`4
`7.3
`28.2
`73.6
`93.9
`
`8
`4.4
`17.1
`44.7
`57.0
`
`16
`2.5
`9.7
`25.4
`32.4
`
`24
`2.0
`7.6
`20.0
`25.5
`
`6
`NC
`4.4
`68.0
`71.7
`
`100
`500
`1500
`2000
`
`100
`500
`1500
`2000
`
`
`Rat : human naloxone ratios based on AUC (ng·h/ml)
` Table 8
`Naloxone dose (mg)
`Dietary concentration
`of Suboxone (ppm)
`
`1
`NC
`15.1
`231.8
`244.5
`
`2
`NC
`8.5
`130.8
`137.9
`
`4
`NC
`5.0
`76.1
`80.3
`
`NC = Not calculated. Plasma concentration less than LLOQ (0.5 ng/ml).
`
`The toxicokinetic data obtained in rats following dietary administration of buprenorphine: naloxone at
`a ratio of 4:1 also suggests that both rats and dogs receiving development formulations of
`buprenorphine: naloxone at a ratio of 1:1 by the oral route would have received toxicologically
`significant exposures to both of the active ingredients.
`
`No novel toxicological aspects rose from the studies with a mixture of buprenorphine/naloxone in
`comparison with knowledge about the compounds alone. Based on toxicokinetic data raised from the
`dietary study with Suboxone, an exposure of animals sufficiently above the maximum therapeutic
`dose in humans has been reached. Clinical signs reflected the pharmacodynamics of the active
`ingredients.
`
` •
`
` Genotoxicity
`Standard in vitro and in vivo genotoxicity tests with buprenorphine and naloxone were negative
`indicating that both compounds are devoid of genotoxic properties.
`
` A
`
` 7-day dietary palatability study in rats, a subacute 28 day dietary toxicity study in rats and
`associated mutagenicity studies have also been completed in order to investigate the potential toxicity
`and genotoxicity of synthesis impurities and degradants of Suboxone.
`
`Synthesis impurity D (7,8-didehydronaloxone) present in naloxone was found clastogenic in vitro
`studies with human lymphocytes. The proposed specification limit of 7,8-didehydronaloxone will
`result in exposure below the Threshold of Toxicological Concern (TTC) of 1.5 µg/day which is
`recommended for setting acceptable daily intake limits of genotoxic impurities by the EU Draft
`Guideline on the Limits of Genotoxic Impurities (CHMP/SWP/5199/02).
`
` A
`
` series of other synthesis impurities and degradants was reviewed for structure-activity relationship
`and were reported to be devoid of structural alerts. However, the process used to determine alerting
`structures was not fully clear and needed further clarification. The applicant further provided sufficient
`information about the approaches used (in house and at FDA) in assessing structural alerts as part of
`
`
`
`©EMEA 2006
`
`9/42
`
`
`
`TEVA EXHIBIT 1011
`TEVA PHARMACEUTICALS USA, INC. V. RB PHARMACEUTICALS LTD.
`
`

`
`the toxicological qualification of impurities and degradants. The applied approach for qualifying the
`impurities/degradants is acceptable.
`
` Carcinogenicity
`A 2-year dietary carcinogenicity study with Suboxone was conducted in rats at doses of 7, 30 and
`120 mg/kg/day, with estimated exposure multiples of 3 to 75 times, based on a human daily sublingual
`dose of 16 mg calculated on a mg/m² basis. Statistically significant increases in the incidence of
`benign testicular interstitial (Leydig's) cell adenomas were observed in all dosage groups.
`
` •
`
` A
`
` dose-related reduction in the

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