throbber
In Situ Gelling Nasal Inserts for
`Prolonged Drug Delivery
`
`
`
`
`
`Inaugural-Dissertation
`zur Erlangung der Doktorwürde
`im Fachbereich Biologie, Chemie, Pharmazie
`der Freien Universität Berlin
`
`
`
`vorgelegt von
`Ulrike Werner
`
`
`
`Berlin 2003
`
`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 1/200
`
`

`
`
`
`1. Gutachter: Prof. Dr. Roland Bodmeier
`
`2. Gutachter: Prof. Dr. Philippe Maincent
`
`Tag der mündlichen Prüfung: 19.11.2003
`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 2/200
`
`

`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 3/200
`
`

`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 4/200
`
`

`
`Acknowledgements
`
`
`First of all, I wish to express my gratitude to my supervisor, Prof. Dr. Roland Bodmeier, for
`his kind supervision, his helpful advice and professional guidance, for providing this
`interesting research topic and his scientific support. It was a great experience and pleasure to
`study, to work and to do research in his international group.
`
`I deeply thank Prof. Dr. Philippe Maincent (Faculté de Pharmacie, Université Poincaré,
`Nancy, France) for useful discussions and for his evaluation of my thesis. I am also very
`grateful to Dr. Christiane Damgé (Centre Européen d'Etude du Diabète, Université Louis
`Pasteur, Strasbourg, France). The opportunity to work in her research group was extremely
`valuable for me and my studies.
`
`I would like to thank all the helpful people at the Free University Berlin for giving me the
`opportunity to use advanced methods of instrumental analytics and their kind help in data
`interpretation: Priv.-Doz. Dr. Gerd Buntkowsky and Thomas Emmler (Institute of Chemistry)
`for performing the 13C-CP/MAS NMR measurements, Prof. Ronald Gust and Ingo Ott
`(Institute for Pharmacy) for atom absorption spectroscopy studies, Bernhard Behrens
`(Institute for Chemistry) for atom emission spectroscopic measurements, Priv.-Doz. Dr. Wolf-
`Dietrich Hunnius (Institute for Chemistry) for performing FT-Raman investigations, Dr.
`Volker Bähr and Petra Exner (University Hospital B. Franklin) for the support with
`radioimmunoassays, and Dr. Christine Prusas (Institute for Poultry diseases) for providing
`chicken erythrocytes.
`
`Furthermore, I am grateful to Prof. Dr. Ronald Gust, Prof. Dr. Hans Hubert Borchert, Priv.-
`Doz. Dr. Ralph Lipp, Dr. Wolfgang Mehnert and Boris Petri for serving as members of my
`thesis advisor committee.
`
`I sincerely wish to thank Dirk Sticha, Heike Friedrich, Oliver Bley, Katrin Johannsen, Mesut
`Ciper, and all my friends and colleagues in the Kelchstraße for their support, proof-readings,
`constructive discussions in early Monday seminars and the stimulating and enjoyable
`atmosphere in the group. I am grateful to Dr. Jürgen Siepmann for his readiness to discuss my
`work and his valuable suggestions. Special thanks go also to Andreas Krause and Eva Ewest
`for their support and to Angelika Schwarz for her help in the jungle of university bureaucracy.
`
`Finally, I want to thank Matthias and my family for their love and support through the years
`and their never-ending encouragement and confidence in me.
`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 5/200
`
`

`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 6/200
`
`

`
`
`___________________________________________________________________________
`
`
`Table of Contents
`
`
`1. INTRODUCTION………………………………………………………………….. 1
`1.1. Transmucosal routes of drug delivery.…………………………………….….. 1
`1.1.1. Nasal drug delivery…………………………………………………….…. 3
`1.1.2. Buccal drug delivery………………………………………………….…... 4
`1.1.3. Ocular drug delivery…………………………………………………..….. 4
`1.1.4. Pulmonary drug delivery…………………………………………...…..… 6
`1.1.5. Rectal drug delivery………………………………………………….…... 7
`1.1.6. Vaginal drug delivery………………………………………………….…. 8
`1.1.7. Comparison of transmucosal drug delivery routes…………………….…. 8
`1.2. Nasal anatomy and physiology……………………………………………….... 12
`1.2.1. Anatomy and air passage……………………………………………….… 12
`1.2.2. Physiology…………………………………………………………….….. 13
`1.2.3. Nasal pathology with relevance to nasal drug absorption…………….….. 20
`1.2.4. The nose as drug delivery site: advantages, barriers, and solutions….…... 21
`1.3. The concept of bioadhesion……………………………………………………. 24
`1.4. Nasal vaccination…………………………………………………………….…. 27
`1.4.1. Nasal immunology…………………………………………………….….. 27
`1.4.2. Nasal vaccine delivery……………………………………………….….... 29
`1.5. Nasal drug delivery systems………………………………………………….... 34
`1.5.1. Nasal solutions as drops or sprays………………………………………... 36
`1.5.2. Viscous nasal solutions and gels including bioadhesive solutions……….. 38
`1.5.3. Nasal suspensions and emulsions…………………………………….…... 41
`1.5.4. Nasal micellar and liposomal formulations…………………………….… 42
`1.5.5. Nasal powders and microparticles………………………………………... 43
`1.5.6. Nasal solid dosage forms……………………………………………….… 48
`1.6. Research objectives…………………………………………………………….. 49
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 7/200
`
`

`
`Table of Contents
`___________________________________________________________________________
`
`2. MATERIALS AND METHODS………………………………………………….... 51
`2.1. Materials……………………………………………………………………...… 51
`2.2. Methods……………………………………………………………………….… 52
`2.2.1. General data presentation in diagrams and tables………………………... 52
`2.2.2. Preparation of samples………………………………………………….... 52
`2.2.3. Polymer solution rheology………………………………………….……. 54
`2.2.4. Spreading of polymer solutions……………………………………….….. 55
`2.2.5. Bioadhesion…………………………………………………………….… 55
`2.2.6. Water uptake and mass loss…………………………………………….… 55
`2.2.7. Moisture sorption……………………………………………………….… 56
`2.2.8. Contact angle……………………………………………………………... 57
`2.2.9. Polymer-drug precipitation…………………………………………….…. 58
`2.2.10. In vitro drug release…………………………………………………….… 58
`2.2.11. Release of MβCD……………………………………………………….... 62
`2.2.12. Flame absorption and flame emission spectroscopy…………….……….. 62
`2.2.13. Scanning electron microscopy (SEM)……………………….…………… 63
`2.2.14. Differential scanning calorimetry (DSC)………………….……………... 63
`2.2.15. Powder X-ray diffraction………………………………………….……… 63
`2.2.16. 13C Nuclear magnetic resonance spectroscopy (13C CP/MAS NMR)…..... 64
`2.2.17. FT-Raman spectroscopy…………………………………………………. 64
`2.2.18. Mechanical properties………………………………………………….… 64
`2.2.19. Hemagglutination test………………………………………………….…. 65
`2.2.20. In vivo studies………………………………………………………….…. 65
`
`3. RESULTS AND DISCUSSION………………………………………………….…. 69
`3.1. Influence of the polymer type……………………………………………….…. 69
`3.1.1. Sponge-structure formation……………………………………………..... 69
`3.1.2. Polymer solution rheology…………………………………………….…. 71
`3.1.3. Effect of polymer type on nasal insert properties…………………….…... 75
`3.2. Drug release rate control………………………………………………….…… 85
`3.2.1. Polymer content…………………………………………………………... 85
`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 8/200
`
`

`
`Table of Contents
`___________________________________________________________________________
`
`3.2.2. Freely water-soluble additives……………………………………………. 88
`3.2.3. Polymer molecular weight………………………………………………... 91
`3.2.4. Polymer blends………………………………………………………….... 99
`3.3. Effect of drug and release medium on drug release……………………….…. 108
`3.3.1. Influence of drug species and drug loading…………………………….… 108
`3.3.2. Influence of release medium……………………………………………... 115
`3.4. Estradiol delivery…………………………………………………………….… 121
`3.5. Nasal influenza vaccination………………………………………………….… 138
`3.5.1. In vitro properties of polymer solutions………….…………...……….…. 138
`3.5.2. In vivo performance of polymer solutions………………….…………..... 143
`3.5.3. Formulation and in vitro properties of nasal inserts………………..…….. 146
`3.5.4. Hemagglutination activity in solutions and inserts………………………. 149
`3.5.5. In vivo performance of nasal inserts……………………………………... 151
`
`
`4. SUMMARY……………………………………………………………………….…. 155
`
`5. ZUSAMMENFASSUNG………………………………………………………….… 161
`
`6. BIBLIOGRAPHY………………………………………………………………….... 167
`
`LIST OF PUBLICATIONS FROM THIS WORK………………………………….... 189
`
`CURRICULUM VITAE…………………………………………………………….….. 191
`
`
`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 9/200
`
`

`
`Table of Contents
`___________________________________________________________________________
`
`
`
`
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 10/200
`
`

`
`
`___________________________________________________________________________
`
`
`1. Introduction
`
`
`
`The aim of this work was the development and characterization of in situ gelling nasal inserts,
`a new, bioadhesive, solid dosage form for the prolonged systemic drug delivery via the nasal
`route. In this chapter conventional and alternative mucosal routes for systemic drug delivery
`are compared (section 1.1) and a basic introduction to the anatomy and physiology of the
`human nose (section 1.2) and to nasal immunology (section 1.4.1) is given. Further on, the
`concept of bioadhesion will be introduced (section 1.3) as a major approach to increase the
`nasal residence time of drug delivery systems. The current state of research concerning nasal
`drug delivery systems and nasal immunization approaches (sections 1.5 and 1.4.2) is
`presented. Finally, the objectives of this work are laid out.
`
`
`1.1. Transmucosal routes of drug delivery
`
`Drugs for systemic medication are administered traditionally and routinely by oral and by
`parenteral routes. Although generally convenient, both routes have a number of
`disadvantages, especially for the delivery of peptides and proteins, a class of drugs that has
`been rapidly emerging over the last decades (Zhou and Li Wan Po, 1991a). Oral
`administration results in the exposure of the drug to the harsh environment of the
`gastrointestinal tract and thus to potential chemical and enzymatic degradation (Langguth et
`al., 1997). After gastrointestinal absorption the drug has to pass the liver, where, dependent on
`the nature of the drug, extensive first pass metabolism can take place with subsequent rapid
`clearance from the blood stream (Lalka et al., 1993; Taki et al., 1998). Low permeability
`across the gastrointestinal mucosa is also often encountered for macromolecular drugs
`(Yamamoto et al., 2001; Pauletti et al., 1997). Parenteral administration avoids drug
`degradation in the gastrointestinal tract and hepatic first pass clearance but due to pain or
`discomfort during injection, patient compliance is poor, particularly if multiple daily
`injections are required as e.g. in the insulin therapy (Hinchcliffe and Illum, 1999). Also
`injection related side effects like tissue necrosis and thrombophlebitis lead to low patient
`
`1
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 11/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`acceptability (Zhou, 1994). In addition, administration by injection requires trained personnel
`which adds to the relatively high costs of parenteral medication.
`
`
`
`Figure 1. 1 Various potential mucosal pathways for systemic delivery of therapeutic
`agents, which bypass the hepatic first pass clearance associated with oral
`administration. The venous drainage system involved in the systemic delivery
`of therapeutic agents following the transmucosal permeation is illustrated
`(Zhou and Li Wan Po, 1991b).
`
`
`
`2
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 12/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`Several mucosal routes have been investigated over the last decades as alternatives to oral and
`parenteral drug administration, including nasal, buccal, rectal, ocular, pulmonary, and vaginal
`mucosa (Banga and Chien, 1988; Zhou and Li Wan Po, 1991b). Their advantages are the easy
`accessibility and circumvention of the hepatic first pass metabolism (Figure 1. 1). Mucosal
`bioavailability can vary between almost 100% for low molecular weight hydrophobic drugs
`(Striebel et al., 1993; Hussain et al., 1980) and below 1% for polar macromolecules (Zhou
`and Li Wan Po, 1991b; Illum, 2003) depending on the nature of the delivered drug. In the
`following, a short overview over the different alternative mucosal drug delivery routes is
`given.
`
`
`
`1.1.1. Nasal drug delivery
`
`The nasal route of administration, which is in the focus of this work, has received a great deal
`of attention in recent years as a convenient and reliable method not only for local but also for
`systemic administration of drugs (Schipper et al., 1991; Sakar, 1992; Merkus and Verhoef,
`1994; Kublik and Vidgren, 1998; Marttin et al., 1998; Davis, 1999; Hinchcliffe and Illum,
`1999; Martini et al., 2000; Chow et al., 2001; Illum, 2003). The nasal cavity offers a number
`of unique advantages such as easy accessibility, good permeability especially for lipophilic,
`low molecular weight drugs, avoidance of harsh environmental conditions and hepatic first
`pass metabolism, potential direct delivery to the brain, and direct contact for vaccines with
`lymphatic tissue and action as inducer as well as effector of the mucosal immune system (see
`section 1.2.4). The nasal epithelium is well suited for the transmucosal drug delivery although
`it is less permeable for hydrophilic and high molecular weight drugs (see section 1.1.7).
`Ciliary movement and the resulting clearance of the delivered drug / dosage form towards the
`throat are challenges when developing a prolonged release dosage form (see sections 1.2.2
`and 1.2.4). Also a considerable enzyme activity, though lower than in the gastrointestinal
`tract, must be considered. Nevertheless, a number of approaches have been used to overcome
`these limitations such as the use of bioadhesive formulations to increase the nasal residence
`time of dosage forms (Morimoto et al., 1991; Soane et al., 2001), addition of absorption
`enhancers to increase the membrane permeability (De Ponti, 1991; Merkus et al., 1993; Illum,
`1999, Natsume et al., 1999), and the use of protease / peptidase inhibitors to avoid enzymatic
`degradation of peptide and protein drugs in the nasal cavity (Morimoto et al., 1995; Dondeti
`et al., 1996). Several nasal dosage forms are under investigation including solutions (drops or
`
`
`
`3
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 13/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`sprays), gels, suspensions and emulsions, liposomal preparations, powders and microspheres,
`as well as inserts (see section 1.5).
`
`
`
`1.1.2. Buccal drug delivery
`
`The oral cavity is lined by a stratified squamous epithelium. The epithelium has a cornified
`surface in regions subject to mechanical forces during mastication, which resembles that of
`the upper epidermis in the skin. Non-keratinized epithelium occupies approximately 60% of
`the total oral cavity including the buccal, lingual, and sublingual mucosa (Chien, 1995;
`Hoogstraate and Wertz, 1998) and is of interest for systemic drug delivery. Although non-
`keratinized, the buccal mucosa contains intercellular lipids which are responsible for its
`physical barrier properties (Hoogstraate and Wertz, 1998; Shojaei, 1998), resulting in poor
`permeability for larger drugs, especially for peptides and proteins (Junginger et al., 1999;
`Veuillez et al., 2001). Transfer of peptides with molecular weights above 500 - 1000 Da
`through buccal mucosa would require use of an absorption enhancer (Merkle et al., 1992).
`Another limitation in buccal drug delivery is the mucosal enzyme activity, especially of
`proteases (Bird et al., 2001; Veuillez et al., 2001; Walzer et al., 2002). The reduced retention
`of the dosage form at the buccal surface due to constant washing with saliva can be overcome
`by the use of bioadhesive formulations (Shojaei, 1998; Veuillez et al., 2001; Langoth et al.,
`2003). The influence of food intake and mastication on the residence time of bioadhesive
`buccal formulations is so far not clear. Thiolated polymers can be used simultaneously as
`bioadhesive carrier and protease inhibitors (Langoth et al., 2003). The sublingual epithelium
`is more permeable than the buccal one but more handicapped by the saliva washing and
`constant mobility (Shojaei, 1998). It is therefore more suitable for immediate release
`products.
`
`Dosage forms for buccal drug delivery include tablets, patches, films, lozenges, sprays,
`hydrogels, lollypops, chewing gums, powders, solutions (Hoogstraate and Wertz, 1998), a
`freeze-dried sublingual dosage form (Vaugelade et al., 2001), wafers (Kalra et al., 2001), and
`liposomal formulations (Veuillez et al., 2001).
`
`
`
`1.1.3. Ocular drug delivery
`
`Ocular delivery of drugs is typically for the treatment of ocular inflammation, corneal
`wounds, and glaucoma. In addition, this route has been investigated for the systemic delivery
`
`
`
`4
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 14/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`of peptides and proteins. Already in 1931, ocular administration of insulin produced sustained
`lowering of the blood glucose level in proportion to the dose instilled (Christie and Hanzal,
`1931). However, today it is known that the majority of the systemic drug absorption after
`ocular instillation takes place across the nasal mucosa after drainage via the nasolachrymal
`duct (Lee et al., 2002). Some absorption occurs also from the conjunctival sac. Drug
`absorption via the cornea is relatively low due to the lipophilicity of the corneal epithelium,
`dilution of the drug in the tear fluid (reflex tearing and reflex blinking) and drug binding to
`proteins in tear fluid (Zhou and Li Wan Po, 1991b). In addition, the corneal and conjunctival
`tissues act also as enzymatic barrier, which contain e.g. proteases (Zhou and Li Wan Po,
`1991b). Therefore, the eye offers no additional advantage over the nose as systemic drug
`delivery site and is of higher interest only for drug administration for local (ophthalmic)
`therapy. However, also the local drug delivery is restricted by the dynamics of the lachrymal
`drainage system, which is the natural defense mechanism of the eye. This system introduces
`tear fluid to the eye and rapidly drains the fluid together with any instilled formulation from
`the precorneal area to the nasal cavity and throat. The high elimination rate results in short
`duration of contact of the drug with its absorption sites and consequently in a low local
`bioavailability. Increased ocular bioavailability can be achieved by the use of viscosity
`enhanced aqueous eye drops, suspensions, oily drops and unguents, mucoadhesive ocular
`delivery systems such as solutions and microparticle suspensions, in-situ gelling systems
`triggered by pH, temperature, or ions, colloidal delivery systems such as liposomes and
`nanoparticles, and ocular inserts (Le Bourlais et al., 1995). Ocular inserts can be divided into
`non-erodible (Chetoni et al., 1998; Kawakami et al., 2001) and erodible inserts. Erodible
`ocular inserts, which do not need to be removed mechanically from the eye, have been
`prepared by powder compression from poly(ethylene oxide) (Di Colo et al., 2001), from
`bioadhesive mixtures of poly(ethylene oxide) with chitosan hydrochloride (Di Colo et al.,
`2002), and from mixtures of Carbopol® 974P with drum dried waxy maize starch (Ceulemans
`et al., 2001; Weyenberg et al., 2003). Also absorbable gelatin sponge (Gelfoam®) soaked with
`an organic drug solution and subsequent solvent removal has been used as erodible ocular
`insert with improved bioavailability compared to eye drops and gels (Simomara et al., 1998).
`Finally, ocular inserts have also been prepared by freeze-drying aqueous solutions of water
`soluble polymers such as HPMC resulting in a sponge-like structure (Diestelhorst et al., 1999;
`Lux et al., 2003).
`
`
`
`
`
`5
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 15/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`1.1.4. Pulmonary drug delivery
`
`Pulmonary drug delivery has traditionally been used for the systemic administration of drugs
`such as anesthetic gases and nicotine (tobacco smoke). Direct delivery of drugs to the lung by
`inhalation for the local treatment of respiratory diseases grew rapidly in the second half of the
`20th century as a result of the availability of effective asthma drugs in convenient, portable
`devices (Gonda, 2000). The lung offers a number of advantages which render it also a suitable
`organ for systemic drug delivery: a large surface area of about 150 m2 and an extremely well
`vascularized, thin epithelium. Thus, various drugs including peptides and proteins (e.g.
`insulin, human growth hormone, luteinizing hormone releasing hormone analogue, glucagon,
`calcitonin) have efficiently been delivered via the lung (Qiu et al., 1997; Adjei and Gupta,
`1998; Edwards et al., 1998). A number of technologies for the delivery of drug formulations
`have been developed (Martini et al., 2000): (i) pressurized metered dose inhalers using
`propellants to deliver micronized drug suspensions (Autohaler®, Spacehaler®), (ii) dry powder
`inhalers which dispense micronized drug particles with / without carrier (lactose) by
`inhalation activation (Spinhaler®, Rotohaler®, Diskhaler®), and (iii) nebulizers and aqueous
`mist inhalers which aerosolize drug solutions using compressed air or ultrasound (AERx®,
`Respimat®). Although the pulmonary route of administration is very promising and the
`available delivery technologies are highly sophisticated, systemic drug delivery via the lung is
`still a challenging area of research. A key issue is the achievement of high delivery efficiency
`to the alveolar region. However, this is handicapped by the 90° bend in the oropharynx and
`the concomitant branching and narrowing of the bronchial tree (Malcolmson and Embleton,
`1998). The particle size should be in the aerodynamic diameter window of 0.5 - 5 µm, ideally
`2 - 3 µm, for deep lung delivery to avoid loss of delivered particles by impaction onto the
`mucus lined epithelia. The aerodynamic diameter relates the geometric particle diameter and
`the particle mass density. Thus, large porous particles are effective means for drug delivery to
`the alveolar region (Edwards et al., 1997; Vanbever et al., 1999; Crowder et al., 2002). Even
`optimized aerosol particles can be deposited in mouth and throat by inertia when delivered
`with too high a velocity (Edwards et al., 1998). In addition, the high humidity in the airways
`furthers particle agglomeration, thus decreasing the delivery efficiency due to hygroscopic
`growth (Malcolmson and Embleton, 1998; Crowder et al., 2002). Once in the lung, the
`particles must release the therapeutic substance at a desired rate and, in some case, escape the
`lung’s natural cleaning mechanisms (mucociliary transport in the conducting airways and
`phagocytosis by macrophages in the alveoli) until their therapeutic payload has been delivered
`(Kim and Folinsbee, 1997). Prolonged drug action after pulmonary delivery is another
`
`
`
`6
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 16/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`challenge in pulmonary drug delivery and is approached by polymeric particle formulations
`(Kawashima et al., 1999; Zhang et al., 2001), mucoadhesive formulations (Takeuchi et al.,
`2001), and protein crystal formulations (Tam et al., 2001). However, in that case
`accumulation of polymeric material in the alveoli has to be taken into consideration as well as
`the possible delivery related development of fibrosis. Finally, the lung contains high levels of
`hydrolytic and other enzymes, which can become significant absorption barriers to drugs,
`although the metabolic activity of the lung is much lower than in the gastrointestinal tract.
`Numerous endoproteases and exoproteases were identified in lung tissue and in the bronchial
`lavage fluid (Adjei, 1997).
`
`
`
`1.1.5. Rectal drug delivery
`
`The lower digestive tract is less harmful to administered drugs than the stomach and the small
`intestine due to the lower enzymatic activity and neutral pH. Also the rectal route of drug
`administration is safe and convenient. In several countries it is generally accepted, especially
`for infants (Lejus et al., 1997; Jensen and Matsson, 2002), although the acceptance can be low
`in other states, particularly among adults. This may be overcome by the use of colon-specific
`drug targeting via the peroral route, which is under intensive investigation (Sinha and Kumria,
`2001; Raghavan et al., 2002) but not within the scope of this work. The adult’s lower intestine
`has also been shown to be relatively impermeable for macromolecules such as high molecular
`weight protein drugs and heparin (Warshaw et al., 1977; Zhou and Li Wan Po, 1991b;
`Lohikangas et al., 1994). Also a considerable protease activity still exists in the rectum and is
`still enhanced by the presence of bacterial flora (Lewin et al., 1986; Hacker et al., 1991; Zhou
`and Li Wan Po, 1991b). Additionally, the circumvention of the hepatic first pass metabolism
`by rectal administration is only partial and depends on the positioning and / or spreading of
`the drug formulation (de Boer and Breimer, 1997; Kurosawa et al., 1998).
`
`Traditional rectal dosage forms are suppositories, unguents and cremes, as well as enemas.
`More recent studies have evaluated thermogelling dosage forms (Miyazaki et al., 1998), gels
`(de Leede et al., 1986), osmotic mini pumps (Teunissen et al., 1985), and hard gelatin
`capsules (Eerikainen et al., 1996) as rectal drug delivery systems. Strategies to improve the
`rectal bioavailability of peptide and protein drugs include the use of absorption enhancers, the
`use of protease inhibitors and structural modifications of peptide and protein drugs
`(Yamamoto and Muranishi, 1997).
`
`
`
`7
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 17/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`1.1.6. Vaginal drug delivery
`
`It has been known for decades that a number of therapeutic agents, such as steroids, can be
`effectively absorbed through the vaginal mucosa (Ho et al., 1976; Alvarez et al., 1983).
`Traditionally, the vagina has been used for the delivery of locally acting drugs such as
`antibacterial, antifungal, antiprotozoal, antiviral, labor-inducing, and spermicidal agents,
`prostaglandins, and steroids (Vermani and Garg, 2000). The large surface area, rich blood
`supply and permeability to a wide range of compounds including peptides and proteins make
`the vagina also attractive for systemic drug administration (Benziger and Edelson, 1983;
`Honkanen et al., 2002; Valenta et al., 2002). The vaginal route has also the potential for
`uterine targeting of active agents such as progesterone and danazol (Bulletti et al., 1997;
`Cicinelli et al., 1998). Commonly used dosage forms are creams, gels, tablets, capsules,
`pessaries, foams, films, tampons, vaginal rings, and douches (Vermani and Garg, 2000). The
`vagina as drug delivery site has a number of unique features which have to be considered
`during the development of dosage forms. The vaginal pH of usually 4 - 5 is maintained by
`lactobacilli which convert glycogen into lactic acid. However, it changes with age, stage of
`menstrual cycle, infections, and sexual arousal (Vermani and Garg, 2000). The variation in
`vaginal pH and secretions may affect the absorption of pH-sensitive and / or solubility-
`dependent therapeutic agents (Chien, 1995). The vaginal microflora is also influenced by a
`number of factors (glycogen content of epithelial cells, pH, hormone levels, birth control
`method etc.) and can potentially contribute to enzymatic drug degradation in addition to the
`membrane-bound enzymes of the vaginal mucosa (Chien, 1995; Vermani and Garg, 2000).
`The changes in the hormone levels during the menstrual cycle vary also the enzyme activity
`of the mucosa as well as the thickness of the epithelial layer and width of the intercellular
`channels (Vermani and Garg, 2000). Limitations of systemic vaginal drug delivery next to the
`physiological barriers are also the gender specificity and the relatively low convenience.
`
`
`
`1.1.7. Comparison of transmucosal drug delivery routes
`
`With nasal, buccal, pulmonary, ocular, rectal, and vaginal mucosa as potential drug delivery
`sites, it is hard to identify the most suitable for clinical use. Only few studies were conducted
`to directly evaluate the different membrane permeabilities between these mucosal sites.
`Rojanaskul et al. (1992) measured the electrical membrane resistance and the flux of the
`hydrophilic probe 6-carboxyfuorescein at various mucosal sites and found a good correlation
`between these two parameters (Table 1. 1). The data indicates that nasal and pulmonary
`
`
`
`8
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 18/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`epithelia are equally or only slightly less permeable than that of the intestine. The high
`permeability values of the respiratory tissue are a result of the presence of numerous aqueous
`pores through which water-soluble molecules can diffuse. Both large and small pores were
`reported in the nasal and pulmonary epithelium. The aqueous pores in the nasal epithelium,
`particularly those of small size (0.4 - 0.8 nm), were found to be more abundant than those
`observed in the jejunum (0.7 - 1.6 nm) (Hayashi et al., 1985). In the pulmonary epithelium,
`pores of 0.6 - 1.0 nm size and large pores of ≥ 8 nm were reported (Taylor and Gaar, 1970).
`
`Table 1. 1 Membrane electrical resistance and flux of 6-carboxyfluorescein of various
`mucosal sites (mean ± SD, n = 6)(Rojanaskul et al., 1992).
`
`Membrane
`tissue
`
`Membrane electrical resistance,
`Ω·cm2
`
`Steady state flux of 6-
`carboxyfluorescein, 106 µg/cm2·h
`
`Skin
`
`Buccal
`
`Corneal
`
`Rectal
`
`Vaginal
`
`Tracheal
`
`Colonic
`
`Bronchial
`
`Ileal
`
`Nasal
`
`Jejunal
`
`Duodenal
`
`
`
`9703 ± 175
`
`1803 ± 175
`
`1012 ± 106
`
`406 ± 70
`
`372 ± 85
`
`291 ± 65
`
`288 ± 72
`
`266 ± 97
`
`266 ± 95
`
`261 ± 55
`
` 224 ± 104
`
`211 ± 91
`
`9
`
` 0.5 ± 0.4
`
` 3.0 ± 1.3
`
` 5.1 ± 1.7
`
` 9.9 ± 2.3
`
`12.4 ± 4.1
`
`14.2 ± 5.4
`
`16.3 ± 6.8
`
`16.7 ± 4.5
`
`19.6 ± 3.9
`
`16.8 ± 1.8
`
`21.1 ± 6.2
`
`21.0 ± 3.9
`
`Teva Pharm. v. Indivior, IPR2016-00280
`INDIVIOR EX. 2024 - 19/200
`
`

`
`Introduction
`1.
`___________________________________________________________________________
`
`The nasal epithelium is leakier for peptide molecules than intestinal epithelia when using
`metabolically stable peptides as permeability tracers (McMartin et al., 1987). Opposite to
`other reports with mannitol and progesterone (Corbo et al., 1990) and 6-carboxyfluorescein
`(Rojanaskul et al., 1992), Aungst et al. (1988) demonstrated that nasal, buccal, and sublingual
`insulin administration were less efficient than administration via rectal mucosa. This finding
`suggests that also other factors like enzyme activity and absorptive surface area may play a
`role in determining the overall bioavailability. The large absorptive surface of the lung would
`make the pulmonary mucosa a very effective route of administration. This was also
`demonstrated by an absorption study in rats with different water-soluble compounds (Phenol
`Red, Trypan Blue, fluorescein isothiocyanate dextran molecular weight 4400 and 9100)
`which revealed bioavailabilities after mucosal administration of the order lung > small
`intestine > nasal cavity > large intestine > buccal cavity (Yamamoto et al., 2001). In the same
`study the pharmacological availability of [ASU1.7]-eel calcitonin gave the order lung > nasal
`cavity > small intestine = large intestine ≥ buccal cavity which was attributed to the higher
`protease content in the small intestine compared to the nasal cavity. The proteolytic activity in
`different animals is relatively high in the rectal and ileal mucosa and comparatively low in the
`buccal, nasal, and vaginal mucosal tissue (Table 1. 2) (Zhou and Li Wan Po, 1991b).
`
`Due to the clear advantages of accessibility, patient convenience, and permeability, nasal and
`pulmonary drug delivery are the most promising transmucosal delivery routes. The quantity
`of drug that can be delivered to the lung may be more limiting than that given nasally

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