throbber
ALL 2015
`MYLAN PHARMACEUTICALS V. ALLERGAN
`IPR2016-01128
`
`1
`
`ALL 2015
`MYLAN PHARMACEUTICALS V. ALLERGAN
`IPR2016-01128
`
`

`

`Mucosal Delivery of Biopharmaceuticals
`
`2
`
`

`

`José das Neves • Bruno Sarmento
`Editors
`
`Mucosal Delivery
`of Biopharmaceuticals
`
`Biology, Challenges and Strategies
`
`2123
`
`3
`
`

`

`Editors
`José das Neves
`IINFACTS – Instituto de Investigação e Formação
`Avançada em Ciências e Tecnologias da Saúde
`Instituto Superior de Ciências da
`Saúde-Norte, CESPU
`Gandra, Portugal
`
`Bruno Sarmento
`NEWTherapies Group
`INEB – Instituto de Engenharia
`Biomédica
`Porto, Portugal
`
`ISBN 978-1-4614-9523-9
`DOI 10.1007/978-1-4614-9524-6
`Springer New York Heidelberg Dordrecht London
`
`ISBN 978-1-4614-9524-6 (eBook)
`
`Library of Congress Control Number: 2013958086
`© Springer Science+Business Media New York 2014
`This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the
`material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
`broadcasting, reproduction on microfilms or in any other physical way, and transmission or information
`storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology
`now known or hereafter developed. Exempted from this legal reservation are brief excerpts in connection
`with reviews or scholarly analysis or material supplied specifically for the purpose of being entered and
`executed on a computer system, for exclusive use by the purchaser of the work. Duplication of this
`publication or parts thereof is permitted only under the provisions of the Copyright Law of the Publisher’s
`location, in its current version, and permission for use must always be obtained from Springer. Permissions
`for use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable to
`prosecution under the respective Copyright Law.
`The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
`does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
`protective laws and regulations and therefore free for general use.
`While the advice and information in this book are believed to be true and accurate at the date of publication,
`neither the authors nor the editors nor the publisher can accept any legal responsibility for any errors or
`omissions that may be made. The publisher makes no warranty, express or implied, with respect to the
`material contained herein.
`
`Printed on acid-free paper
`Springer is part of Springer Science+Business Media (www.springer.com)
`
`4
`
`

`

`Contents
`
`Part I Biology of Mucosal Sites
`
`1 Concepts in Mucosal Immunity and Mucosal Vaccines . . . . . . . . . . . . .
`Simona Gallorini, Derek T. O’Hagan and Barbara C. Baudner
`
`2 Mucoadhesion and Characterization of Mucoadhesive Properties . . . .
`Tao Yu, Gavin P. Andrews and David S. Jones
`
`3 Mucus as a Barrier for Biopharmaceuticals and Drug
`Delivery Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Hongbo Zhang, Mohammed-Ali Shahbazi, Patrick V. Almeida
`and Hélder A. Santos
`
`4 Epithelial Permeation and Absorption Mechanisms
`of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Hanne Mørck Nielsen
`
`3
`
`35
`
`59
`
`99
`
`Part II Delivery Strategies for Specific Mucosal Sites
`
`5 Oral Delivery of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
`Catarina Oliveira Silva, Bruno Sarmento and Catarina Pinto Reis
`
`6 Buccal Delivery of Biopharmaceuticals: Vaccines and Allergens . . . . . 149
`Sevda ¸Senel, Merve Cansız and Michael J. Rathbone
`
`7 Pulmonary Delivery of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . 169
`Fernanda Andrade, Catarina Moura and Bruno Sarmento
`
`8 Nasal Delivery of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197
`Eiji Yuba and Kenji Kono
`
`9 Ocular Delivery of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
`Holly Lorentz and Heather Sheardown
`
`ix
`
`5
`
`

`

`x
`
`Contents
`
`10 Vaginal Delivery of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . 261
`José das Neves
`
`Part III Case Studies of Mucosal Delivery of Biopharmaceuticals
`
`11 Nanoparticles-in-Microsphere Oral Delivery Systems (NiMOS)
`for Nucleic Acid Therapy in the Gastrointestinal Tract . . . . . . . . . . . . . 283
`Shardool Jain and Mansoor Amiji
`
`12 Bacteria-Based Vectors for Oral Gene Therapy . . . . . . . . . . . . . . . . . . . . 313
`Yong Bai, Rachael Burchfield, Sangwei Lu and Fenyong Liu
`
`13 Self-Assembled Polysaccharide Nanogels for Nasal Delivery
`of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 325
`Tomonori Nochi, Yoshikazu Yuki, Kazunari Akiyoshi and Hiroshi Kiyono
`
`14 PheroidTM Vesicles and Microsponges for Nasal Delivery
`of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 333
`Lissinda H. du Plessis and Awie F. Kotzé
`
`15 Delivery Strategies for Developing siRNA-Based
`Vaginal Microbicides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345
`Joseph A. Katakowski and Deborah Palliser
`
`16 Delivery Strategies for Developing Vaginal DNA Vaccine Combining
`Cell-Penetrating Peptide and Jet Injection . . . . . . . . . . . . . . . . . . . . . . . . 367
`Takanori Kanazawa and Hiroaki Okada
`
`17 Vaccine Delivery Systems for Veterinary Immunization . . . . . . . . . . . . . 379
`Juan M. Irache, Ana I. Camacho and Carlos Gamazo
`
`18 Eligen® Technology for Oral Delivery of Proteins and Peptides . . . . . . 407
`Sunita Prem Victor, Willi Paul and Chandra P. Sharma
`
`19 The RapidMistTM System for Buccal Delivery of Insulin . . . . . . . . . . . . 423
`Meena Bansal, Sanjay Bansal and Rachna Kumria
`
`20 The Pharmaceutical Development of rhDNase (Dornase Alpha)
`for the Treatment of Cystic Fibrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 437
`Steven J. Shire and Thomas M. Scherer
`
`21 Development of the Exubera® Insulin Pulmonary Delivery System . . . 461
`Cynthia L. Stevenson and David B. Bennett
`
`22 Technosphere®: An Inhalation System for Pulmonary Delivery
`of Biopharmaceuticals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 483
`António J. Almeida and Ana Grenha
`
`6
`
`

`

`Contents
`
`xi
`
`23 ChiSys® as a Chitosan-Based Delivery Platform
`for Nasal Vaccination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499
`Peter Watts, Alan Smith and Michael Hinchcliffe
`
`24 Development of a Cationic Nanoemulsion Platform (Novasorb®)
`for Ocular Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
`Frédéric Lallemand, Philippe Daull and Jean-Sébastien Garrigue
`
`Part IV Regulatory, Toxicological and Market Issues
`
`25 Regulatory Aspects and Approval of Biopharmaceuticals
`for Mucosal Delivery: Quality, Toxicology, and Clinical Aspects . . . . . 539
`Karen Brigitta Goetz, Yuansheng Sun, Katrin Féchir,
`Evelyne Kretzschmar and Isabel Buettel
`
`Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 591
`
`7
`
`

`

`Chapter 24
`Development of a Cationic Nanoemulsion
`Platform (Novasorb®) for Ocular Delivery
`
`Frédéric Lallemand, Philippe Daull and Jean-Sébastien Garrigue
`
`24.1 Introduction
`
`Drug delivery across mucosal barriers has always been a challenge, and crossing the
`eye mucosa is no exception. The eye surface is a unique and complex mucosa with
`its own physiology and mechanisms of protection. This chapter illustrates how, from
`a clear understanding of the eye mucosal barrier structure, a new delivery system
`was designed to better treat ocular surface diseases. This case study describes the
`development of a new drug delivery system, the Novasorb® platform, designed to
`overcome ocular barriers to improve ophthalmic drug delivery. This technology is
`based on cationic emulsions primarily developed in the late 1990s by University of
`Jerusalem professor Simon Benita. Several years later, and after the creation of a spin-
`off company, Novagali Pharma (Evry, France) in 2001, the Novasorb® technology
`was successfully transferred to clinical use. The main steps of the development are
`briefly presented; from concept formulations to preclinical pharmacokinetics (PK)
`and toxicity studies to the clinic.
`
`24.1.1 Eye Protection Systems
`
`Of the sensory organs, the eye is probably the most precious and the organ upon which
`our daily activities depend most. As an extension of the central nervous system, the
`eye needs to be well protected although continuously exposed to and threatened by
`an external and aggressive environment. As a consequence, the body has developed
`several effective protection mechanisms to preserve the eye’s structure and function.
`However, these protection mechanisms make it particularly difficult to access the
`eye’s inner tissues, even the different corneal layers, when treating the eye becomes
`necessary.
`J.-S. Garrigue ((cid:2)) · F. Lallemand · P. Daull
`Novagali Pharma, 1 rue Pierre Fontaine, 91058 Evry Cedex, France
`e-mail: jean-sebastien.garrigue@santen.fr
`
`J. das Neves, B. Sarmento (eds.), Mucosal Delivery
`of Biopharmaceuticals, DOI 10.1007/978-1-4614-9524-6_24,
`© Springer Science+Business Media New York 2014
`
`517
`
`8
`
`

`

`518
`
`F. Lallemand et al.
`
`For other mucosal tissues (pulmonary, nasal, intestine, etc.), mucus production
`and excretion is one of the main protection mechanisms, primarily by preventing
`pathogen adherence and tissue infection through their entrapment in a thick external
`mucus layer. For example, the thickness of the mucus layer varies from 100 μm in the
`large intestine to only 0.5 μm on the ocular surface. Hence, the membrane-tethered
`mucins (the mucus layer) of the ocular surface cannot be the only mechanism of
`protection.
`The ocular mucosa is protected by both physical and dynamic mechanisms. The
`eye is firstly protected by the eyelid, which blinks every 4–5 s. The eyelids possess
`several important ocular functions, with the primary objective of protecting both the
`anterior globe (cornea) from injury and the retina from excessive incoming light.
`Eyelid blinking also helps spread and maintain the ocular tear film. Eyelid behaviors
`achieving these functions include blinking (voluntary, spontaneous, or reflexive) and
`voluntary eye closure (gentle or forced) [1]. Blinking swipes away the overloads of
`tears as well as xenobiotic or solid particles present on the ocular surface, including
`any active ingredients administered topically.
`The precorneal tear film is also of major importance in the protection and health of
`the eye. The tear film is a nourishing, lubricating, and protecting layer that bathes the
`ocular surface. It is continuously replenished through cycles of production and elim-
`ination via evaporation, absorption, and drainage. These processes are often referred
`to as tear-film dynamics [2]. Tears fight desiccation, microbial contamination, and
`the effects of xenobiotic and solid particles. This film also maintains surface humid-
`ity to provide transparency and optical quality of the cornea as a refracting surface
`[3]. The tear film comprises three layers: a thin superficial layer of meibomian lipid;
`an intermediate aqueous layer containing dissolved mucins, salts and proteins; and
`an internal layer of mucus network, secreted mainly from conjunctival goblet cells
`whose chemical structure has now been fully described [4]. These ocular mucins are
`highly negatively charged, with the majority terminated by sialic acid, while those
`from rabbits are mainly neutral and terminated by alpha 1-2 fucose and/or alpha
`1-3 N-acetylgalactosamine [3]. In addition, these O-glycan mucins prevent bacte-
`rial adhesion and endocytic activity and maintain epithelial barrier function through
`interactions with galectins [5]. However, in addition to their traditional protective
`functions (selective barrier to the penetration of xenobiotics, antiadhesive that pre-
`vent pathogen adherence, and lubrication), the membrane mucins are also signaling
`molecules via their cytoplasmic tails and epidermal growth factor (EGF) domains [6].
`While providing lubrication and protection of the ocular surface, this layer reduces
`the efficacy of pharmaceutical treatment by limiting tissue penetration. However, as
`we will see below, this negatively charged mucosa may also be diverted from its
`original function and may be a critical player in the improvement of ocular drugs
`formulated in cationic emulsions.
`The presence of a constant tear flow of approximately 1.2 μL/min (0.5–2.2
`μL/min) is also an important protection system. This constant flow results in a tear
`turnover rate of 16 % per minute during waking hours, and the reflex lachrymation
`may increase this rate up to 100-fold, to 300 μL/min. The high turnover rate of
`this precorneal tear film contributes to the protection of the eye, but also to the low
`availability of topically administered drugs [7].
`
`9
`
`

`

`24 Development of a Cationic Nanoemulsion Platform (Novasorb®) for Ocular Delivery
`
`519
`
`The inner part of the eye is protected by the cornea, which has a complex structure
`of three different layers with varying physiological properties alternating between
`lipophilicity and hydrophilicity [8]. This layered and alternated construction makes
`corneal crossing of most drugs very difficult. Optimal permeant molecules should
`have a log D of 2–3 [9]. Examining in greater detail the corneal structure, first the
`outermost layer of the cornea is distinguished, i.e., lipophilic epithelium, which
`is formed by epithelial cells linked by tight junctions providing a strong barrier
`to the molecules present in the tear fluid. The corneal epithelium is consequently
`almost impermeable to any substance larger than 500 Da [10]. The next layer is
`made up of the stroma, a hydrophilic layer composed of fibrous tissue made of large
`collagen fibers and proteoglycans that form the major part of the cornea. Finally,
`the endothelium is a monolayer of hexagonal cell interfaces, which is also quite
`lipophilic [11].
`In addition, it should be noted that the cornea is innervated by sensory nerve ter-
`minals of the trigeminal ganglion. Physical and chemical agents acting on the ocular
`surface (extreme environmental temperatures, wind, foreign bodies, and chemicals)
`induce conscious sensations and reflex motor and autonomic responses (blinking,
`lacrimation, conjunctival vasodilation) aimed at protecting the eye from further injury
`and drug penetration [12].
`These successive physical and biological protections result in less than10 % of
`an instilled drug being absorbed by ocular tissues [13], leading to poor efficacy and
`the need for repeated instillations for the vast majority of eye drops. As a result of
`extending and maintaining the efficacy of eye drop solutions, the drug concentration
`needs to be increased but with potential exacerbation of local and/or systemic side
`effects with potent drugs such as timolol (a beta-blocker). Consequently, there is a
`need for new formulations that will improve efficacy while also limiting the risk of
`local side effects.
`
`24.1.2 Options to Overcome Physiological Barriers
`
`Ocular drug absorption from the lacrimal fluid to the anterior ocular tissues via trans-
`corneal absorption is determined by two major factors: drug permeability through
`the cornea and contact time of the product with ocular tissues. Based on these
`two principles, scientists have created several valuable approaches to overcome the
`barriers.
`
`24.1.3 Enhancing Penetration
`
`Firstly, to promote drug permeability, penetration enhancers have been added to
`aqueous eye drops with some success [14]. These excipients, based on their surface-
`active property, are able to open corneal epithelium tight junctions and desmosomes
`leading to penetration toward the anterior chamber. The literature is very rich in
`examples of studies testing penetration enhancers, their effect toward hydrophilic
`
`10
`
`

`

`520
`
`F. Lallemand et al.
`
`and lipophilic drugs, the size and charge of molecules, etc. [15]. However, all authors
`agree on one fact: Although penetration enhancers can be good adjuvants for ocular
`penetration, as regards their surfactant nature they are intrinsically deleterious for
`the ocular surface [16]. Long-term use of penetration enhancers might result in poor
`patient compliance due to chronic discomfort, thus limiting the use of penetration
`enhancers in ophthalmology.
`Another way to increase corneal penetration is to increase the specific uptake
`of the drug through the use of vectors, such as nanoparticles or liposomes, which
`have been described as being specifically taken up by corneal cells, as shown and
`discussed by Calvo et al. [17] and Diebold et al. [18]. However, the exact mechanism
`is not yet fully elucidated, therefore, limiting the use of this promising approach.
`Corneal penetration can also be influenced by the specific contact surface created
`by the colloidal system with the cornea. Emulsions are a typical example of such
`systems. An oil-in-water emulsion is a dispersion of oil into a water phase. This
`dosage form has been used in pharmacy for decades but only recently in ophthal-
`mology as eye drops. Ophthalmic emulsions have shown promise in topical ocular
`delivery as they are nontoxic systems, easy and inexpensive to manufacture, and able
`to deliver a lipophilic active agent with enhanced corneal penetration [19]. In addi-
`tion, ophthalmic emulsions are able to protect unstable active agents from chemical
`degradation (such as latanoprost) and to mask the irritation potential of some drugs.
`The first ophthalmic emulsion to be approved in the USA was Restasis® in 2002.
`This dosage form is now routinely used to treat dry eye conditions. A few years
`later, Durezol® was marketed to treat ocular inflammation. The exact mechanistic
`processes regarding enhanced corneal penetration has still not been fully elucidated
`[20] but they most likely involve several physicochemical and biological mechanisms
`whose increased specific surface of exchange with the cornea has major importance.
`
`24.1.4 Increasing Retention Time
`
`Increased retention time by enhanced viscosity or bioadhesion has been widely used
`and described during the past 20 years [8], leading to a number of ophthalmic prod-
`ucts. Mucoadhesion is based on noncovalent bonds (hydrogen or electrostatic bonds)
`between polymers and mucus or physical entanglement. In spite of their current wide
`use to decrease the frequency of administration and/or concentration in the solution,
`hydrogels present only a limited value because such aqueous formulations are elim-
`inated by the usual routes in the ocular domain and cause blurred vision and patient
`discomfort.
`
`24.1.5 Enhancing Penetration and Increasing Retention Time
`
`Improving the eye surface’s drug retention alone is inadequate in bringing a signif-
`icant improvement of drug bioavailability. It should be combined with penetration
`
`11
`
`

`

`24 Development of a Cationic Nanoemulsion Platform (Novasorb®) for Ocular Delivery
`
`521
`
`Drop of emulsion
`
`Ca(cid:415)onic nanodroplet of oil
`loaded with ac(cid:415)ve ingredient
`
`+
`
`+
`
`+
`
`+
`
`+
`+
`
`+
`
`+
`
`+
`
`+
`
`+
`
`+
`+
`+
`
`+
`+
`
`+
`
`Nega(cid:415)vely charged
`mucins layer
`
`Cornea
`
`Corneal epithelium
`
`•
`
`•
`
`•
`
`•
`
`Stroma
`
`+ + + + + + + + +
`• • • • •
`
`• • • •
`
`•
`
`•
`
`•
`
`•
`
`•
`
`•
`
`•
`
`• •
`
`•
`
`•
`
`•
`
`Fig. 24.1 Novasorb® enhanced the spread of a drop emulsion on the corneal surface and increased
`the contact surface of oily drops on the ocular surface
`
`enhancement. Several examples of combined systems (penetration and retention)
`have been described in the literature, such as liposomes combined with hydrogels
`[21], liposomes combined with a collagen shield [22], and even more solid lipid
`nanoparticles associated with a bioadhesive hydrogel such as chitosan [23]. Although
`promising, these examples all remain today at the prototype stage. Nevertheless, a
`new and innovative system has come to the market combining the effect of a very
`large specific surface of colloidal systems with improved bioadhesion properties. This
`drug-delivery system is the cationic nanoemulsion registered under the trademark
`Novasorb® technology (Fig. 24.1).
`
`24.2 Cationic Emulsions
`
`24.2.1 Cationic Agent
`
`While ophthalmic emulsions are becoming an essential tool for topical delivery,
`cationic emulsions combining intrinsic advantages of emulsions with a bioadhesive
`mechanism have provided enhanced efficacy of emulsions [24, 25]. This innovative
`approach uses the physiological barrier of mucus as a tool to increase retention
`
`12
`
`

`

`522
`
`Fig. 24.2 Benzalkonium
`chloride is a mixture of
`various aliphatic chain
`lengths starting from C8 to
`C18 with C12, C14, and C16
`representing the major
`entities of the mixture
`
`F. Lallemand et al.
`
`time on the ocular surface. As discussed above, the last protection layer of the tears
`is composed of mucins that have a negative charge. Cationic nanoemulsions use
`this negative charge to interact with the ocular surface via a strong electrostatic
`interaction leading to a prolonged residence time of the cationic nanodroplets on the
`ocular surface [26].
`Basically, cationic emulsions are composed of oil that is dispersed in ultrafine
`droplets into a physiologically acceptable aqueous external phase (pH and osmot-
`ically adjusted). These droplets are stabilized by an interfacial film of surfactants
`such as cremophors, polysorbates, poloxamers, and tyloxapol in which a cationic
`charge is included. The cationic agent can be chosen over those commonly used
`and described in the literature. One can cite the primary amines stearylamine and
`oleylamine, the cationic phospholipid DOTAP and the polymers poly-L-lysine and
`polyethylenimine. Yet, these cationic agents are not suitable for use in pharmaceu-
`tical products in terms of regulatory requirements, stability, or toxicity issues. The
`alternative is to use registered excipients such as quaternary amines usually used
`as preservative agents in ophthalmic aqueous solutions. The most widely used are
`cetylpyridinium chloride, benzalkonium chloride (BAK), and benzethonium chlo-
`ride. However, the current tendency is to withdraw these preservatives from eye drops,
`due to long-term intolerance to them [27, 28], and to replace them either with soft
`preservatives (sodium perborate, SofziaTM) or single-use containers or preservative-
`free multidose containers. Nonetheless, BAK has several significant advantages over
`the other cationic agents: it is listed in all pharmacopeias, used in more than 80 %
`of eye drops at a concentration of 0.02 %, and has excellent interfacial properties,
`making this excipient a potential cationic agent candidate.
`Ten years ago, Sznitowskaat et al. [29] noted that when used in combination with
`oil-in-water emulsions, the preservative efficacy of BAK was drastically decreased.
`This observation was explained by the inclusion of part of BAK in the oily phase of
`the emulsion, leading to a lower molecular concentration available in the aqueous
`phase. Only the freely soluble molecules of BAK present in the aqueous phase can
`exert their antimicrobial effects on bacteria. Concretely, BAK is a mixture of sev-
`eral quaternary ammoniums (Fig. 24.2) with varying lipophilicity. According to US
`and European pharmacopeias, three main entities should be present in the mixture:
`benzododecinium chloride (C12-substituted alkyl chain), myristalkonium chloride
`(C14), and cetalkonium chloride (C16), as presented in Fig. 24.3. In presence of oil
`droplets, the most lipophilic entity of BAK, cetalkonium chloride, is rearranged at the
`oil or water interface, providing a cationic charge on the droplets while hydrophilic
`
`13
`
`

`

`24 Development of a Cationic Nanoemulsion Platform (Novasorb®) for Ocular Delivery
`
`523
`
`Fig. 24.3 Benzalkonium
`chloride made of three
`different entities of increasing
`lipophilicity (cetalkonium
`chloride, myristalkonium
`chloride, and benzododecin-
`ium chloride) and cetalkoni-
`um chloride schematic
`developed formulae
`
`entities remain in the aqueous phase. Based on this observation, it was decided to use
`only cetalkonium chloride at a very low concentration (0.002–0.005 %) as a cationic
`agent to make cationic nanoemulsions [30].
`
`24.2.2 Physicochemical Considerations Regarding Novasorb®
`
`Cationic nanoemulsions are characterized by two main physicochemical properties.
`First, the zeta potential, which is defined as the electrical potential difference between
`the dispersion medium (i.e., water) and the stationary layer of fluid attached to the
`dispersed oil nanodroplets [31]. This surface charge will provide the system with its
`bioadhesion property on the ocular surface as well as the stability of the emulsion by
`providing electric repulsion between droplets. For optimal electrostatic interaction
`between cornea and product, a zeta potential of about + 20 mV is sufficient [30].
`The second main property is the oil droplet size. As described by the Stokes
`law (Fig. 24.4), the smaller the droplet size, the slower the dispersed system will
`separate, thus providing greater stability to the system. Even more importantly, the
`size of the droplets significantly participates in the penetration rate of the drug in the
`ocular tissue. With the active ingredient being solubilized in the lipid nanodroplets,
`a smaller particle size should provide a greater contact surface, hence a higher tissue
`concentration. In the case of the Novasorb® technology, a size between 100 and
`200 nm was demonstrated to be small enough to provide a stable emulsion.
`Surface bioadhesion was demonstrated by measurement of the spreading prop-
`erties of the emulsion in contact angle studies. On excised rabbit eyes, a drop of
`cationic emulsion is applied and compared to anionic emulsion and hyaluronic gel
`in terms of contact angle (Fig. 24.5). Immediately after drop deposit, the cationic
`
`Fig. 24.4 Stokes law equation where vs is the particle creaming velocity, g is the gravitational
`acceleration, ρp is the mass density of the particles, ρf is the mass density of the fluid and μ is the
`viscosity of the continuous phase
`
`14
`
`

`

`524
`
`F. Lallemand et al.
`
`Fig. 24.5 Dynamic contact angle measurements on rabbit eyes confirm optimal and immediate
`spreading of cationic emulsions compared to anionic emulsions and hyaluronic acid-based product
`(adapted from Lallemand et al. [30])
`
`emulsion based on Novasorb® spread all over the cornea while other tested products
`remained with high contact angles.
`Other physicochemical parameters are important for the development and use of
`Novasorb®. It should be noted that to avoid reflex tearing and blinking the emulsions
`should be compatible with biological parameters (pH and osmolality). pH is adjusted
`to physiological pH (about 7–7.2). Osmolality should be adjusted to avoid osmotic
`stress to the epithelial cells. Neutral molecules should be used to avoid charge mask-
`ing and emulsion destabilization such as mannitol, sorbitol or, glycerol. Glycerol is
`favored because this compound possesses an intrinsic beneficial demulcent property
`on the eye surface that is particularly useful in dry eye disease (DED) [32].
`Finally, several lipophilic active ingredients were added to the cationic emulsion
`either to address unmet medical needs or to improve existing products: cyclosporin
`A (CsA), latanoprost, antihistaminics, anti-inflammatories, antibiotics, and antifun-
`gals. For example, latanoprost, the active ingredient of the antiglaucoma blockbuster
`product Xalatan®, was included in the emulsion providing new properties. La-
`tanoprost is an unstable molecule in presence of water due to ester hydrolysis. When
`encapsulated in oil, the concerned ester function is masked in the oily phase, thus
`avoiding hydrolysis and providing an advantage over Xalatan®. In addition, as dis-
`cussed below, the beneficial effect of Novasorb® on the ocular surface should be
`beneficial to patients with combined glaucoma and ocular surface disease.
`Another example is CsA. This molecule has always been a huge challenge to
`formulate in an adequate and efficient product [33] due to its high lipophilicity.
`Novasorb® seems the most appropriate technology to administer this molecule to
`patients.
`
`24.3 Nonclinical Evaluation
`
`24.3.1 Pharmacokinetics and Efficacy of Cationic Emulsions
`
`The ocular delivery of Novasorb® cationic emulsions loaded CsA and latanoprost
`was evaluated in rabbits. The first indications that cationic emulsions were effective
`ocular drug-delivery vehicles with prolonged precorneal residence time come from
`
`15
`
`

`

`24 Development of a Cationic Nanoemulsion Platform (Novasorb®) for Ocular Delivery
`
`525
`
`the work of Benita et al., who used delta 8-tetrahydrocannabinol and pilocarpine as
`model drugs [34, 35]. These results were confirmed later with CsA cationic emul-
`sions, which have an increased ocular absorption, especially in the conjunctiva and
`cornea, when compared to anionic emulsions [20, 36]. However, these first cationic
`emulsions all used noncompendial excipients, or primary amines, such as steary-
`lamine and oleylamine as the cationic agent, which are not devoid of toxicity. Based
`upon these results, the Novasorb® technology was developed to create cationic emul-
`sions of CsA and latanoprost with improved ocular tolerance safety profiles. CsA
`cationic emulsions were compared to Restasis®, a commercially available unpre-
`served BAK-free anionic emulsion of 0.05 % CsA, in single- and multiple-dose
`PK studies [37]. The corneal absorption of CsA following a single instillation of a
`0.05 % CsA cationic emulsion was approximately twice that observed with Restasis®
`(Fig. 24.6a), and a 0.025 % CsA cationic emulsion was as effective as Restasis® at
`delivering CsA to the cornea. The area under the curve (AUC) in the cornea following
`a single instillation was 14,210, 14,476, and 26,476 ng h/g for Restasis®, 0.025 %
`CsA cationic emulsion, and 0.05 % CsA cationic emulsion, respectively (Fig. 24.6b).
`Interestingly, a second peak is observed 4 h post instillation with the 0.05 % CsA
`cationic emulsion, which is not present with the Restasis® anionic emulsion. This
`suggests that the cationic emulsion does indeed possess a prolonged precorneal res-
`idence time. With the 0.1 % CsA cationic emulsion, this second peak was observed
`12 h post instillation [37], confirming that for such a peak to be present, a prolonged
`residence time in the precorneal space was necessary. This prolonged residence time
`can be explained by the presence of the positive charge on the oil droplets of the
`emulsion, which can interact with the negatively charged corneal epithelium. In the
`multiple-dose PK studies, bis in die (BID) (twice daily) instillations for 7 days of
`the 0.05 % CsA cationic emulsion or once daily instillation for 7 days of the 0.1 %
`CsA cationic emulsion were not accompanied by an increased systemic absorption
`of CsA.
`Confirmation of the good absorption following instillations of the CsA cationic
`emulsions was obtained with a 0.005 % latanoprost cationic emulsion. In a monkey
`model of laser-induced elevated intraocular pressure (IOP), the 0.005 % latanoprost
`cationic emulsion was as effective as Xalatan® (0.005 % latanoprost) at reducing
`elevated IOP [38], suggesting an equivalence in the latanoprost-delivered dose be-
`tween the two 0.005 % latanoprost formulations. Xalatan® contains 0.02 % BAK, a
`quaternary ammonium that at this high concentration acts both as a preservative for
`the eye drop solution and a permeation enhancer for latanoprost. For example, Al-
`lergan increased the BAK concentration, from 0.005 to 0.02 %, while decreasing the
`concentration of bimatoprost (a prostaglandin analog, PGA) from 0.03 to 0.01 % in
`order for the new Lumigan® 0.01 % bimatoprost eye drop solution to have the same
`efficacy as the original 0.03 % bimatoprost formulation. In this regard, increasing the
`BAK concentration potentiated the absorption of bimatoprost and helped in main-
`taining its efficacy. This formulation change was motivated by the side effect of the
`PGA class: hyperemia, i.e., conjunctival redness, which results from PGA-induced
`conjunctival vein vasodilation. Hyperemia is the major cause of antiglaucoma PGA
`treatment cessation and poor compliance.
`
`16
`
`

`

`526
`
`F. Lallemand et al.
`
`CsA concentra(cid:415)on in the cornea
`
`Restasis®
`(0.05% CsA)
`0.025% CsA
`Ca(cid:415)onic EM
`0.05% CsA
`Ca(cid:415)onic EM
`
`1600
`
`1400
`
`1200
`
`1000
`
`800
`
`600
`
`400
`
`200
`
`0
`
`0
`
`4
`
`8 12 16 20 24 28 32 36 40 44 48 52 56 60 64 68 72
`Time (hour)
`AUC
`
`30000
`25000
`2000

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