throbber

`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`Jeremy C. Wright - Diane J. Burgess
`Editors
`
`Long Acting Injections
`and Implants
`
`@ Springer
`
`AstraZeneca Exhibit 2157 p. 1
`InnoPharma Licensing LLC V. AstraZeneca AB IPR2017-00905
`
`

`

`Editors
`
`Jeremy C. Wright
`DURECT Corporation
`Cupertino, CA, USA
`jeremy.wright@durect.com
`
`Diane J. Burgess
`Department of Pharmaceutical Sciences
`School of Pharmacy
`University of Connecticut
`Storrs, CT, USA
`d .burgess @ uconn.edu
`
`ISSN 2192-6204
`ISBN 978-1—4614—0553-5
`DOI 10.1007/978—1-4614—0554-2
`
`e-ISBN 978-1-4614-0554-2
`
`Springer New York Dordrecht Heidelberg London
`
`Library of Congress Control Number: 2011937763
`
`© Controlled Release Society, 2012
`All rights reserved. This work may not be translated or copied in whole or in part without the written
`permission of the publisher (Springer Science+Business Media, LLC, 233 Spring Street, New York,
`NY 10013, USA), except for brief excerpts in connection with reviews or scholarly analysis. Use in
`connection with any form of information storage and retrieval, electronic adaptation, computer software,
`or by similar or dissimilar methodology now known or hereafter developed is forbidden.
`The use in this publication of trade names, trademarks, service marks, and similar terms, even if they
`are not identified as such, is not to be taken as an expression of opinion as to whether or not they are
`subject to proprietary rights.
`
`Printed on acid—free paper
`
`Springer is part of Springer Science+Business Media (www.springer.com)
`
`AstraZeneca Exhibit 2157 p. 2
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`Contents
`
`1
`
`2
`
`3
`
`4
`
`5
`
`6
`
`7
`
`8
`
`9
`
`11
`
`An Introduction to Long Acting Injections and Implants ............... ..
`Diane J. Burgess and Jeremy C. Wright
`
`Historical Overview of Long Acting Injections and Implants ......... ..
`Jeremy C. Wright and Allan S. Hoffman
`
`Host Response to Long Acting Injections and Implants .................. ..
`James M. Anderson
`
`Anatomy and Physiology of the Injection Site:
`Implications for Extended Release Parenteral Systems ................... ..
`Arlene McDowell and Natalie J. Medlicott
`
`Drugs for Long Acting Injections and Implants ............................... ..
`Jie Shen and Diane J. Burgess
`
`Diseases and Clinical Applications that Can Benefit
`from Long Lasting Implants and Injections ...................................... ..
`Roshan James, Udaya S. Toti, Sangarnesh G. Kumbar,
`and Cato T. Laurencin
`
`Oily (Lipophilic) Solutions and Suspensions ..................................... ..
`Susan W. Larsen, Mette A. Thing, and Claus Larsen
`
`Aqueous Suspensions ........................................................................... ..
`Susan M. Machkovech and Todd P. Foster
`
`In Situ Forming Systems (Depots) ...................................................... ..
`Jeremy C. Wright, Michael Sekar, William van Osdol,
`Huey Ching Su, and Andrew R. Miksztal
`
`Microsphere Technologies ................................................................... ..
`Yan Wang and Diane J. Burgess
`
`Liposomes as Carriers for Controlled Drug Delivery ...................... ..
`Xiaoming Xu and Diane J. Burgess
`
`1
`
`11
`
`25
`
`57
`
`73
`
`93
`
`113
`
`137
`
`153
`
`167
`
`195
`
`xi
`
`AstraZeneca Exhibit 2157 p. 3
`
`U}..
`
`E
`Ba
`E'U
`V}
`5
`E
`.2..
`
`8
`EDD
`.5...
`
`O<D
`
`D
`‘5
`.4
`
`S"
`
`8a
`
`;
`NNI:
`.23;
`f3._.
`g:
`.23Q“
`mm
`a:
`MBan
`“9.9
`0%.>,0
`uE’>-‘
`§%
`2‘3-
`can)
`:2,
`3':D.
`
`

`

`xii
`
`Contents
`
`12 Micro- and Nanoemulsions (Controlled Release Parenteral
`Drug Delivery Systems) ....................................................................... ..
`Jacqueline M. Morais and Diane J. Burgess
`
`13 Nanosuspensions .................................................................................. ..
`Sumit Kumar and Diane J. Burgess
`
`14 PEGylated Pharmaceutical Nanocarriers ......................................... ..
`Vladimir Torchilin
`
`15 Protein PEGylation .............................................................................. ..
`Francesco M. Veronese and Gianfranco Pasut
`
`16 Self-Assembling Lipid Formulations .................................................. ..
`Fredrik Tiberg, Markus Johnsson, Catalin Nistor,
`and Fredrik Joabsson
`
`17
`
`Implantable Drug Delivery Systems Based
`on the Principles of Osmosis ............................................................... ..
`John A. Culwell, Jose R. Gadea, Clarisa E. Peer,
`
`and Jeremy C. Wright
`
`18 Microtechnologies for Drug Delivery ................................................. ..
`Kristy M. Ainslie and Tejal A. Desai
`
`19 Drug-Eluting Stents ............................................................................. ..
`Jonathon Zhao and Lori Alquier
`
`20 Delivery of Peptides and Proteins via Long Acting
`Injections and Implants ....................................................................... ..
`Cynthia L. Stevenson, Christopher A. Rhodes,
`and Steven J. Prestrelski
`
`21
`
`Injectable PLGA Systems for Delivery of Vaccine Antigens ............ ..
`Vesna Milacic, Brittany Agius Bailey, Derek O’Hagan,
`and Steven P. Schwendeman
`
`22 Methods of Sterilization for Controlled Release Injectable
`and Implantable Preparations ............................................................ ..
`Alpaslan Yaman
`
`23
`
`In Vitro Drug Release Testing and In Vivo/In Vitro
`Correlation for Long Acting Implants and Injections ...................... ..
`Michail Kastellorizios and Diane J. Burgess
`
`24 Regulatory Issues and Challenges Associated
`with the Development of Performance Specifications
`for Modified Release Parenteral Products ......................................... ..
`
`Marilyn N. Martinez and Mansoor A. Khan
`
`221
`
`239
`
`263
`
`295
`
`315
`
`335
`
`359
`
`383
`
`409
`
`429
`
`459
`
`475
`
`505
`
`Index .............................................................................................................. ..
`
`537
`
`AstraZeneca Exhibit 2157 p. 4
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`Chapter 4
`Anatomy and Physiology of the Injection Site:
`Implications for Extended Release Parenteral
`Systems
`
`Arlene McDowell and Natalie J. Medlicott
`
`Abstract Understanding how the biological environment contributes to drug
`release following administration is increasingly becoming a focus for drug delivery
`research. Achieving therapeutic levels of a bioactive relies on appropriate drug
`release following parenteral administration that must be complimentary to subse—
`quent drug absorption, distribution, metabolism and elimination. The biological
`characteristics of the injection site can have an influence on the drug absorption
`process. In this chapter the intravenous, intramuscular and subcutaneous routes for
`parenteral administration of extended release products will be discussed.
`
`4.1
`
`Introduction
`
`There are many extended and controlled release injectable systems used to deliver
`drugs in human and veterinary medicine [1—5]. These systems are prepared from a
`variety of biocompatible materials and aim to release drug for an extended period
`following injection or implantation. Drug release is governed by the design of the
`dosage form, although the biological environment often influences drug release
`[6—9]. Understanding how the biological environment contributes to drug release
`following administration is increasingly becoming a focus for drug delivery research.
`Extended release parenteral delivery systems range from relatively simple aque-
`ous suspensions that prolong drug release due to slow dissolution at the injection
`site to more sophisticated in situ gelling implants and polymeric biodegradable
`microparticulate systems [4, 10, 11]. For example, long acting intramuscular aque—
`ous suspensions of penicillin have been available since the 1950s [12] and oily
`
`A. McDowell - N.J. Medlicott (8)
`School of Pharmacy, University of Otago, PO. Box 56, Dunedin 9054, New Zealand
`e-mail: natalie.medlicott@otago.ac.nz
`
`J.C. Wright and DJ. Burgess (edsi), Long Acting Injections and Implants,
`Advances in Delivery Science and Technology, DOI 10.1007/978-14614-0554-2_4,
`© Controlled Release Society, 2012
`
`57
`
`AstraZeneca Exhibit 2157 p. 5
`
`

`

`58
`
`A. McDowell and NJ. Medlicott
`
`.
`inJeCtlonl.
`Implantation site
`depm
`
`
`
`Implant
`
`or
`depot
`
`
`release
`
`free
`
`drug in
`
`tissue
`
`
`
`
`
`:
`:
`E
`
`E
`
`E
`E
`:
`:
`l
`
`metabolites
`
`degraded drug
`
`I
`:
`l
`
`l
`
`E
`:
`E
`;
`l
`
`bound drug
`(tissue or
`protein)
`
`‘
`
`to systemic
`circulation
`
`
`
`absorption
`
`lg
`
`
`.
`Blood or lymphatic
`
`
`l\dug in
` protein
`
`distribution
`
`plasma /
`
`bound drug
`
`metabolism
`and
`elimination
`
`Fig. 4.1 Schematic showing pharmacokinetics for a drug administered as an extended release
`intramuscular or subcutaneous system
`
`formulations of neuroleptic drugs since the 1970s [13]. More recently, sustained
`release microparticulate polymeric formulations of leuprolide have been developed
`for prostate cancer [1, 14, 15]. For all implanted extended release delivery systems,
`balancing the in vivo drug release from the delivery system with drug absorption,
`distribution, metabolism and elimination processes is key to achieving target drug
`levels in the body (Fig. 4.1). Hence, careful consideration of drug pharmacokinetics
`and knowledge of target plasma and tissue drug concentrations can guide the devel-
`opment of extended and controlled release parenteral drug delivery systems. One
`useful way to categorize parenteral delivery systems has been suggested by
`Washington et al. for intramuscular injections [16]. According to this, injectable
`delivery systems can be divided into those in which the pharrnacokinetics are
`predominately controlled by the implant (or device) and those controlled by the
`process of absorption into blood or lymphatic capillaries at the implant site (i.e.
`perfusion limited pharmacokinetics), as shown in Fig. 4.2. If drug release from the
`device is slow and drug absorption from the tissue is fast, then a situation results
`where the appearance of drug in the blood is closely controlled by the release char-
`acteristics of the extended release device (Type I). At the other extreme, an aqueous
`
`AstraZeneca Exhibit 2157 p. 6
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`4 Anatomy and Physiology of the Injection Site...
`
`59
`
`ka
`drug in
`1"
`Device
`or depot —') tissue _’ Type 1: kr < ka : device-limited PK
`
`kl”
`
`drug in
`
`ka
`
`—> tissue —) Type 2: kr > ka:perfUS10n-llmlted PK
`
`'
`
`_
`
`_
`
`Fig. 4.2 Pharmacokinetic (PK) classification of intramuscular implantable delivery systems.
`Modified from Washington et al. [16]
`
`Imflar
`
`Epidemis
`Dennis
`
`Muscle
`
`Subcutaneous tissue
`
`Fig. 4.3 Potential sites for injection of extended release parenteral dosage forms
`
`solution of a water—soluble drug provides its dose immediately following injection
`and in this case, the absorption processes (k3) may limit the appearance of drug in
`the blood (Type H). Many extended release parenteral delivery systems fall between
`these two extremes so it is important to consider the biological processes at injec—
`tion sites that contribute to drug absorption. A further example of extended release
`systems are those designed to deliver drugs locally at the implantation site. For
`these local delivery systems, a balance between drug release and drug absorption
`that maintains a constant local drug concentration may be the goal. The previous
`chapter has shown that while implantable extended release systems must be bio—
`compatible, they cannot be considered biologically inert because the patient will
`react to the presence of the implant.
`The aims of this Chapter are to review the anatomy and physiology of injection
`sites (intravenous, intramuscular and subcutaneous — Fig. 4.3) and to summarize the
`biological variables that affect drug release and absorption at these sites.
`
`AstraZeneca Exhibit 2157 p. 7
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`60
`
`A. McDowell and NJ. Medlicott
`
`4.2
`
`Intravenous Route
`
`The intravenous route of administration for injectable products provides direct
`access into the blood stream and so therapeutic agents delivered by this route are
`available immediately in the systemic circulation. The plasma concentration is
`determined by the initial dose injected, the drug distribution and rates of metabo-
`lism and elimination according to Eqs. (4.1) and (4.2) for first-order elimination
`kinetics.
`
`
`Dose
`C0 = V ,
`
`C, = C0 x e‘“,
`
`(4.1)
`
`(4.2)
`
`where: C0=initial plasma concentration, V= volume of distribution, Ctzplasma
`concentration at time t and k: first—order elimination rate constant.
`
`Controlled drug release can be achieved using the intravenous route when par-
`ticulate systems such as liposomes or polymeric nanoparticles are used [2, 17, 18].
`The prolonged effect of these dosage forms is primarily attributed to the time taken
`for the bioactive to be released from the circulating particles. Properties of nanopar-
`ticles such as: method of preparation, method of drug association (e.g., encapsulated
`or surface-adsorbed) and the type of polymer used can be manipulated to alter the
`drug release profile and the in vivo fate of the drug. For example, nanoparticles that
`have the drug dispersed uniformly throughout a matrix generally release drug in a
`first-order process controlled by diffusion and polymer degradation [19].
`Nanoparticulate systems administered by the intravenous route can also be used to
`facilitate drug targeting to specific tissues and cell types. Active targeting can be
`achieved through surface modifications, such as conjugation of ligands, e.g., biotin
`to poly(D,L—1actide-co-glycolide) nanoparticles to deliver paclitaxel to tumors [20].
`Restrictions are placed on the size of particulates that can be injected into the
`circulatory system so that controlled release systems designed for intravenous use
`are typically within colloidal range with diameters less than 500 nm [21]. With
`increasing research into particulate delivery systems, understanding of the tissue
`distribution of colloidal particles following administration into the blood has
`advanced. Moghimi et al. [22], in a review of long-circulating and target-specific
`nanoparticles, reported that passive tissue distribution patterns of intravenously
`administered particles depended on size and defonnability of the particles as well as
`their surface chemistry. Slack et a1.
`[23] showed polystyrene-divinylbenzene
`microparticles with diameters 7.4 and 11.6 um were deposited mainly in the lungs,
`while particles with diameter 3.4 pm deposited in the liver and spleen following
`intravenous administration. Smaller nanoparticles (less than 150 nm) appear to be
`distributed more widely than larger colloids with particles seen in the liver, spleen,
`bone marrow, bone, heart, kidney and stomach [21, 24, 25]. At tumor sites and sites
`of tissue inflammation, the spaces between endothelial cells lining blood capillaries
`are enlarged so that correctly sized nanoparticles can escape the vasculature into
`
`AstraZeneca Exhibit 2157 p. 8
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`4 Anatomy and Physiology of the Injection Site...
`
`61
`
`underlying tissue in these sites [25—27]. This mechanism is exploited in the target-
`ing of cytotoxic drugs to tumors [28, 29] or anti-inflammatory drugs to sites of
`tissue inflammation [27]. Tumors at different sites of the body, however, appear to
`have different levels of microvasculature porosity, which may affect the accumula-
`tion of particulate delivery systems [25]. Pore sizes ranging from 200 nm to 1.2 pm
`were reported with experimental subcutaneous tumors, but lower pore size (less
`than 500 nm) was reported in brain tumors. The microvascular pore size also varied
`depending on the tumor cell line [25]. At even smaller sizes, particles may be elimi-
`nated from the body by filtration through the kidney. Choi et al. suggested that
`particles less than 5 nm diameter were freely filtered in the kidneys, while those
`with diameter greater than 5 nm were retained in the body [30]. This suggests that
`there is also a lower particle size limit (5 nm) as well as the higher one (500 nm) for
`particulate systems administered via the intravenous route.
`
`4.3
`
`Intramuscular Route
`
`The intramuscular site is reached by injection through the hypodemris into the
`underlying skeletal muscle. The structure of musculature is such that vasculature
`extends into the muscle and each muscle fiber is surrounded by a number of capil-
`laries lying parallel to each fiber with transverse vessels between muscle fibers [31].
`Thus, muscle tissue is typically highly perfused with blood for the delivery of
`oxygen and nutrients to muscle cells and for the removal of waste material and so
`can be utilized for the systemic delivery of therapeutics. Extracellular fluid in skel-
`etal muscle is reported to have a pH of 7.1 at rest but decreases with exercise to 6.8
`due to lactate accumulation [32].1 This slightly acidic pH may influence release and
`absorption properties of weakly acidic or basic drugs. Lymphatic vessels are also
`present within the connective tissue that surrounds the muscle fibers and bundles;
`however, the lymph system is more extensive in the subcutaneous site compared to
`the intramuscular site [33]. Drug characteristics that promote absorption into the
`lymphatic system are discussed later.
`The most common muscles into which injections are made in humans are the
`gluteus maximus, vastus lateralis and deltoid. Blood flow in these muscles is reported
`to be fastest in the deltoid, and slowest in the gluteus maxirnus giving rise to potential
`differences in drug absorption rates at different sites of administration [34].
`Differences in muscle perfusion can be expected to have the greatest effect on drug
`absorption when uptake into injection site capillaries is the rate-limiting step in drug
`absorption. For example, more rapid absorption of diazepam from injections into the
`deltoid muscle compared with the vastus lateralis has been reported [35]. Additionally,
`when muscle perfusion is the rate—limiting step for drug absorption, activities that
`increase local blood flow such as exercise, and local muscle massage may be expected
`to increase the rate of drug absorption following intramuscular injection [36].
`Increased absorption has been reported for both intramuscular administration of
`
`1pH can also be lowered by the foreign body reaction to implants (see Chap. 3).
`
`AstraZeneca Exhibit 2157 p. 9
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`62
`
`A. McDowell and NJ. Medlicott
`
`b01o01
`
`.0 01
`
`
`
`Relativespreadingofradioactivity
`
`0
`
`4
`
`8
`
`12
`
`16
`
`20
`
`24
`
`28
`
`Time (h)
`
`Fig. 4.4 Graph of the spread following intramuscular injection of hydrophilic radioactive markers
`into thigh muscle of rabbit. Spreading is represented as relative to aprotinin encapsulated in an
`emulsion with 60% w/w aqueous phase. (Open circle) 30% w/w aqueous phase emulsion contain-
`ing aprotinin; open inverted triangle 60% w/w aqueous phase emulsion containing aprotinin;
`(Filled cirrle)30% w/w aqueous phase emulsion containing radioactive pertechnetate; (filled
`inverted triangle) 60% w/w aqueous phase emulsion containing radioactive pertechnetate; (filled
`diamond) solution of aprotinin in PBS administered intramuscularly. Figure with permission from
`Bjerregaard et a1. [39]
`
`penicillin and diazepam with exercise [37]. However, when drug release from the
`depot is slow, then the effects of increased muscle perfusion may not be great. Soni
`et al. showed no significant effects of injection site, massage or muscle activity on
`plasma levels from a depot injection of fluphenazine decanoate [38]. Muscle activity
`may also have an effect on the surface area available for drug release for low to inter-
`mediate viscosity systems as exercise could affect the spreadability of the extended
`release depot within the muscle tissue. For example, absorption of the protein apro—
`tinin [39] was found to be greater from formulations with a lower viscosity that
`spread more extensively within the muscle compared to the formulations with higher
`viscosity, 30% and 60% w/w emulsions, respectively (Fig. 4.4).
`The amount of fat associated with the muscle can also modulate absorption from
`intramuscular injections and has been given as an explanation for the slower rate of
`drug absorption following injection into the gluteus maximus in females compared
`to males [40]. Cockshott et a]. have shown that injections intended for intramuscular
`administration into the gluteal muscle may not reach the muscle and may indeed
`be made most of the time into the fat surrounding the muscle [41]. Their study of
`63 men and 60 women indicated that at any given weight, the skin to muscle dis-
`tance in the gluteal region was approximately 2.5 cm greater in women than in men.
`If a standard 3.5 cm length needle was used, then under 5% of women would receive
`
`AstraZeneca Exhibit 2157 p. 10
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,Diane1.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`4 Anatomy and Physiology of the Injection Site...
`
`63
`
`the intramuscular injection at the desired depth of at least 0.5 cm into the muscle
`[41]. Hence, the reported slower absorption following intramuscular injection into
`the gluteus maximus site may be due to injection into fat overlying the muscle rather
`than muscle itself.
`
`4.4 Subcutaneous Route
`
`The subcutaneous tissue or hypoderrnis is situated directly below the dermis layer
`of the skin and exterior to the muscle layer [16]. The characteristic feature of this
`site is the storage of dietary adipose within loose connective tissue in the interstitial
`space [16]. The interstitial space comprises a collagen network embedded in a gel
`of glycosaminoglycans, salts and proteins [42] and has a pH of 7.3 [43]. The acces-
`sibility of the subcutaneous site and relative ease of injection with short, fine needles
`contribute to its popularity for self—administration of medications such as insulin.
`Implants, particulates or in situ gelling systems can sit comfortably within the
`connective tissue of the subcutaneous site and release drug that is then absorbed
`into surrounding blood or lymph capillaries.
`Extended release injections containing contraceptive hormones have been in use
`since the late 1960s, initially with intramuscular Depo Provera®, a long acting
`aqueous suspension of medroxyprogesterone acetate administered once every three
`months [44] and more recently a lower dose subcutaneous form, depo—subQ Provera
`[45]. The duration of effective contraceptive action was further extended with the
`introduction of levonorgestrel containing polydimethylsiloxane implants. The first
`of these were the Norplant® implants of the 19805, which gave slow release over
`5 years following subcutaneous administration. Six rod shaped implants were
`needed for treatment with Norplant, and re-design of this product allowed the
`number of implants to be reduced to two [46]. More recently a single rod implant
`for subcutaneous administration has been developed incorporating etonogestrel in
`ethylene vinyl acetate (ImplanonTM) to provide contraceptive effects over a period
`of three years [47].
`Blood perfusion in the subcutaneous tissue is recognized to be lower than in the
`intramuscular site, which translates into comparatively slower absorption, lower
`maximum plasma concentrations and longer times to maximum plasma concentra—
`tion as illustrated for the antibiotic cefotaxime in sheep (Fig. 4.5). A further delay
`in the appearance of drug within the systemic circulation results if drug is absorbed
`into subcutaneous lymphatic capillaries because time is required for the drug to
`transverse the lymphatic system and enter the blood circulation. The lymphatic
`system has unidirectional flow and maintains interstitial pressure through the col-
`lection of fluid and proteins from the interstitial fluid through a series of draining
`lymph nodes and is eventually returned to systemic circulation [49]. Lymph capil-
`laries are more permeable than blood capillaries because they are lined with a single
`layer of endothelial cells that have an incomplete basal layer and lack coherent tight
`junctions between adjacent endothelial cells [42]. The composition of protein in the
`lymph is similar to that in blood plasma and lymph flow rate is 100—500 times
`
`AstraZeneca Exhibit 2157 p. 11
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`64
`
`A. McDowell and NJ. Medlicott
`
`(ug/mL)
`PlasmaConcentration
`
`
`0
`
`l
`
`2
`
`3
`
`4
`
`5
`
`Time (hours)
`
`Fig. 4.5 Comparative plasma concentrations following injection of an aqueous solution cefo—
`taxime (50 mg/kg) via the (filled circle) intramuscular and (open circle) subcutaneous routes in
`sheep. Data with permission from Guerrini et a1. [48]
`
`slower than the flow in blood vessels [49]. The importance of the lymphatic system
`for absorption of drugs is increasingly recognized for large molecular weight pro-
`tein drugs that do not partition well into blood capillaries [50]. Small drug mole-
`cules (<1 kDa) are predominantly absorbed into blood capillaries as their small size
`means they can partition relatively easily across the capillary endothelium and their
`diffusion through the interstitial fluid is not restricted [51, 52]. For larger peptide
`and protein compounds, absorption into blood vessels is limited primarily by their
`poor permeability across the capillary endothelial cell wall due to their large size.
`Consequently, these remain in the extracellular fluid until taken up by the lymphatic
`system. Porter and Charman have shown that the fraction of a dose absorbed into the
`lymphatic system is directly proportional to molecular weight [50]. Compounds
`with a molecular weight greater than 30 kD have been shown to be predominantly
`absorbed from into the lymphatic capillaries [52]. Colloidal particles may also be
`absorbed into the lymphatic system, with an optimal size for uptake reported to be
`10—100 nm [49] and larger particles taking longer to be absorbed [53]. Whilst
`absorption into the lymphatic vessels appears not to be selective, the rate of
`
`AstraZeneca Exhibit 2157 p. 12
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`4 Anatomy and Physiology of the Injection Site...
`
`65
`
`diffusion of the solute through the extracellular matrix of the interstitial fluid is
`determined by size, charge and hydrophilicity of the drug and so will influence
`lymphatic uptake [49, 50].
`The spread of formulations within the subcutaneous tissue is another potentially
`important variable affecting in vivo drug release as it will have an effect on the
`available surface area across which drug can escape from the delivery system. The
`influence of depot spreadability on drug absorption can be demonstrated by the
`effects of co—administration of formulations with the enzyme hyaluronidase. This
`enzyme degrades hyaluronic acid within the subcutaneous interstitial matrix and
`basement membrane, reducing the resistance to flow of injected material through
`the subcutaneous tissue [54]. Bookbinder et al. [55] described the effects on sub—
`cutaneous injection spread on co—administration of a recombinant form of
`hyaluronidase with trypan blue dye. The area of spread of the trypan blue dye was
`dose dependant over 0.05—5.0 units hyaluronidase per injection and because the
`enzyme acts only on the hyaluronic acid and not the collagen network, a significant
`structural matrix still existed. This enzyme also allowed the spread of small particu—
`lates (less than 200 nm) within the subcutaneous tissue [55].
`
`4.5 Effects of the Tissue Response on Extended
`Release Parenteral Systems
`
`For many extended release parenteral systems, extensive in vitro characterization is
`carried out and understanding of the effects of formulation and processing variables on
`the release characteristics are determined. In vitro release methods to study the release
`of drugs from extended release parenteral delivery systems make only small attempts
`to simulate the in vivo environment and methods used may be categorized as: sample
`and separate, continuous flow and dialysis—based methods as recently reviewed by
`Larsen et al. [51]. These methods mimic the poorly stirred, limited fluid conditions that
`are expected in intramuscular and subcutaneous sites and little is done to account for
`the effect of the tissue inflammatory reaction on release characteristics.
`For intravenous nanoparticulate systems, protein adsorption to the particulate
`surface and uptake by the reticuloendothelial system reduce the circulation time
`within the blood. Much recent work has shown that these effects can be reduced by
`surface pegylation so that residence time of nanoparticulate systems in the circula-
`tion are now significantly extended [56]. This strategy has had wide application in
`the formulation of particulate-targeted delivery systems. Ideally, the drug remains
`within the particulate system while it is circulating in the blood, but is released
`following deposition in the target tissue (e.g. tumor). In vitro evaluation of particle
`size and retention of the drug within the nanoparticle are important studies to char-
`acterize these delivery systems prior to in vivo use. However, it is the rate of in vivo
`particle opsonization, uptake by reticuloendothelial system cells and interactions
`with microvasculature that will ultimately determine the in vivo fate and effective-
`ness of intravenously injected nanoparticles.
`
`AstraZeneca Exhibit 2157 p. 13
`
`
`
`
`
`
`
`
`
`
`
`NewYork,ISBN:9781461405542
`
`
`
`
`
`
`
`
`
`©Wright,JeremyC.;Burgess,DianeJ.,Jan29,2012,LongActingInjectionsandImplantsSpringer,
`
`
`
`

`

`66
`
`A. McDowell and NJ. Medlicott
`
`toxic concentration
`
`
`
`Plasmaconcentration
`
`
`
`therapeutic range
`
`. minimum effective
`concentration
`
`Time
`
`Fig. 4.6 Plasma versus time profile for an ideal controlled release delivery system
`
`Intramuscular and subcutaneous extended release systems are expected to stay at
`the administration site and release drug slowly over time to achieve constant plasma
`drug concentrations as shown in Fig. 4.6. For some systems, the biological environ-
`ment has a large influence on the in vivo release profile and understanding how drug
`release occurs in vivo may allow optimization of in vivo release profiles to achieve
`target plasma drug concentrations. The biological environment can trigger physical
`change in the injected materials such as the solution to semi—solid transformation
`occurring for in situ gelling systems ([57—60]; see also Chap. 9). For others the
`opposite occurs and a solid material may be implanted, which hydrates to form a
`semi-solid depot at the injection site [61, 62]. In situ gelling implant materials tran—
`sition from liquid to solid in response to environmental changes such as temperature
`changes, e.g., poloxamer gels [63, 64], or loss of organic solvent into surrounding
`tissue, e.g., polylactide—co-glycolide organic solutions [65—67]. The tissue reaction
`at the implantation site can additionally influence the in vivo performance and may
`be involved in drug release. Kempe et al. described the in vivo solidification process
`for PLGA implants from n-methyl-2—pyrrolidone solutions as a two—stage process
`involving firstly surface solidification to form a shell over about 30 min, followed
`by a slower process of complete solidification over 24 h [67]. In the first stage of
`solidification they reported about 75% of the polymer precipitated. As well as alter-
`ing the physical form of the implanted material, the biological environment may
`affect drug release and implant degradation and erosion rates. An early example was
`reported by Olanoff et al. for a tetracycline containing methacrylate tri-laminate
`film system [68, 69]. These films released tetracycline with a zero-order profile in
`both in vitro studies [69] and following subcutaneous implantation in rats [68]. The
`tissue response 1—2 weeks after implantation was described as mild inflammatory
`with tissue oedema, loose granulation tissue and the beginning of fibrous capsule
`formation. The fibrous capsule developed further ove

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