throbber
CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 237
`
`Liposomal Formulations for Nucleic Acid Delivery
`
`CHAPTER 9
`
`Ian MacLachlan
`
`CONTENTS
`
`9.1 Liposomes for the Delivery of Nucleic Acid Drugs.............................................................237
`9.2 Liposome Constituents .........................................................................................................239
`9.2.1 Cationic Lipids .........................................................................................................239
`9.2.2 The Role of Helper Lipids in Promoting Intracellular Delivery..............................240
`9.2.3 PEG–Lipids...............................................................................................................241
`9.2.4 Active Targeting........................................................................................................242
`9.3 Methods of Encapsulating Nucleic Acids ............................................................................242
`9.3.1 Passive Nucleic Acid Encapsulation.........................................................................243
`9.3.2 The Ethanol Drop (SALP) Method of Nucleic Acid Encapsulation........................247
`9.3.3 Encapsulation of Nucleic Acid in Ethanol-Destabilized Liposomes .......................247
`9.3.4 The Reverse-Phase Evaporation Method of Nucleic Acid Encapsulation ...............248
`9.3.5 The Spontaneous Vesicle Formation by Ethanol Dilution (SNALP) Method of
`Nucleic Acid Encapsulation .....................................................................................249
`9.4 Analytical Methods...............................................................................................................251
`9.4.1 Measuring Particle Size ............................................................................................251
`9.4.2 Zeta Potential............................................................................................................253
`9.4.3 Encapsulation............................................................................................................253
`Pharmacology of Liposomal NA..........................................................................................254
`9.5.1 Pharmacokinetics and Biodistribution of Liposomal NA Following Systemic
`Administration ..........................................................................................................254
`9.5.2 Toxicity of Liposomal NA Formulations .................................................................256
`9.5.3 Immune Stimulation .................................................................................................258
`9.5.4 Immunogenicity........................................................................................................259
`9.5.5 The Efficacy of Liposomally Formulated NA Drugs...............................................260
`References ......................................................................................................................................262
`
`9.5
`
`9.1 LIPOSOMES FOR THE DELIVERY OF NUCLEIC ACID DRUGS
`
`Liposomes are artificial vesicles made up of one or more bilayers of amphipathic lipid encap-
`sulating an equal number of internal aqueous compartments. They are distinguished on the basis
`of their size and the number and arrangement of their constituent lipid bilayers (Figure 9.1).
`
`© 2007 by Taylor & Francis Group, LLC
`
`237
`
`Moderna Ex 1024-p. 1
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 238
`
`238
`
`ANTISENSE DRUG TECHNOLOGIES, SECOND EDITION
`
`Figure 9.1 Liposomes. Mulilamellar vesicles (MLVs) are large (hundreds of nm in diameter) complex structures
`containing a series of concentric bilayers separated by narrow aqueous compartments. Large
`unilamellar vesicles (LUVs) are between 50 and 500 nm in diameter, while the smallest liposomes
`namely small unilamellar vesicles (SUVs) are ⬍50 nm. LUVs are the preferred systems for delivery
`of NA drugs. Lipids are drawn roughly to scale.
`
`Multilamellar vesicles (MLVs) are formed by the aqueous hydration of dried lipid films. Typically
`hundreds of nanometers in diameter, they are large, complex structures containing a series of con-
`centric bilayers separated by narrow aqueous compartments. Simple unilamellar vesicles between
`50 and 500 nm in diameter are referred to as large unilamellar vesicles (LUVs) while the smallest
`liposomes, vesicles smaller than 50 nm in diameter, are small unilamellar vesicles (SUVs).
`Liposomes have received attention not only for their utility as model membrane systems, but
`also for use in drug delivery. Typically, liposomes are used as drug carriers, with the solubilized
`drug encapsulated in the internal aqueous space formed by the liposomal lamellae. Liposomal drug
`formulations can be used to overcome a drug’s nonideal properties, such as limited solubility,
`serum stability, circulation half-life, biodistribution, and target tissue selectivity. Experience with
`conventional small molecule drugs has shown that the drugs which benefit the most from liposo-
`mal delivery, are those that are chemically labile, subject to enzymatic degradation and have an
`intracellular site of action [1]. For this reason, there is considerable interest in exploiting lipo-
`somes as carriers of nucleic acids (NAs), either as plasmid vectors for gene therapy applications
`or to deliver smaller NA species such as antisense oligonucleotides, ribozymes and, more recently,
`siRNA for the purposes of downregulating target genes. Because of their ability to achieve favor-
`able drug/lipid ratios and their more predictable drug release kinetics LUV are the preferred lipo-
`some delivery system for NA drugs.
`An advantage of liposomal drug delivery is that the pharmacokinetics, biodistribution, and intra-
`cellular delivery of the liposome payload are largely determined by the physicochemical properties
`of the carrier. For example, the biodistribution of a NA entrapped within a small, long circulating
`liposome is independent of the type of NA, which can be a relatively stable double-stranded plasmid
`DNA molecule or single-stranded antisense DNA, or one of the more labile ribonucleotide
`molecules such as ribozymes or a duplex siRNA. This is only true if the liposome is truly acting as
`a carrier, rather than a mere excipient. Liposomes function as excipients when used to formulate
`hydrophobic drugs that would otherwise be difficult to administer in aqueous dosage form.
`Hydrophobic drugs rapidly exchange into lipoproteins or other lipid-rich environments soon after
`injection, resulting in comparably uncontrolled pharmacology. In the context of NA drug delivery,
`liposomes are considered excipients if used to enable vialing and aqueous dosing of hydrophobic
`lipid–NA conjugates [2–5]. (These applications are not considered in this chapter, nor are those that
`use preformed, cationic lipid-containing vesicles to form “lipoplex” or “oligoplex” systems.)
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 2
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 239
`
`LIPOSOMAL FORMULATIONS FOR NUCLEIC ACID DELIVERY
`
`239
`
`An objective inherent in all pharmaceutical development is to minimize the risks associated with
`treatment while maximizing the benefit to patient health. The most important risk to patients is the
`toxicity associated with the administration of poorly tolerated compounds, often exacerbated by
`attempts to increase efficacy by escalating the administered dose. A well-designed liposomal delivery
`system will be capable of reducing the toxicity and increasing the potency of NA-based drugs by
`optimizing NA delivery to target tissues. Liposomal NA delivery will be determined by the physical
`and biochemical properties of the liposome including stability, size, charge, hydrophobicity, interac-
`tion with serum proteins, and interaction with nontarget cell surfaces. Ideally, liposomal carriers for
`NA delivery will have the following properties: (i) they will be safe and well tolerated; (ii) they will
`have appropriate pharmacokinetic attributes to ensure delivery to intended disease sites; (iii) they will
`mediate effective intracellular delivery of intact NA; (iv) they will be nonimmunogenic, enabling
`the use of multidosing treatment regimes; and (v) they will be stable upon manufacture so that large
`batches can be prepared with uniform, reproducible specifications. In this chapter we discuss the
`physical makeup, manufacturing methods, and pharmacological considerations specific to liposomal
`systems for the delivery of NA-based drugs, with emphasis on those that enable systemic delivery of
`synthetic polynucleotides such as antisense ODN, ribozymes, and siRNA.
`
`9.2 LIPOSOME CONSTITUENTS
`
`NA encapsulation was first described in the late 1970s, prior to the development of cationic lipid-
`containing lipoplex, using naturally occurring, neutral lipids to encapsulate high-molecular-weight
`DNA [6–8]. The first reports of low-molecular-weight oligo- or polynucleotide encapsulation
`similarly used passive techniques to entrap NA in neutral liposomes [9–11]. The advent of cationic
`lipid-mediated lipofection [12] saw a shift in emphasis away from encapsulated systems in favor of
`“lipoplex” or “oligoplex” systems. More recently, improvements in formulation technology have
`allowed for a return to encapsulated systems that contain cationic lipids as a means of facilitating
`both encapsulation and intracellular delivery. More advanced systems typically contain multiple lipid
`components, each of which play a role in determining the physical and pharmacological properties
`of the system as a whole.
`
`9.2.1 Cationic Lipids
`
`Cationic lipids play two roles in liposomal NA formulations. In the first case, they encourage
`interaction between the lipid bilayer and the negatively charged NA, allowing for the enrichment
`of NA concentrations over and above that which would be achieved using passive loading in charge neu-
`tral liposomes. Cationic lipids allow for encapsulation efficiencies greater than 40% when using
`coextrusion methods, and greater than 95% when using more sophisticated techniques [13–15].
`Cationic lipids also function by providing the liposome with a net positive charge, which in turn
`enables binding of the NA complex to anionic cell surface molecules. The most abundant anionic
`cell surface molecules, sulfated proteoglycans and sialic acids, interact with and are responsible for
`the uptake of cationic liposomes [16–18]. The role of cationic lipids in liposomal uptake presents
`a dilemma: highly charged systems are rapidly cleared from the blood, thereby limiting accumula-
`tion in target tissues. Particles with a neutral charge however, display good biodistribution profiles,
`but are poorly internalized by cells. This supports the concept of a modular delivery solution, that
`is, an engineered nanoparticle with individual components fulfilling different functions in the
`delivery process, and in particular, a system which responds to the microenvironment in a manner
`that facilitates transfection. Titratable, ionizable lipids are components that allow for the adjust-
`ment of the charge on the system by simply changing the pH after encapsulation [19]. At reduced
`pH when the system is strongly charged, NAs are efficiently encapsulated. When liposomes
`containing titratable, ionizable lipids are at a pH closer to the pKa of the cationic lipid, such as
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 3
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 240
`
`240
`
`ANTISENSE DRUG TECHNOLOGIES, SECOND EDITION
`
`physiological pH, they become more charge neutral and are able to avoid opsonization by blood
`components [19]. More recently, the use of novel, pH titratable cationic lipids with distinct
`physicochemical properties that regulate particle formation, cellular uptake, fusogenicity, and
`endosomal release of NA drugs have been described [20]. The chemical and biological properties
`of pH-titratable cationic lipids are influenced by their degree of lipid saturation. In particular, the
`phase transition properties, as measured using 31P-NMR, are affected. Above the phase transition
`temperature, Tc, lipids adopt the more highly fusogenic reverse hexagonal HII phase [20–22]. By
`noting the temperature at which this phase transition occurs, the relative ease with which lipids
`form the HII phase and become “fusogenic” can be determined. On this basis it has been shown
`that the fusogenicity of liposomal systems increases as the titratable cationic lipid becomes less
`saturated. The lipid pKa also correlates with the degree of saturation. pK measurements confirm
`that saturated lipids carry more residual charge at physiological pH. For this reason, liposomes
`containing the more highly saturated cationic lipids are taken up more readily by cells in vitro [20].
`However, liposomes containing the more fusogenic unsaturated cationic lipids DLinDMA and
`DLenDMA are more effective at mediating RNA interference in both in vitro cell culture systems
`and in vivo. The apparently conflicting results between cellular uptake and silencing potency are
`a reminder that cellular uptake per se is insufficient for effective delivery of NA. Cellular uptake,
`fusogenicity, and endosomal release are distinct processes, each of which need to be enabled by
`the delivery vehicle and each of which are profoundly affected by the physicochemical properties
`of the cationic lipids used.
`
`9.2.2 The Role of Helper Lipids in Promoting Intracellular Delivery
`
`Although we have just shown that cationic lipids may have inherent fusogenic properties of
`their own, cationic lipids were originally believed to require fusogenic “helper” lipids for effi-
`cient NA delivery [23–26]. Fusogenic liposomes facilitate the intracellular delivery of complexed
`plasmid DNA by fusing with the membranes of the target cell. Fusion may occur at a number of
`different stages in delivery, either at the plasma membrane, endosome or nuclear envelope.
`Fusion of first-generation, nonencapsulated lipoplex systems with the plasma membrane is
`expected to be a particularly inefficient method of introducing NA into the cytosol. Since
`lipoplex-NA is predominantly attached to the surface of the liposome, lipoplex fusion events
`resolve with NA, formerly attached to the liposome surface, deposited on the outside surface of
`the plasma membrane. Encapsulated systems are significantly different from lipoplex in this
`respect. Upon fusion with either the plasma or endosomal membrane(s), encapsulated carriers
`deliver their contents directly into the cytosol.
`Lipids that preferentially form nonbilayer phases, in particular the reverse hexagonal HII phase,
`such as the unsaturated phosphatidylethanolamine DOPE, promote destabilization of the lipid
`bilayer and fusion. Similar to fusogenic cationic lipids, decreasing the degree of lipid saturation
`increases the lipid’s affinity for the fusogenic HII phase [27–32]. However, some cationic lipids can
`function in the absence of these so-called helper lipids, either alone [24,25] or in the presence of
`the nonfusogenic lipid cholesterol [33]. This would suggest that either these lipids have properties
`which promote delivery through a mechanism which does not require membrane fusion, or that
`their own fusogenic properties are adequate to support delivery. As described above, cationic lipids
`are readily designed for optimal fusogenicity by controlling lipid saturation. This provides for
`multiple opportunities for modulating the fusogenicity of a liposomal lipid bilayer [20].
`Attempts to address the role of fusogenic lipids in vivo have yielded confounding results. In this
`regard it is important to distinguish the effect of fusogenic lipids on NA delivery to target tissue
`from their effect on intracellular delivery. Fusogenic formulations are more likely to interact
`with the vascular endothelium, blood cells, lipoproteins, and other nontarget systems while in the
`blood compartment. For this reason there may be an advantage to transiently shield the fusogenic
`potential of systemic carriers using shielding agents such as polyethylene glycol (PEG).
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 4
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 241
`
`LIPOSOMAL FORMULATIONS FOR NUCLEIC ACID DELIVERY
`
`241
`
`9.2.3 PEG–Lipids
`
`An ideal delivery system would be one that is transiently shielded upon administration, facili-
`tating delivery to the target site, yet becomes increasingly charged and fusogenic as it reaches the
`target cell. PEG lipids partially address this challenge. PEG–lipid conjugates are readily incorpo-
`rated in liposomal NA formulations. They provide a benefit during the formulation process, stabi-
`lizing the nascent particle and contribute to formulation stability by preventing aggregation in the
`vial [13]. PEG conjugates sterically stabilize liposomes by forming a protective hydrophilic layer
`that shields the hydrophobic lipid layer. By shielding the liposome’s surface charge they prevent the
`association of serum proteins and resulting uptake by the reticuloendothelial system when liposomes
`are administered in vivo [34,35]. In this way, cationic liposome NA formulations are stabilized in a
`manner analogous to PEGylated liposomal drug formulations that exhibit extended circulation life-
`times [36–41]. Although this approach has been investigated with a view towards improving the sta-
`bility and pharmacokinetics of lipoplex containing either plasmid DNA [42] or antisense
`oligonucleotides [43], PEG–lipid-containing lipoplex systems suffer from the heterogeneity and
`suboptimal pharmacology common to most nonencapsulated NA–cationic lipid complexes.
`Although PEG–lipid-containing systems are promising with respect to their ability to deliver NA
`to disease sites, improvements are required to increase their potency. Early PEGylated liposomes for
`the delivery of small molecule chemotherapeutic drugs utilized stably integrated PEG lipids such as
`PEG-DSPE [39]. These systems are designed to function as carriers that facilitate the accumulation of
`active drug compound at disseminated disease sites. The drug is released at the cell surface at a “leak-
`age rate” determined by the liposomal bilayer composition. NA-based drugs differ in this respect in
`that they require effective intracellular delivery, hence the use of the cationic and fusogenic lipids
`described earlier. PEGylated systems typically exhibit relatively low-transfection efficiencies. This is
`mainly due to the ability of the PEG coating to inhibit cell association and uptake [23,44,45]. Ideally,
`PEG–lipid conjugates would have the ability to dissociate from the carrier and transform it from a sta-
`ble, stealthy particle to a transfection-competent entity at the target site. Various strategies have been
`applied to this problem. A number of investigators have explored the use of chemically labile
`PEG–lipid conjugates [46–52], in particular those that are “pH sensitive.” Typically, these systems
`invoke a chemically labile linkage between the lipid and PEG moieties that reacts via acid-catalyzed
`hydrolysis to destabilize the liposomes by removal of the sterically stabilizing PEG layer. Although
`this approach results in improved performance both in vitro and in vivo, it may be regarded as sub-
`optimal for two reasons. First, pH-sensitive PEG lipids are designed to be rapidly hydrolyzed in the
`reduced pH environment encountered within the endosome, but since PEG lipids are known to inhibit
`cellular uptake, a prerequisite to endosomal localization and hydrolysis, their use actually limits the
`amount of material delivered to the endosome [53]. Second, the incorporation of pH-sensitive or oth-
`erwise chemically labile lipids results in a truncation of formulation shelf life relative to systems that
`use more stable PEG–lipids. An alternative to the use of acid-labile PEG–lipids involves the use of
`chemically stable, yet diffusible PEG lipids.
`The concept of diffusible PEG lipids arose from the observation that the length of the PEG lipid
`anchor has an influence on PEG lipid retention and the stability and circulation lifetime of empty
`lipid vesicles [54]. It has been found that by modulating the alkyl chain length of the PEG lipid
`anchor [55–59], the pharmacology of encapsulated NA can be controlled or “programmed” in a
`predictable manner. Upon formulation, the liposome contains a full complement of PEG in steady-state
`equilibrium with the contents of the vial. In the blood compartment, this equilibrium shifts and the
`PEG–lipid conjugate is free to dissociate from the particle over time, revealing a positively charged
`and increasingly fusogenic lipid bilayer that transforms the particle into a transfection-competent
`entity. Diffusible PEG lipids differing in the length of the their lipid anchors have been incorporated
`into liposomal systems containing plasmid DNA (SPLP) [13,55], antisense oligonucleotides (PFV,
`SALP) [19,56,60], and siRNA (SNALP) [14,15,61]. This approach may help to resolve the two
`conflicting demands imposed upon NA carriers. First, the carrier must be stable and circulate long
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 5
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 242
`
`242
`
`ANTISENSE DRUG TECHNOLOGIES, SECOND EDITION
`
`enough to facilitate accumulation at disease sites. Second, the carrier must be capable of interact-
`ing with target cells to facilitate intracellular delivery.
`
`9.2.4 Active Targeting
`
`Active targeting refers to processes that aim to increase the accumulation, retention or internal-
`ization of a drug through the use of cell-specific ligands. This is to be distinguished from the passive
`“disease site targeting” or the “enhanced permeability and retention” (EPR) effect, which results
`in the accumulation of appropriately designed carriers in target sites such as tumor tissue. Active
`targeting has been successfully applied to liposomal small molecule drug formulations and generally
`has the effect of improving the therapeutic index of the liposomal drug when measured in preclinical
`studies. NA delivery systems stand to benefit from targeting in two ways, first through improving the
`accumulation and binding of formulations to target cells and second by facilitating intracellular
`delivery through endocytosis. The perceived benefits of active targeting have encouraged numerous
`investigators in this area and targeting of NA formulations has been achieved through the use of
`molecules as diverse as antibodies directed against cell surface proteins [62–65], protein ligands of
`cell surface receptors [66–69], vitamins [70–72], and glycolipids [73,74].
`The earliest reports of targeted liposomal formulations of encapsulated NA were attempts to
`improve the intracellular delivery characteristics of charge neutral liposomes encapsulating either
`synthetic antisense DNA [63,65] or in vitro transcribed antisense RNA [64]. The results of these
`studies were encouraging, suggesting a significant benefit associated with the use of targeted
`systems. Although these in vitro studies effectively demonstrated the potential advantage of targe-
`ting at the level of intracellular delivery, they were unable to address important pharmacological
`considerations such as those that influence accumulation at disease sites. It is unlikely that addition
`of targeting ligands to delivery systems that are rapidly removed from the circulation will result
`in delivery exceeding that achieved by systems that display passive disease site targeting. For this
`reason many investigators have pursued approaches involving the addition of targeting ligands to
`sterically stabilized and charge shielded systems, such as those containing PEG lipids [71,72,75–77].
`This approach has been advanced, in part, by the development of the so-called postinsertion technique
`[78]. Postinsertion allows for the insertion of ligand–PEG–lipid conjugates into preformed liposomes
`containing encapsulated NA. This represents a significant improvement on earlier approaches in
`which ligands were chemically coupled to preformed liposomes, an approach limited by suboptimal
`coupling efficiencies, or where ligand–lipid conjugates were incorporated in the first stages of the
`formulation process, an approach limited by the resulting negative impact on NA encapsulation
`efficiency and subsequent suboptimal presentation of the targeting ligand.
`A number of reports suggest that it is possible to design encapsulated systems containing
`targeting ligands that retain extended circulation lifetimes and passive disease site targeting the
`following systemic administration. It remains to be seen if the benefits of active targeting outweigh
`the increased cost, manufacturing complexity and immunogenicity that often accompanies the use
`of such technology.
`
`9.3 METHODS OF ENCAPSULATING NUCLEIC ACIDS
`
`To capitalize on the pharmacology of liposomal drug carriers it is necessary to completely entrap
`NA within the contents of a liposome. In this regard it is important to distinguish first-generation
`“lipoplex” or “oligoplex” systems from those that truly encapsulate their NA payload. Lipoplex are elec-
`trostatic complexes formed by mixing preformed cationic lipid-containing vesicles with NA [12,79,80].
`The result is a heterogenous, metastable aggregate that is effective when used to transfect cells in cul-
`ture but has relatively poor performance in vivo. Upon systemic administration, lipoplex systems are
`rapidly cleared from the blood, accumulating in the capillary bed of first-pass organs such as the lung.
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 6
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 243
`
`LIPOSOMAL FORMULATIONS FOR NUCLEIC ACID DELIVERY
`
`243
`
`Lipoplex are effectively taken up by the cells of the innate immune system, contributing to their pro-
`found toxicities and off-target effects. These side effects may manifest as “efficacy” in antitumor or anti-
`infective applications, confounding data interpretation and encouraging the acceptance of false-positive
`results. For these reasons, an abundance of caution is encouraged when initiating in vivo studies that use
`liposomes to deliver NA. Of particular importance is the use of appropriate analytical methodology,
`described in Section 9.4, to properly characterize lipid-based systems prior to and during use.
`
`9.3.1 Passive Nucleic Acid Encapsulation
`
`Liposomal encapsulation of small molecule drugs may be achieved by either “passive” or “active”
`loading. Unlike small molecule drugs, NAs are not readily packaged in preformed liposomes using
`pH gradients or other similar active loading techniques. This is predominantly due to the large size
`and hydrophilic nature of NA, which conspire to prevent them from crossing intact lipid bilayers. For
`this reason, much of the work on NA encapsulation has utilized passive loading technology.
`Passive encapsulation typically involves the preparation of a “lipid film,” the lipidic residue that
`remains after evaporation of the organic phase of a lipid solution (Figure 9.2). Rehydration of the
`
`Lipid solution in solvent
`
`Nucleic acid solution
`in buffer
`
`Dried lipid film
`
`Lipid hydration with
`nucleic acid solution
`
`MLV formation
`by freeze/thaw (5−10×)
`
`MLV extrusion (10x)
`
`LUV collection
`
`Free nucleic acid removal
`
`Sample concentration
`
`Sterile filtration
`
`Figure 9.2 Passive method of NA encapsulation. Passive encapsulation utilizes a dried lipid film prepared by
`evaporating the organic phase of a lipid solution. The resulting lipid film is rehydrated in an aqueous
`solution of NA in buffer, forming MLV. Multiple freeze-thaw cycles increase the extent of NA encap-
`sulation within the MLV bilayers. The vesicles are then extruded through polycarbonate filters
`producing LUV.
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 7
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 244
`
`244
`
`ANTISENSE DRUG TECHNOLOGIES, SECOND EDITION
`
`lipid film in aqueous media, typically buffer containing NA, followed by vigorous mixing, results
`in the formation of MLV. This is followed by multiple cycles of freezing and thawing to increase
`the extent to which the NA solute is entrapped by the MLV bilayers. The MLV preparation is then
`subjected to multiple rounds of extrusion through polycarbonate filters to produce LUV (Figure 9.2
`and Figure 9.3) [81]. The size of the LUV is determined by the size of the filter pores. This process
`suffers from a number of limitations. When used to encapsulate NA, the efficiency of passive encap-
`sulation is generally quite low, ranging from 3 to 45%, depending on the composition of the lipid
`bilayer and other factors (Table 9.1). The low encapsulation efficiency, consequently, necessitates
`the incorporation of a postencapsulation separation step such as dialysis, size exclusion chro-
`matography or ultrafiltration to remove nonencapsulated NA. In an effort to improve the efficiency
`of encapsulation, excess lipid is often incorporated in the formulation process, resulting in low
`NA/lipid ratios which ultimately impact toxicity and cost of goods. Finally, the extrusion process is
`inherently difficult to scale. Preparation of large batches requires the use of custom-built extruders
`to accommodate large filters. The probability of filter tears, resulting in batch failure, increases
`as the size and cost of the batch increases. In spite of these process limitations, extrusion-based
`methods for liposome preparation have been successfully adopted by many laboratories, presum-
`ably because the technology is readily accessible to the casual investigator. Furthermore, significant
`progress has been made adapting or enhancing extrusion-based processes for the liposomal formu-
`lation of NA-based drugs. These include the use of cationic and anionic lipids [82,83], ionizable
`cationic lipids [19,84], PEG lipids [85], and detergent or organic solvents such as ethanol [19,60]
`to control bilayer assembly.
`
`Figure 9.3 The Lipex™ thermobarrel extruder for the preparation of uniformly sized liposomes by extrusion. An
`MLV or other vesicle preparation is introduced to the top of the extruder and the extruder is pres-
`surized with nitrogen, forcing the MLV through a polycarbonate filter of defined pore size. The resulting
`LUVs are collected via the outlet port at the bottom of the device. Extrusion is repeated, typically for
`a total of 10 passes. The unit permits thermostatic operation by virtue of the thermobarrel, which
`can be coupled to a circulating water bath. Photo courtesy Northern Lipids Inc., Vancouver, Canada,
`http://www.northernlipids.com.
`
`© 2007 by Taylor & Francis Group, LLC
`
`Moderna Ex 1024-p. 8
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`CRC_8796_ch009.qxd 2/21/2007 2:43 PM Page 245
`
`LIPOSOMAL FORMULATIONS FOR NUCLEIC ACID DELIVERY
`
`245
`
`(Continued)
`
`[183]
`[182]
`[181]
`[180]
`[72]
`[179]
`[178]
`[177]
`[176]
`
`[175]
`[174]
`[173]
`[172]
`[93]
`[171]
`[170]
`[169]
`[168]
`[167]
`[92]
`[166]
`[165]
`[164]
`[163]
`[162]
`[161]
`[160]
`[159]
`[158]
`[157]
`[85]
`[156]
`#1626
`
`[155]
`[154]
`[63]
`[153]
`
`[65]
`
`Reference
`
`Payload
`
`Encapsulation (%)
`
`Size (nm)
`
`Lipid Composition
`
`Formulation Method
`
`Table 9.1Liposomal Formulations of Oligo- and Polynucleotide Drugs
`
`ODN
`ODN
`ODN
`ODN
`Various
`ODN
`ODN
`ODN
`
`ODN
`ODN
`ODN
`ODN
`ODN
`ODN
`TFD, ODN
`ODN
`ODN
`pDNA
`ODN
`ODN
`RNA Aptamer
`Plasmid/ODN
`ODN
`ODN
`Ribozyme
`ODN
`ODN
`ODN
`ODN
`ODN
`ODN
`
`ODN
`ODN
`ODN
`ODN
`ODN
`
`Up to 20
`
`10–30
`
`ND
`2–5
`ND
`
`ND
`7–15
`ND
`⬎85
`
`43.5⫾4
`
`ND
`
`ND
`
`10–15
`80–100
`⬃16
`ND
`10
`ND
`ND
`
`ND
`88
`60
`⬍10
`⬎90
`⬍10
`⬍10
`30–40
`⬍10
`
`10–75
`
`ND
`
`24–32
`15–20
`
`2–3
`10
`⬍2
`
`3
`
`⬍10
`
`467.2⫾72.0
`
`250–300
`
`ND
`
`90–100
`90–110
`240–370
`
`130
`ND
`ND
`
`⬍2000
`220⫾55
`110⫾40
`100–140
`
`ND
`
`ND
`ND
`
`100–150
`
`ND
`ND
`
`50–65
`
`200–300
`
`400–500
`316–562
`
`ND
`
`110⫹30
`⬍200
`
`ND
`ND
`ND
`ND
`
`50–70
`
`ND
`
`100–140
`
`170
`
`460⫾200
`
`⬃200
`220⫾55
`
`DSPC:Chol:CPL
`HVJ liposome:PS:PC:Chol
`Thiocationic lipid:oleic acid:Vitamin D
`PE:CHEMS:LLO
`Folate liposomes:EPC:Chol:DSPE-PEG-Pteroate
`PE:CHEMS:LLO
`HVJ liposome:PS:PC:Chol
`DDAB:EPC:Chol
`CHEMS:DOPE or conventional SPC liposomes
`immunoliposomes
`HVJ liposome:PE-DTP:PS:PC:Chol
`DPPC:DMPG
`HVJ liposome:PS:PC:Chol
`EPC:Chol
`PC40:Chol:PEG-DSPE:DOTAP
`DOPE:CHEMS or SPC
`HVJ liposome
`DPPE:Cetyltrimethyl ammonium bromide
`HVJ liposome:PC:DOPE:Sph:PS:Chol
`HVJ liposome:PS:PC:Chol
`DPPC:Chol:DPPS or DPPA
`EPC:Chol:Folate-PEG-DSPE
`DSPC:Chol
`HVJ liposome:Chol:PC:PS
`HVJ liposome
`DOGS:DOPE
`HVJ liposome:PC:Chol:DC-Chol
`PC:Chol:PS
`DDAB:PC:Chol
`DOPE:Chol:Oleic Acid:Palmitoyl-CD4
`PC:Ch

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