throbber
(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT)
`(19) World Intellectual Property
`Organization
`International Bureau
`(43) International Publication Date
`28 December 2017 (28.12.2017) WIPO I PCT
`
`(10) International Publication Number
`WO 2017/223135 A1
`
`(51) International Patent Classification:
`A 61K 9/127 (2006.01)
`B82 F 40/00 (2011.01)
`A61K 9/133 (2006.01)
`A61M5/20 (2006.01)
`B82Y5/00 (2011.01)
`(21) International Application Number:
`PCT/US2017/038426
`
`(22) International Filing Date:
`
`21 June 2017 (21.06.2017)
`English
`English
`
`(25) Filing Language:
`(26) Publication Language:
`(30) Priority Data:
`24 June 2016 (24.06.2016)
`62/354,351
`US
`(71) Applicant: MODERNATX, INC. [US/US]; 200 Technol­
`ogy Square, Cambridge, MA 02139 (US).
`(72) Inventor: GELDHOF, Benjamin, Frank; 165 Woodside
`Avenue, Winthrop, MA 02152 (US).
`= (74) Agent: BELLIVEAU, Michael, J.; Clark & Elbing LLP,
`101 Federal Street, 15th Floor, Boston, MA 02110 (US).
`(81) Designated States (unless otherwise indicated, for every
`kind of national protection available)'. AE, AG, AL, AM,
`AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ,
`
`CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO,
`DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN,
`HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP,
`KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME,
`MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ,
`OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA,
`SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN,
`TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW.
`(84) Designated States (unless otherwise indicated, for every
`kind of regional protection available)'. ARIPO (BW, GH,
`GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ,
`UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ,
`TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK,
`EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, LV,
`MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM,
`TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW,
`KM, ML, MR, NE, SN, TD, TG).
`
`Declarations under Rule 4.17:
`as to applicant's entitlement to apply for and be granted a
`patent (Rule 4.17(ii))
`as to the applicant's entitlement to claim the priority of the
`earlier application (Rule 4.17(Hi))
`of inventorship (Rule 4.17(iv))
`
`(57) Abstract: The invention features methods and apparatus for producing lipid
`nanoparticles. Methods of the invention include inj ecting a lipid solution into an aque­
`ous solution at an automated rate (e.g., a rate controlled by a servo pump). The in­
`vention provides methods and apparatus for making lipid nanoparticles possessing a
`wide range of lipid components and hydrophilic encapsulants, including nucleic acids
`(e.g., mRNA). Also provided are nanoparticles and compositions thereof made by
`methods and apparatus of the invention.
`
`(54) Title: LIPID NANOPARTICLES
`
`FIG. 1
`
`Stepper Motor
`
`mi r
`
`I ead Screw with
`plunger attachment
`
`Syringe Barrel
`
`Pipette Tip Adaptor
`
`Pipette Tip
`
`'
`
`IT) m
`m
`CJ
`CJ
`
`o
`CJ o
`
`[Continued on next page]
`
`ARBUTUS - EXHIBIT 2037
`Moderna Therapeutics, Inc. v. Arbutus Biopharma Corporation
`IPR2019-00554
`
`

`

`WO 2017/223135 A1 llll II II11 III I II III III I III 11 III II III
`
`Published:
`with international search report (Art. 21(3))
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`LIPID NANOPARTICLES
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`BACKGROUND OF THE INVENTION
`Nanoparticles are useful for the delivery of various therapeutic, diagnostic, or experimental
`agents to cells and tissues. Nanoparticles are hypothesized to have enhanced interfacial cellular uptake
`because of their sub-cellular size, achieving a local effect. It is also hypothesized that there is enhanced
`cellular uptake of agents encapsulated in nanoparticles compared to the corresponding agent
`administered in free form. Thus, nanoparticle-entrapped agents have enhanced and sustained
`concentrations inside cells, thereby increasing therapeutic effects. Furthermore, nanoparticle-entrapped
`agents are protected from metabolic inactivation before reaching the target site, as often happens upon
`systemic administration of free agents. Therefore, the effective local nanoparticle dose required for the
`local pharmacologic effect may be several fold lower than with systemic or oral doses. Lipid
`nanoparticles, in particular, are useful in enhancing the delivery of agents such as nucleic acids.
`Widespread utility of lipid nanoparticles is limited, in part, due to manufacturing and processing
`constraints. In particular, large-scale production of lipid nanoparticle formulations can introduce variability
`in lipid nanoparticle characteristics, such as chemical composition, surface charge, size, batch-to-batch
`concentration, and purity. Such processing limitations have generated a need in the field for new
`methods and apparatus for synthesizing lipid nanoparticles.
`
`SUMMARY OF THE INVENTION
`The invention provides a method for producing lipid nanoparticles, the method including the steps
`of providing an aqueous solution; providing a lower alkanol solution including lipids; and injecting at an
`automated rate (e.g., at a rate controlled by a servo pump) the lower alkanol solution to the aqueous
`solution to produce the lipid nanoparticles. In some embodiments, the steps of the method of the
`invention are repeated one or more (e.g., 1,2,3, 4, 5, 6, 7, 8, 9, 10, or more) times.
`In some embodiments, the lipid nanoparticles have a mean diameter between 80 nm and 100 nm
`(e.g., between 82 nm and 98 nm, between 84 nm and 96 nm, between 86 nm and 94 nm, between 88 nm
`and 92 nm, about 80 nm, about 81 nm, about 82 nm, about 83 nm, about 84 nm, about 85 nm, about 86
`nm, about 87 nm, about 88 nm, about 89 nm, about 90 nm, about 91 nm, about 92 nm, about 93 nm,
`about 94 nm, about 95 nm, about 96 nm, about 97 nm, about 98 nm, about 99 nm, or about 100 nm) and
`a polydispersity index of 0.25 or less (e.g., 0.25, 0.24, 0.23, 0.22, 0.21, 0.20, 0.19, 0.18, 0.17, 0.16, 0.15,
`0.14, 0.13, 0.12, 0.11, 0.10, 0.09, 0.08, 0.07, 0.06, 0.05, 0.04, 0.03, 0.02, 0.01, or less).
`In some embodiments, the aqueous solution includes a nucleic acid (e.g., DNA, RNA, e.g.,
`mRNA). The nucleic acid may be at a concentration between 50 jjg per ml and 200 jjg per ml of the
`aqueous solution (e.g., about 50 jjg/ml, about 60 jjg/ml, about 70 jjg/ml, about 80 jjg/ml, about 90 jjg/ml,
`about 100 jjg/ml, about 110 jjg/ml, about 111 jjg/ml, about 111.11 jjg/ml, about 120 jjg/ml, about 130
`jjg/ml, about 140 jjg/ml, about 150 jjg/ml, about 175 jjg/ml, or about 200 jjg/ml). All of or a portion of the
`nucleic acid may be encapsulated in the lipid nanoparticles. In some embodiments, the method yields a
`nucleic acid encapsulation efficiency of at least 80% (e.g., 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%,
`88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater). In some embodiments,
`the method yields a nucleic acid encapsulation efficiency of at least 94%.
`
`1
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`In some embodiments, the lower alkanol solution provides 50% or less (e.g., between 25% and
`50%, e.g., 50%, 49%, 48%, 47%, 46%, 45%, 44%, 43%, 42%, 41%, 40%, 39%, 38%, 37%, 36%, 35%,
`34%, 33%, 32%, 31%, 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%,
`16%, 15%, 14%, 13%, 12%, 11 %, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1 %, or less) of the total
`volume. In some embodiments, the lower alkanol solution provides about 33% or about 25% of the total
`volume.
`In some embodiments, the injecting is at a rate from about 1,000 to about 5,000 microliters per
`second (|jl/s; e.g., from about 1,000 to about 5,000 |jl/s, from about 1,500 to about 4,500 jjl/s, from about
`2,000 to about 4,000 jjl/s, or from about 2,500 to about 3,000 jjl/s). For example, injecting may be at a
`rate of 2,600 (Jl/s.
`In some embodiments, the aqueous solution further includes a buffer. Examples of suitable
`buffers include, but are not limited to, a citrate buffer (e.g., 100 mM citrate buffer), a phosphate buffer
`(e.g., phosphate buffered saline (PBS)), or a IRIS buffer (e.g., TRIS/Sucrose). In some embodiments,
`the aqueous solution has a pH from about 3.0 to about 8.0 (e.g., about 3.0, about 3.1, about 3.2, about
`3.3, about 3.4, about 3.5, about 3.6., about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, about 4.2,
`about 4.3, about 4.4, about 4.5, about 4.6., about 4.7, about 4.8, about 4.9, about 5.0, about 5.1, about
`5.2, about 5.3, about 5.4, about 5.5, about 5.6., about 5.7, about 5.8, about 5.9, about 6.0, about 6.1,
`about 6.2, about 6.3, about 6.4, about 6.5, about 6.6., about 6.7, about 6.8, about 6.9, about 7.0, about
`7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.9, or about 8.0). The
`aqueous solution can have an osmolality from about 200 mOsm to about 400 mOsm (e.g., from about
`250 mOsm to about 350 mOsm, or about 260 mOsm, 270 mOsm, 280 mOsm, 290 mOsm, 300 mOsm,
`310 mOsm, 320 mOsm, 330 mOsm, 340 mOsm, or 350 mOsm).
`In some embodiments, the lower alkanol solution includes heptatriaconta-6,9,28,31-tetraen-19-yl-
`4-(dimethylamino)butanoate (DLin-MC3-DMA), phosphatidylcholine (1,2-distearoyl-sn-glycero-3-
`phosphocholine (DSPC), cholesterol, and polyethylene glycol-dimyristolglycerol (PEG-DMG). The ratio of
`DLin-MC3-DMA:DSPC:Cholesterol:PEG-DMG may be, for example, 50:10:38.5:1.5.
`In some embodiments, the method further includes purifying and/or concentrating the dispersion
`of lipid nanoparticles, e.g., through the use of a desalting column, dialysis, or tangential flow filtration.
`Purification and/or concentration may be performed as part of a buffer exchange procedure, e.g.,
`complete buffer exchange. The method may further include sterilizing the dispersion of lipid
`nanoparticles by filtration, e.g., microfiltration.
`In some embodiments, the invention provides a method of producing nanoparticles having an
`encapsulation efficiency of greater than 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%,
`99%, or 100%).
`In another aspect, the invention provides a lipid nanoparticle produced by injecting a lower
`alkanol solution into an aqueous solution having a lipid, wherein the injecting is automated at a rate from
`about 1,000 to about 5,000 microliters per second (jjl/s; e.g., from about 1,000 to about 5,000 jjl/s, from
`about 1,500 to about 4,500 jjl/s, from about 2,000 to about 4,000 jjl/s, or from about 2,500 to about 3,000
`|j|/s). In some embodiments, the injecting is at a rate of 2,600 jjl/s.
`In another aspect, the invention provides an apparatus for producing lipid nanoparticles, the
`apparatus having an injector configured to transfer a lower alkanol solution from a first reservoir to a
`
`2
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`second reservoir configured to hold an aqueous solution; and a servo pump configured to operate the
`injector at a rate from 1,000 to 5,000 jjl/s (e.g., from 1,500 to 4,000 jjl/s, 2,000 to 3,500 jjl/s, or from 2,500
`to 3,000 |jl/s. In some embodiments, the servo pump is configured to operate the injector at a rate of
`2,600 MI/S.
`In some embodiments, the injector is configured to move in three dimensions relative to the
`second reservoir. In some embodiments, the injector is configured to move in three dimensions relative
`to the first reservoir and second reservoir.
`In some embodiments, the invention provides a pharmaceutical composition including the lipid
`nanoparticles and a pharmaceutically acceptable carrier.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`FIGURE 1 is a schematic diagram of an injector including a stepper motor, lead screw with
`plunger, syringe barrel, pipette tip adaptor, and pipette tip.
`
`DEFINITIONS
`The term "nucleic acid" refers to a molecule of two or more nucleotides or alternative nucleotides.
`The term, "nucleotide" refers to a nucleoside including a phosphate group. The term "nucleoside" refers
`to a compound containing a sugar molecule (e.g., a pentose or ribose) or derivative thereof in
`combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to
`herein as a "nucleobase"). Examples of nucleic acids include but are not limited to DNA, RNA, tRNA
`(transfer RNA), mRNA (messenger RNA), siRNA (small interfering RNA), miRNA (micro RNA), shRNA
`(short hairpin RNA), ncRNA (non-coding RNA), aptamers, ribozymes, and shorter oligonucleotide
`sequences of any of the foregoing. Alterations of the base, sugar, and phosphate moiety of a nucleotide
`are encompassed by this definition. Herein, in a nucleotide, nucleoside or polynucleotide (such as the
`nucleic acids of the invention, e.g., mRNA molecule), the terms "alteration" or, as appropriate,
`"alternative" refer to alteration with respect to A, G, U or C ribonucleotides. Generally, herein, these
`terms are not intended to refer to the ribonucleotide alterations in naturally occurring S'-terminal mRNA
`cap moieties.
`As used herein, the terms "alteration" or "alternative" of a nucleotide, nucleoside, or
`polynucleotide (such as the polynucleotides of the invention, e.g., mRNA molecule), refer to alteration
`with respect to A, G, U or C ribonucleotides. Generally, herein, these terms are not intended to refer to
`the ribonucleotide alterations in naturally occurring 5'-terminal mRNA cap moieties.
`As used herein, the term "nanoparticle" refers to a particle having one or a plurality of
`components, the particle having any one structural feature on a scale of less than about 1000 nm that
`exhibits novel properties as compared to a bulk sample of the same material or component materials.
`Routinely, nanoparticles have any one structural feature on a scale of less than about 500 nm, less than
`about 400 nm, less than about 300 nm, less than about 200 nm or less than about 100 nm. In exemplary
`embodiments, a nanoparticle is a particle having one or more dimensions of the order of about 10-500
`nm. In other exemplary embodiments, a nanoparticle is a particle having one or more dimensions of the
`order of about 10-1000 nm. A spherical nanoparticle would have a diameter, for example, of between 10­
`100 nm or 10-1000 nm.
`
`3
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`A nanoparticle most often behaves as a unit in terms of its physical or biophysical properties,
`e.g., transport. It is noted that novel properties that differentiate nanoparticles from the corresponding
`bulk material typically develop at a size scale of under 1000 nm, or at a size of under 500 nm, but
`nanoparticles can be of a larger size, for example, for particles that are oblong, tubular, and the like. The
`size at which materials display different properties as compared to the bulk material is material-dependent
`and can be seen for many materials much larger in size than 100 nm and even for some materials larger
`in size than 1000 nm. Nanoparticles can be employed in a variety of drug delivery technologies (e.g.,
`nucleic acid drug delivery technologies) and can be employed for various purposes including, but not
`limited to, controlled drug delivery, protection of the drugs from degradation, and protection of the body
`from the toxic effects of the drugs.
`As used herein, the term "microparticle" refers to a small particle or particulate system, generally
`larger than about one micrometer (1 |im) in diameter and can be used to describe both microcapsules
`and microspheres.
`The term "lipid nanoparticle" refers to a nanoparticle as described above that includes lipids and
`that is stable and dispersible in aqueous media. In exemplary embodiments, lipid nanoparticles may be
`from 10 nm to 500 nm in diameter, e.g., from 70 nm to 120 nm.
`The term "lower alkanol" refers to an alcohol with 6 or fewer carbon atoms. Examples of lower
`alkanols include but are not limited to methanol, ethanol, propanol, pentanol, their isomers, and mixtures
`thereof.
`The term "injector" refers to a structure, through which a solution passes, that is configured to
`guide the flow of a solution into a reservoir.
`The term "injection rate" refers to the volume of fluid that is injected per unit time.
`The term "total volume after injecting" refers to the volume of suspension (e.g., including the
`lower alkanol solution, the aqueous solution, and any encapsulants) at a time point immediately following
`the termination of injecting, prior to any subsequent processing, such as filtration, lyophilization, etc.
`The term "particle size" or "particle diameter" refers to the mean diameter of the particles in a
`sample, as measured by dynamic light scattering (DLS), multiangle light scattering (MALS), nanoparticle
`tracking analysis, or comparable techniques. It will be understood that a dispersion of lipid nanoparticles
`as described herein will not be of uniform size but can be described by the average diameter and,
`optionally, the polydispersity index.
`In preferred embodiments, the lipid nanoparticles in the formulation of the present invention have
`a single mode particle size distribution (i.e., they are not bi- or poly-modal). The particle size distribution
`relates to the amount of particles by size within a given population. This is derived using Mie theory,
`where the assumption is that all particles are spherical, and the optical properties of the particles are
`known. A particle size distribution can be measured by dynamic light scattering (DLS) or other particle
`tracking systems (e.g., diffraction tracking and Brownian motion analysis).
`The term "encapsulation efficiency" as used herein refers to the percentage of nucleic acid in the
`lipid nanoparticles that is not degraded after exposure to serum or a nuclease assay that would
`significantly degrade free nucleic acids. Encapsulation efficiency can be measured as follows: Dilute the
`lipid nanoparticle formulation to about 1 -10 jjg/ml in 1 x TE buffer. Place 50 |il of the sample in a well in a
`polystyrene 96 well plate, and 50 |il in the well below it. Add 50 |il of 1x TE buffer to the top well, and 50
`
`4
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`of 2% Triton X-100 to the bottom well. For the reference wells, replace the sample with 50 |il of 1x TE
`buffer. Allow the 96 well plate to incubate at 370C for 15 minutes. During this time, allow RiboGreen® to
`thaw. Once thawed, dilute the RiboGreen 1:100 in 1x TE buffer. After the 15 minute incubation, add 100
`|il of diluted RiboGreen reagent to each well, mixing thoroughly by pipetting. Once addition of the
`RiboGreen is complete, the plate is then read by a fluorescence plate reader (FITC settings); after
`subtracting the fluorescence values of the blanks from each sample well, the percent of free mRNA may
`be determined by dividing the fluorescence of the intact liposome sample (no Triton X-100) by the
`fluorescence value of the disrupted liposome sample (with Triton X-100).
`
`Entrapped fraction = 1 - free fraction.
`Encapsulation efficiency = 100 x entrapped fraction.
`
`The term "fully encapsulated" as used herein indicates that the nucleic acid in the particles is not
`significantly degraded after exposure to serum or a nuclease assay that would significantly degrade free
`nucleic acids. In a fully encapsulated system, preferably less than 25% of particle nucleic acid is
`degraded in a treatment that would normally degrade 100% of free nucleic acid, more preferably less than
`10% and most preferably less than 5% of the particle nucleic acid is degraded. Fully encapsulated also
`indicates that the particles are serum stable, that is, that they do not rapidly decompose into their
`component parts upon in vivo administration.
`
`DETAILED DESCRIPTION
`The invention provides methods and apparatus for producing lipid nanoparticles that involve
`injecting a lipid solution into an aqueous solution at an automated rate. The methods can be used to
`make lipid nanoparticles possessing a wide range of lipid components including, but not limited to,
`cationic lipids, anionic lipids, neutral lipids, polyethylene glycol (PEG) lipids, hydrophilic polymer lipids,
`fusogenic lipids, and sterols. Hydrophobic agents can be incorporated into the organic solvent (e.g.,
`ethanol) with the lipid, and nucleic acids can be added to an aqueous component. The methods and
`apparatus can be used to prepare homogeneous dispersions of lipid nanoparticles.
`
`Methods
`Lipid nanoparticles can be produced (e.g., allowed to self-assemble, e.g., spontaneously) by
`injecting a lower alkanol solution containing lipids into an aqueous solution. Various lipids can be used to
`achieve desired properties, such as size, surface charge, and capacity for encapsulants. Such properties
`can also be influenced by the composition of the aqueous solution. Lipid nanoparticles of the invention
`can encapsulate a wide range of hydrophilic molecules, e.g., nucleic acids such as DNA and RNA, or
`alternative versions of DNA or RNA. Variations in lipid nanoparticle size can be affected by controlling
`process parameters. In particular, the rate at which the lower alkanol solution is injected into the aqueous
`solution is inversely related to the resulting lipid nanoparticle size. Similarly, minimizing variance in the
`rate of injection will minimize variance in lipid nanoparticle size, yielding homogeneous suspensions of
`lipid nanoparticles, e.g., within a single batch or among multiple batches. A precise rate of injection can
`be attained, e.g., through a servo pump.
`
`5
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`Typically, the lipid nanoparticles are liposomes with a lipid bilayer surrounding an aqueous
`interior. Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV)
`which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers
`separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller
`than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 nm and 500 nm
`in diameter. Liposome design may include, but is not limited to, opsonins or ligands in order to improve
`the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to,
`endocytosis. Liposomes may contain a low or a high pH in order to improve the delivery of the
`pharmaceutical formulations.
`The formation of liposomes may depend on the physicochemical characteristics such as, but not
`limited to, the pharmaceutical formulation entrapped and the liposomal ingredients, the nature of the
`medium in which the lipid vesicles are dispersed (e.g., osmolality or pH), the effective concentration of the
`entrapped substance and its potential toxicity, any additional processes involved during the application
`and/or delivery of the vesicles, the optimization size, polydispersity and the shelf-life of the vesicles for the
`intended application, and the batch-to-batch reproducibility and possibility of large-scale production of
`safe and efficient liposomal products.
`The invention further provides methods of injecting the lipid-containing lower alkanol solution into
`the aqueous solution at a precise rate. Without wishing to be bound by theory, a high rate of injection
`creates greater turbulence and provides more energy for lipid assembly, yielding small lipid nanoparticles.
`In some embodiments, the volume of the aqueous solution is at most 5.0 ml. In conditions in which the
`volume of the aqueous solution is 5.0 ml, the rate of injection of the lower alkanol solution can be
`between 1,000 |jl/s and 5,000 jjl/s (e.g., between 1,500 |jl/s and 4,000 jjl/s, between 2,000 and 3,000
`|j|/s, between 2,400 jjl/s and 2,800 jjl/s, or about 2,600 jjl/s), or between 20% and 100% (e.g., between
`30% and 80%, between 40% and 60%, or between 48% and 56%, e.g., about 52%) of the total volume
`per second. Lipid nanoparticles of the invention may have a diameter of, e.g., between 70 nm and 110
`nm (e.g., between 75 nm and 105 nm, between 80 nm and 100 nm, between 82 nm and 98 nm, between
`84 nm and 96 nm, between 86 nm and 94 nm, between 88 nm and 92 nm, about 85 nm, about 90 nm, or
`about 95 nm) with a polydispersity index of 0.25 or less (e.g., 0.25, 0.24, 0.23, 0.22, 0.21, 0.20, 0.19,
`0.18, 0.17, 0.16, 0.15, 0.14, 0.13, 0.12, 0.11, 0.10, 0.09, 0.08, 0.07, 0.06, 0.05, 0.04, 0.03, 0.02, 0.01, or
`less), as measured by dynamic light scattering (DLS). In some embodiments, all or a portion of the
`nucleic acid is encapsulated in the lipid nanoparticles. In some embodiments, the method yields a nucleic
`acid encapsulation efficiency of at least 80% (e.g., at least 80%, at least 85%, at least 90%, at least 92%,
`at least 94%, at least 95%, at least 96%, at least 97%, or at least 98%, e.g., 80%, 81%, 82%, 83%, 84%,
`85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater).
`The volume fraction may be modified according to parameters known in the art. For example, a
`lower alkanol solution having a greater concentration of lipid molecules may necessitate a lesser volume
`of lower alkanol solution relative to aqueous solution, e.g., to maintain the molar ratio of lipid to
`hydrophilic encapsulants.
`The lower alkanol solution can be injected automatically (e.g., at an automated rate, e.g., by an
`automatic injector, e.g., a servo-powered injector, e.g., as part of a robotic pipetting apparatus) into a
`volume of aqueous solution, e.g., contained within a reservoir, e.g., a well of a multi-well plate, test tube,
`
`6
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`

`

`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`WO 2017/223135
`
`PCT/US2017/038426
`
`or flask. Reservoirs can be part of a multi-reservoir unit, e.g., a multi-well plate or a rack of multiple tubes
`(e.g., 0.25 ml tubes, 0.5 ml tubes, 1.0 ml tubes, 1.5 ml tubes, 2.0 ml tubes, 2.5 ml tubes, 5.0 ml tubes, 10
`ml tubes, 15 ml tubes, or 50 ml tubes, e.g., included as part of a multiple tube rack, e.g., a 2-tube rack, a
`4-tube rack, a 6-tube rack, an 8-tube rack, a 10-tube rack, a 12-tube rack, a 16 tube rack, a 24-tube rack,
`a 48 tube rack, or a 96-tube rack). In some embodiments, one or more multi-reservoir units (e.g., tube
`racks) can be processed in a single run. The aqueous solution may be stationary or under mixing or
`agitation. Multiple, repeated injections, each into a corresponding reservoir of aqueous solution, can
`produce multiple suspensions of lipid nanoparticles. The injector can be moved relative to the aqueous
`solution reservoir, or, alternatively, the aqueous solution reservoir can be moved relative to the injector.
`An injector may additionally move in the Z direction, relative to the aqueous solution reservoir, e.g., such
`that ejection of lower alkanol solution from the injector occurs below the surface of the aqueous solution.
`Alternatively, ejection of the lower alkanol solution from the injector can occur above the surface of the
`aqueous solution, e.g., to prevent spillage and/or crossover of aqueous solution between reservoirs.
`The total volume of the lower alkanol solution per injection is generally equal to or less than the
`total volume of suspension upon injection completion (e.g., the mixture of lower alkanol solution and
`aqueous solution). In some embodiments, the volume of the lower alkanol solution per injection is
`between 10% and 100% (e.g., between 20% and 90%, between 30% and 80%, between 40% and 75%,
`between 50% and 70%, or between 60% and 70%, e.g., about 20%, about 30%, about 40%, about 50%,
`about 60%, about 65%, about 66.67%, about 70%, about 80%, about 90%, or about 100%) of the volume
`of the aqueous solution or between 5% and 50%, between 10% and 45%, between 15% and 40%,
`between 20% and 38%, or between 25% and 35%, e.g., about 20%, about 30%, about 33%, about 40%,
`or about 40% of the total volume of suspension upon injection completion). Other volume fractions can
`be used. In general, the greater the volume of the lower alkanol solution relative to the aqueous solution,
`the more concentrated the suspension of lipid nanoparticles will be upon completion of injection.
`Conventional downstream processing can be employed as part of the present invention. For
`example, lipid nanoparticles can be purified or concentrated, e.g., by tangential flow filtration, dialysis, or
`desalting column (e.g., a PD-10 desalting column). In methods involving dialysis, the filter membrane
`geometry, including the area of the filter and the fiber diameter can be varied to achieve an optimal rate of
`filtration according to known parameters, such as lipid nanoparticle size, encapsulants size, and liquid
`viscosity (e.g., buffer viscosity). A person of skill in the art will understand that the effect of varying each
`of these parameters can be informed by the Stokes-Einstein equation, below, where D is the diffusion
`constant of a particle, ka is Boltzmann's constant, Tis temperature, 77 is viscosity, and ris the radius of
`the particle.
`
`knT
`D =
`Gnrjr
`
`For tangential flow filtration procedures, the effect of liquid flow reduces the influence of diffusion
`on buffer exchange rate. In this case, increasing the recirculation rate will increase the shear rate and
`enhance the rate of buffer exchange. The transmembrane and permeate pressures can also be varied.
`
`7
`
`

`

`WO 2017/223135
`
`PCT/US2017/038426
`
`In some embodiments, the nanoparticle suspension can be exchanged with a solution containing
`a cryoprotectant (e.g., for long term storage in, e.g., frozen or lyophilized form). Physiologically suitable
`cyroprotectants for lipid nanoparticles are known in the art and include, e.g., sucrose, glucose, mannitol,
`glycerol, and other carbohydrates and polyalcohols. In one non-limiting example, a lipid nanoparticle
`solution is dialyzed against a sucrose solution (e.g., a TRIS/sucrose buffer).
`A sterile filtration step may also be employed, and the membrane area, pore size and filtration
`force can be varied, as described above. The lipid nanoparticles described herein may be made in a
`sterile environment by the system and/or methods described in U.S. Publication No. 20130164400.
`
`Lower Alkanol Solution
`A lower alkanol solution of the invention provides the lipid components that assemble into lipid
`nanoparticles upon injection into the aqueous solution. Lipid components that can be included in the
`lower alkanol solution include, but are not limited to, cationic lipids, anionic lipids, neutral lipids,
`polyethyleneglycol lipids, hydrophilic polymer lipids, and fusogenic lipids.
`In one embodiment, the lower alkanol solution includes at least one lipid. The lipid may be
`selected from, but is not limited to, L604, DLin-DMA, DLin-K-DMA, 98N12-5, C12-200, DLin-MC3-DMA,
`DLin-KC2-DMA, DODMA, PLGA, PEG, PEG-DMG, PEGylated lipids and amino alcohol lipids. In another
`aspect, the lipid may be a cationic lipid such as, but not limited to, DLin-DMA, DLin-D-DMA, DLin-MC3-
`DMA, DLin-KC2-DMA, DODMA and amino alcohol lipids. The amino alcohol cationic lipid may be the
`lipids described in and/or made by the methods described in U.S. Publication No. US20130150625,
`herein incorporated by reference in its entirety. As a non-limiting example, the cationic lipid may be 2-
`amino-3-[(9Z,12Z)-octadeca-9,12-dien-1 -yloxy]-2-{[(9Z,2Z)-octadeca-9,12-dien-1 -yloxy]methyl}propan-1 -
`ol (Compound 1 in US20130150625); 2-amino-3-[(9Z)-octadec-9-en-1-yloxy]-2-{[(9Z)-octadec-9-en-1-
`yloxy]methyl}propan-1-ol (Compound 2 in US20130150625); 2-amino-3-[(9Z,12Z)-octadeca-9,12-dien-1-
`yloxy]-2-[(octyloxy)methyl]propan-1-ol (Compound 3 in US20130150625); and 2-(dimethylamino)-3-
`[(9Z,12Z)-octadeca-9,12-dien-1-yloxy]-2-{[(9Z,12Z)-octadeca-9,12-dien-1-yloxy]methyl}propan-1-ol
`(Compound 4 in US20130150625); or any pharmaceutically acceptable salt or stereoisomer thereof.
`Lipid nanoparticle formulations may include, e.g., an ionizable cationic lipid, for example, 2,2-
`dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane (DLin-KC2-DMA), dilinoleyl-methyl-4-
`dimethylaminobutyrate (DLin-MC3-DMA), or di((Z)-non-2-en-1-yl) 9-((4-
`(dimethylamino)butanoyl)oxy)heptadecanedioate (L319), and further comprise a neutral lipid, a sterol and
`a molecule capable of reducing particle aggregation, for example a PEG or PEG-modified lipid.
`In one embodiment, the lower alkanol solution includes (i) at least one lipid selected from the
`group consisting of 2,2-dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane (DLin-KC2-DMA), dilinoleyl-methyl-
`4-dimethylaminobutyrate (DLin-MC3-DMA), and di(

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