throbber
Letter
`
`pubs.acs.org/NanoLett
`
`Optimization of Lipid Nanoparticle Formulations for mRNA Delivery
`in Vivo with Fractional Factorial and Definitive Screening Designs
`‡
`†,‡
`‡,§
`∥
`∥
`Kevin J. Kauffman,
`J. Robert Dorkin,
`Jung H. Yang,
`Michael W. Heartlein,
`Frank DeRosa,
`‡
`‡,⊥
`and Daniel G. Anderson*,†,‡,#,∇
`Faryal F. Mir,
`Owen S. Fenton,
`†
`‡
`David H. Koch Institute for Integrative Cancer Research, §Department of Biology,
`Department of Chemical Engineering,
`⊥
`∇
`#
`Department of Chemistry,
`Institute for Medical Engineering and Science, and
`Harvard MIT Division of Health Science and
`Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
`∥
`Shire Pharmaceuticals, Lexington, Massachusetts 02421, United States
`*S Supporting Information
`
`ABSTRACT: Intracellular delivery of messenger RNA (mRNA) has
`the potential to induce protein production for many therapeutic
`applications. Although lipid nanoparticles have shown considerable
`promise for the delivery of small
`interfering RNAs (siRNA), their
`utility as agents
`for mRNA delivery has only recently been
`investigated. The most common siRNA formulations contain four
`components: an amine-containing lipid or
`lipid-like material,
`phospholipid, cholesterol, and lipid-anchored polyethylene glycol,
`the relative ratios of which can have profound effects on the
`formulation potency. Here, we develop a generalized strategy to
`optimize lipid nanoparticle formulations for mRNA delivery to the
`liver in vivo using Design of Experiment (DOE) methodologies including Definitive Screening and Fractional Factorial Designs.
`By simultaneously varying lipid ratios and structures, we developed an optimized formulation which increased the potency of
`erythropoietin-mRNA-loaded C12-200 lipid nanoparticles 7-fold relative to formulations previously used for siRNA delivery. Key
`features of this optimized formulation were the incorporation of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and
`increased ionizable lipid:mRNA weight ratios. Interestingly, the optimized lipid nanoparticle formulation did not improve siRNA
`delivery, indicating differences in optimized formulation parameter design spaces for siRNA and mRNA. We believe the general
`method described here can accelerate in vivo screening and optimization of nanoparticle formulations with large
`multidimensional design spaces.
`KEYWORDS: Lipid nanoparticle, mRNA, design of experiment, nucleic acid, in vivo
`
`N ucleic acids have tremendous therapeutic potential to
`
`modulate protein expression in vivo but must be
`delivered safely and effectively. Because the delivery of naked
`nucleic acids results in poor cellular internalization, rapid
`degradation, and fast renal clearance,1,2 lipid nanoparticles
`(LNPs) have been developed to encapsulate and deliver nucleic
`acids to the liver. Most notably, the field has seen orders-of-
`in the delivery of 21−23
`magnitude potency advances
`nucleotide-long double stranded small
`interfering RNAs
`(siRNAs) due in part to the creation of new synthetic ionizable
`lipids and lipid-like materials.2 Whereas some of these novel
`lipids were synthesized with rational design approaches by
`systematically varying the lipid head and tail structures (e.g.,
`DLin-KC2-DMA, DLin-MC3-DMA, L319),3−5 other materials
`were discovered by creating large combinatorial
`libraries of
`lipid-like materials (e.g., C12-200, cKK-E12, 503O13).6−8
`When formulated into LNPs, these amine-containing ionizable
`lipids and lipid-like materials electrostatically complex with the
`negatively charged siRNA and can both facilitate cellular uptake
`and endosomal escape of the siRNA to the cytoplasm.6,9 In
`particular, the ionizable lipid-like material C12-200 has been
`
`widely used to make siRNA-LNP formulations for various
`therapeutic applications in vivo to silence protein expres-
`sion.10−12
`In addition to the ionizable material, three other excipients
`are also commonly used to formulate LNPs: (1) a
`phospholipid, which provides structure to the LNP bilayer
`and also may aid in endosomal escape;2,13 (2) cholesterol,
`which enhances LNP stability and promotes membrane
`fusion;14,15 and (3) lipid-anchored polyethylene glycol
`(PEG), which reduces LNP aggregation and “shields” the
`LNP from nonspecific endocytosis by immune cells.16 The
`particular composition of the LNP can also have profound
`effects on the potency of the formulation in vivo. Several
`previous efforts to study the effect of formulation parameters
`on siRNA-LNP potency utilized the one-variable-at-a-time
`method,17,18 in which formulation parameters were individually
`
`Received:
`June 23, 2015
`Revised: October 12, 2015
`
`© XXXX American Chemical Society
`
`A
`
`DOI: 10.1021/acs.nanolett.5b02497
`Nano Lett. XXXX, XXX, XXX−XXX
`
`Moderna Ex 1019-p. 1
`Moderna v Arbutus
`
`

`

`Nano Letters
`
`Letter
`
`Figure 1. Formulation of lipid nanoparticles. Lipid nanoparticles (LNPs) are synthesized by the mixing of two phases: (1) a four-component ethanol
`phase containing ionizable lipid, helper phospholipid, cholesterol, and lipid-anchored PEG; (2) an acidic aqueous phase containing mRNA.
`
`Table 1. Library A, B, and C Formulation Parameters
`
`parameter
`C12-200:mRNA weight ratio
`phospholipid
`
`original formulation
`5:1
`DSPC
`
`Library A
`Library B
`2.5:1 to 7.5:1
`7.5:1 to 12.5:1
`DSPC, DSPE
`DSPC
`DOPC, DOPE
`DOPE
`C12-200 molar composition
`50%
`40% to 60%
`30% to 40%
`35%
`phospholipid molar composition
`10%
`4% to 16%
`16% to 28%
`16%
`cholesterol molar composition
`38.5%
`21.5% to 55.5%
`28.5% to 51.5%
`46.5%
`PEG molar composition
`1.5%
`0.5% to 2.5%
`2.5% to 3.5%
`2.5%
`aPhospholipid abbreviations: DS = 1,2-distearoyl-sn-glycero- (saturated tail), DO = 1,2-dioleoyl-sn-glycero- (Δ9-cis unsaturated tail), PC = 3-
`phosphocholine (primary amine headgroup), PE = 3-phosphoethanolamine (quaternary amine headgroup).
`
`Library C
`5:1 to 25:1
`DOPE
`
`varied to maximize LNP potency; this approach, however, does
`not allow for examination of potentially important second-
`order interactions between parameters. Inspired by statistical
`methodologies commonly used in the engineering and
`combinatorial chemistry literature,19,20 we chose to utilize
`Design of Experiment (DOE) to better optimize LNP
`formulations for nucleic acid delivery. Using DOE, the number
`of
`individual experiments required to establish statistically
`significant trends in a large multidimensional design space are
`considerably reduced, which is particularly relevant for the
`economical screening of LNP formulations: in vitro screens are
`often poor predictors of in vivo efficacy with siRNA-LNPs,21
`and it would be both cost- and material-prohibitive to test large
`libraries of LNP formulations in vivo.
`To demonstrate the application of DOE to LNP formulation
`optimization in vivo, we formulated LNPs with a different type
`of nucleic acid than siRNA. Recently, messenger mRNA
`(mRNA) has been investigated for
`therapeutic protein
`production in vivo, including applications in cancer immuno-
`therapy, infectious disease vaccines, and protein replacement
`therapy.22,23 Unlike plasmid DNA, mRNA need only access the
`cytoplasm rather than the nucleus to enable protein translation
`and has no risk of inducing mutation through integration into
`the genome.24 Because there are inherent chemical and
`structural differences between mRNA and siRNA in terms of
`length, stability, and charge density of the nucleic acid,25 we
`hypothesized that LNP delivery formulations for mRNA may
`require significant variation from those developed for siRNA
`delivery. We further hypothesized that formulated mRNA may
`
`pack differently and with different affinity into nanoparticles
`than siRNA. To optimize LNP formulation parameters
`specifically for mRNA delivery, we developed a novel strategy
`in which we used DOE methodologiesincluding both
`Fractional Factorial and Definitive Screening Designsto
`synthesize several smaller LNP libraries to screen in vivo.
`Using the formulation conditions of the original siRNA-LNPs
`as a starting point, each successive generation of library was
`designed to improve protein expression based upon the
`parameters in the previous library that were found to correlate
`with improved efficacy. Through this approach, we aimed to
`develop an optimized C12-200 LNP with increased protein
`expression over the original LNP formulation.
`EPO mRNA Delivery with Original siRNA-Optimized
`LNP. The formulation process for synthesizing LNPs is
`described in Figure 1. The organic phase containing the lipids
`was mixed together with the acidic aqueous phase containing
`the nucleic acid in a microfluidic channel,26 resulting in the
`formation of mRNA-loaded LNPs. We chose to use unmodified
`mRNA coding for erythropoietin (EPO), a secreted serum
`protein that has previously been successfully translated in
`vivo.25,27 It has further been recently reported28 that LNP-
`delivered unmodified EPO mRNA is more potent than EPO
`mRNA with pseudouridine and/or 5-methylcytidine modifica-
`tions in vitro and in mice. To establish a baseline from which to
`improve, EPO mRNA was first formulated into LNPs using the
`formulation parameters previously published6 for
`original
`siRNA delivery in vivo (Table 1). The formulation was dosed
`intravenously at 15 μg of total mRNA per mouse and resulted
`
`B
`
`DOI: 10.1021/acs.nanolett.5b02497
`Nano Lett. XXXX, XXX, XXX−XXX
`
`Moderna Ex 1019-p. 2
`Moderna v Arbutus
`
`

`

`Nano Letters
`
`Letter
`
`Figure 2. Efficacy results of LNPs in Libraries A, B, and C. (a) Serum EPO concentration 6 h post-intravenous injection of 15 μg total mRNA for
`each formulation in Libraries A and B, including the original formulation (data presented as mean + SD, n = 3). (b) A statistically significant trend of
`increasing serum EPO concentration was observed with increasing C12-200:mRNA weight ratio and with DOPE phospholipid for Library B
`formulations, independent of the other formulation parameters. Furthermore, a statistically significant second-order effect was observed between
`DOPE and increasing weight ratio, as indicated by the larger relative slope of the DOPE best-fit line compared to the DSPC best-fit line. (1 data
`point = 1 mouse) (c) Serum EPO concentration 6 h post-intravenous injection of 15 μg total mRNA for formulation B-26 and Library C, which had
`similar formulation parameters as B-26 with differing C12-200:mRNA weight ratios. (Data presented as mean + SD, n = 3.)
`
`in an average EPO serum level of 963 ± 141 ng/mL at 6 h
`post-injection.
`Optimization of mRNA LNPs with Design of Experi-
`ment. Some previous efforts
`to optimize nanoparticle
`formulations have involved varying each of
`the important
`parameters individually and then possibly combining each
`optimized parameter
`for an overall optimized formula-
`tion.17,18,29 Because pilot experiments
`suggested strong
`second-order effects between parameters in our system, we
`chose instead to vary all five independent parameters
`simultaneously. In an attempt to maximize EPO expression in
`mice and thereby optimize the C12-200 LNPs for mRNA
`delivery, we chose to simultaneously vary the C12-200:mRNA
`weight
`ratio,
`the phospholipid identity, and the molar
`composition of the four-component LNP formulation. Three
`additional phospholipids structurally similar to DSPC but with
`differing head groups (primary vs quaternary amine) and tail
`saturation (saturated vs Δ9-cis unsaturated) were incorporated
`into the LNP formulations.
`Library A: Definitive Screening Design. We designed the
`first library, Library A, to be centered around the original
`siRNA-optimized LNP formulation parameters (Table 1). With
`four three-level quantitative factors (C12-200:mRNA weight
`ratio and three independent formulation molar compositions)
`and one four-level qualitative factor (phospholipid type), this
`
`large five-dimensional design space required DOE to reduce the
`number of formulations (3 × 3 × 3 × 3 × 4 = 324) to a
`reasonable number for in vivo experiments. An initial library of
`14 formulations (coded A-01 through A-14, see Table S1 for
`parameters) was created using a Definitive Screening Design, a
`recently described economical DOE in which main effects are
`not confounded with two-factor interactions and nonlinear
`correlations can be detected.30 The purpose of this first screen
`was to sample the large design space in a controlled fashion to
`eliminate unimportant formulation parameters and/or find a
`local maximum in efficacy from which a second-generation
`library could be generated.
`Out of 14 formulations in Library A, two formulations (A-02
`and A-09) resulted in higher EPO serum levels (6445 ± 1237
`and 2072 ± 302 ng/mL,
`respectively) than the original
`formulation (Figure 2a). Although the results from Library A
`were insufficient to deduce statistically significant effects for
`EPO production in vivo, there were statistically significant (p <
`0.05) orthogonal trends (Figure S2). We hypothesize that the
`increased encapsulation efficiency with increasing C12-
`200:mRNA weight ratio (Figure S2a) is caused by better
`complexation of more positively charged ionized C12-200 lipid
`with negatively charged mRNA. We also observed decreased
`LNP size with increasing PEG composition (Figure S2b), a
`phenomenon that has been previously observed in the
`
`C
`
`DOI: 10.1021/acs.nanolett.5b02497
`Nano Lett. XXXX, XXX, XXX−XXX
`
`Moderna Ex 1019-p. 3
`Moderna v Arbutus
`
`

`

`Nano Letters
`
`literature18,31 and has been speculated to be caused by
`increased lipid bilayer compressibility and increased repulsive
`forces between liposomes.32 The two top-performing for-
`mulations of Library A (A-02 and A-09) possessed similar
`attributes:
`increased weight ratio (7.5:1 vs 5:1),
`increased
`phospholipid content (16% vs 10%), and either DSPC or
`DOPE as the phospholipid; moreover, A-02 had decreased
`C12-200 content (40% vs 50%) and A-09 had increased PEG
`content (2.5% vs 1.5%).
`Library B: Fractional Factorial Screening Design. A more
`robust second-generation library, Library B (coded B-15 to B-
`32, Table S1), was generated using a L18-Taguchi Fractional
`Factorial Design29 with new parameter ranges which shifted in
`the direction of the two top-performing LNPs from the first
`library (Table 1). Out of 18 formulations in Library B, 11
`formulations resulted in higher EPO serum levels than the
`original
`formulation (Figure 2a). The top-performing for-
`mulation was B-26 with an average serum EPO concentration
`of 7485 ± 854 ng/mL. A standard least squares linear
`regression model was applied to the data from Library B, and
`several statistically significant factors were found with respect to
`efficacy (Table S2). Several second-order effects were found to
`be statistically significant as well, including the second-order
`interaction between DOPE and C12-200:mRNA weight ratio as
`shown by the best-fit line (p < 0.05) for DOPE in Figure 2b.
`Additional description of the statistical model and significant
`effects may be found in the Supporting Information (Table S2,
`Figure S1).
`that
`trend from Library B was
`The most apparent
`formulations with DOPE as the phospholipid resulted in
`significantly higher EPO production than formulations with
`DSPC, the original phospholipid (Figure 2b). In fact, the
`presence of DOPE in the formulation was the single strongest
`predictor of
`in vivo efficacy in our study. Whereas DSPC
`contains a quaternary amine headgroup and a fully saturated
`tail, DOPE contains a primary amine headgroup and a tail with
`one degree of unsaturation. It has been reported that conical
`lipids, such as DOPE, tend to adopt the less stable hexagonal
`phase, while cylindrical lipids, such as DSPC, tend to adopt the
`more stable lamellar phase.33 Upon fusion with the endosomal
`membrane, LNPs containing DOPE may reduce membrane
`stability, ultimately promoting endosomal escape.34,35 Another
`possible explanation involves their different encapsulation
`efficiencies: independent of other varying formulation param-
`eters, formulations with DSPC entrapped mRNA on average
`significantly better than DOPE (51% vs 36%), so it may be
`possible that the stronger complexation of mRNA to lipid in
`DSPC LNPs hinders the subsequent decomplexation of mRNA
`from lipid once inside the cell, thus inhibiting translation of the
`mRNA to protein.
`Library C: Maximizing Lipid:mRNA Weight Ratio with
`DOPE. As was initially hypothesized, we observed several
`second-order effects on EPO production between formulation
`parameters in Library B, most notably the synergistic effect
`between increasing the C12-200:mRNA weight ratio along with
`the use of DOPE as the phospholipid (Figure 2b). In an effort
`to further increase in vivo potency, a third and final library was
`generated (Library C, Table 1) to exploit
`this discovered
`second-order effect. The top-performing formulation (B-26)
`from Library B was reformulated with C12-200:mRNA weight
`ratios varying from 5:1 to 25:1 (coded C33−C38, Table S1).
`Surprisingly,
`increasing the weight ratio only increased the
`serum EPO concentration up to a certain point (Figure 2c); it
`
`Letter
`
`appears that increasing the weight ratio beyond 10:1 confers no
`significant efficacy advantage in vivo. Because no significant
`increases in EPO production were observed beyond 10:1 and
`to mitigate any concerns with possible lipid toxicity caused by
`increased lipid doses, we chose the 10:1 C12-200:mRNA
`weight
`ratio (C-35) as the final mRNA-optimized LNP
`formulation (Table 2).
`
`Table 2. LNP Characteristics of C-35 Compared to the
`Original Formulationa
`
`optimized formulation
`(C-35)
`10:1
`DOPE
`35%
`16%
`
`46.5%
`2.5%
`
`7065 ± 513
`102
`0.158
`43
`
`original
`formulation
`5:1
`DSPC
`50%
`10%
`
`38.5%
`1.5%
`
`962 ± 141
`152
`0.102
`24
`
`C12-200:mRNA weight ratio
`phospholipid
`C12-200 molar composition
`phospholipid molar
`composition
`cholesterol molar composition
`C14 PEG 2000 molar
`composition
`serum EPO (ng/μL)
`diameter (nm)
`polydispersity index (PDI)
`mRNA encapsulation efficiency
`(%)
`6.96
`7.25
`pKa
`−5.0
`−25.4
`zeta potential (mV)
`aPhospholipid abbreviations: DSPC = 1,2-distearoyl-sn-glycero-3-
`phosphocholine, DOPE = 1,2-dioleoyl-sn-glycero-3-phosphoethanol-
`amine, Serum EPO reported as mean ± SD (n = 3) 6 h after 15 μg of
`total mRNA intravenous injection into mice.
`
`Evaluation of Methodology. Although only 14% (2 of 14)
`of the Library A formulations resulted in increased potency
`compared to the original parameters, 61% (11 of 18) of the
`Library B formulations and 100% of Library C formulations (6
`of 6) did so (Figures 2a,c). This suggests that formulation
`parameters can be optimized and are critically important for
`efficient mRNA delivery with C12-200 LNPs. Furthermore, the
`increasing percentage of formulations that performed better
`than the original in each subsequent library demonstrates the
`predictive success of the generated statistical models (Table
`S2). A flowchart of the complete methodology we developed
`for in vivo nanoparticle optimization can be found in Figure S3.
`Characterization of mRNA-Optimized LNP. The opti-
`mized formulation C-35 had the following formulation
`parameters: 10:1 C12-200:mRNA weight
`ratio with 35%
`C12-200, 16% DOPE, 46.5% cholesterol, and 2.5% C14-
`PEG2000 molar composition. The average efficacy of C-35
`with 15 μg of total EPO mRNA injection in vivo, 7065 ± 513
`ng/mL, was increased over 7-fold compared to the original
`traditional LNP formulation (963 ± 141 ng/mL). C-35 was
`further characterized and compared to the original formulation
`with regard to size, polydispersity, encapsulation efficiency, and
`pKa (Table 2). No significant morphological differences were
`observed between the two formulations with transmission
`electron microscopy (TEM) (Figure S4). Although others have
`reported increases
`in siRNA nanoparticle potency with
`decreasing size,36 we found no such trend with all 38 mRNA
`formulations tested in our LNP system. Jayaraman et al.4 found
`that pKa was an important characteristic in predicting the
`efficacy of liver-targeting siRNA LNPs with an optimal pKa of
`between 6.2 and 6.5. It appears that in our C12-200 mRNA
`
`D
`
`DOI: 10.1021/acs.nanolett.5b02497
`Nano Lett. XXXX, XXX, XXX−XXX
`
`Moderna Ex 1019-p. 4
`Moderna v Arbutus
`
`

`

`Nano Letters
`
`Letter
`
`Figure 3. Efficacy and biodistribution of original and C-35 formulation with Luc mRNA. (a) Efficacy of original and C-35 LNP formulations
`synthesized with mRNA coding for luciferase in three organs of interest as measured by total flux from luminescence 6 h after intravenous injection
`of 15 μg total mRNA. (Data presented as mean + SD, n = 3). (b) Representative biodistribution image of luciferase expression for original and C-35
`LNP in seven organs as measured with an IVIS imaging system 6 h after intravenous injection of 15 μg of total mRNA.
`
`system, the in vivo efficacy is not significantly correlated with
`pKa of the LNP, although the slightly lower pKa of C-35 (pKa =
`6.96) compared to the original formulation (pKa = 7.25) may
`partially explain its improved efficacy. The surface charge of the
`LNP may also partially explain differences in efficacy: the
`optimized formulation C-35 is less negatively charged (zeta
`potential = −5.0 mV) than the original formulation (−25.4
`mV). C-35 contains twice the amount of amine-rich ionizable
`lipid C12-200 than the original formulation, which is likely the
`predominant reason C-35 is more positively charged. Although
`one study found no relationship between surface charge and
`hepatocellular delivery in vivo with siRNA-loaded lipid
`nanoparticles,21 other reports have noted that more positively
`charged nanoparticles bind better to negatively charged cellular
`membranes and this electrostatic interaction might facilitate
`uptake.37
`In order to determine whether C-35 would similarly improve
`the efficacy of mRNAs with different lengths, we formulated
`LNPs with firefly luciferase (Luc) mRNA, an mRNA which has
`a coding region roughly three times longer than that of EPO
`mRNA (1653 vs 582 nucleotides). Luciferase protein generated
`by C-35 LNPs was expressed predominately in the liver and
`likewise resulted in a statistically significant, approximately 3-
`fold increase in luciferase expression as measured by liver
`luminescence compared to the original formulation (Figure 3).
`Although LNPs made with Luc mRNA had similar
`encapsulation efficiencies as those made with shorter EPO
`mRNA (Tables 1, S3), we anticipate that significantly longer
`mRNAs would eventually become too large to effectively load
`into LNPs.
`siRNA Delivery with mRNA-Optimized LNP. Having
`optimized the formulation for mRNA delivery, we then wanted
`to examine the potential
`for siRNA delivery with C-35 as
`compared to the original siRNA-optimized formulation. We
`formulated siRNA coding for Factor VII (FVII), a serum
`clotting factor expressed exclusively in hepatocytes, using both
`the C-35 LNP and the original LNP formulation to determine
`their
`relative silencing in hepatocytes. FVII
`levels were
`measured 72 h after intravenous injection of siRNA-loaded
`LNPs ranging from 0.01 mg/kg to 0.1 mg/kg, and there was no
`significant difference between the original and optimized
`formulations at any dose (Figure 4, Table S4) despite having
`significantly different formulation parameters. The ED50 of both
`C-35 and the original
`formulations with FVII siRNA were
`
`Figure 4. Efficacy of original and C-35 formulation with siRNA.
`Efficacy of original versus optimized C-35 formulation made with C12-
`200 and siRNA coding against Factor VII (FVII) protein as measured
`by serum FVII levels 72 h post-intravenous injection of various doses
`of total siRNA. FVII levels were normalized with respect to PBS-
`injected control mice. (Data presented as mean + SD, n = 3.)
`
`approximately 0.03 mg/kg of total siRNA content, consistent
`with previous reports.6
`Interestingly, siRNA-loaded LNPs may be more tolerant than
`mRNA-loaded LNPs of design space differences. Over the past
`decade in the siRNA delivery field, many groups have focused
`on developing new ionizable lipids to increase the potency of
`siRNA-LNPs but have generally used the same standard
`formulation parameters in consecutive studies.3,4,6−8 The
`discovery of new ionizable lipids and lipid-like materials,
`however,
`is an endeavor which is often time- and material-
`intensive,
`requiring large-scale combinatorial
`libraries or
`chemically difficult rational design approaches. Meanwhile, we
`have shown that for one of the most commonly used ionizable
`materials for siRNA delivery, C12-200, merely changing the
`formulation parameters can significantly increase the potency of
`the LNP when loaded with two different mRNAs of varying
`lengths, EPO or Luc (Table 2, Figure 3).
`In this study, we have demonstrated a new general method
`for optimizing previously used siRNA lipid nanoparticle
`technology for a new class of RNA therapeutics and identified
`a lead optimized formulation for mRNA delivery, coded C-35.
`To the best of our knowledge, this study represents the first
`optimization of nanoparticle potency in vivo using Design of
`Experiment principles. Although C-35 significantly improved
`
`E
`
`DOI: 10.1021/acs.nanolett.5b02497
`Nano Lett. XXXX, XXX, XXX−XXX
`
`Moderna Ex 1019-p. 5
`Moderna v Arbutus
`
`

`

`Nano Letters
`
`mRNA delivery with mRNA’s of two different lengths, C-35
`was surprisingly equally as efficacious for siRNA delivery as the
`original siRNA-optimized formulation. We believe that the
`optimized formulations described here may provide a basis for
`further formulation optimization with other mRNA delivery
`materials as well. Furthermore, the generalized approach we
`described for in vivo optimization of multicomponent nano-
`particle formulations may accelerate the discovery of more
`potent formulations with other materials and drug payloads.
`Methods. Lipid Nanoparticle Synthesis. The ethanol phase
`was prepared by solubilizing with ethanol a mixture of C12-200
`(prepared as previously described,6 courtesy of Alnylam
`Pharmaceutics, Cambridge, MA), 1,2-distearoyl-sn-glycero-3-
`phosphocholine (DSPC, Avanti Polar Lipids, Alabaster, AL),
`1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE,
`Avanti), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC,
`Avanti), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine
`(DOPE, Avanti), cholesterol (Sigma), and/or 1,2-dimyristoyl-
`sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene
`glycol)-2000] (ammonium salt) (C14-PEG 2000, Avanti) at
`predetermined molar ratios. The aqueous phase was prepared
`in 10 mM citrate buffer (pH 3) with either EPO mRNA
`(human erythropoietin mRNA, courtesy of Shire Pharmaceut-
`icals, Lexington, MA), Luc mRNA (Firefly luciferase mRNA,
`Shire), or FVII siRNA (Factor VII siRNA,7 Alnylam). Syringe
`pumps were used to mix the ethanol and aqueous phases at a
`3:1 ratio in a microfluidic chip device.26 The resulting LNPs
`were dialyzed against PBS in a 20 000 MWCO cassette at 4 °C
`for 2 h.
`mRNA Synthesis. mRNA was synthesized by in vitro
`transcription from a plasmid DNA template encoding the
`gene, which was followed by the addition of a 5′ cap structure
`(Cap 1) using a vaccinia virus-based guanylyl
`transferase
`system. A poly(A) tail of approximately 300 nucleotides was
`incorporated via enzymatic addition employing poly-A
`polymerase. Fixed 5′ and 3′ untranslated regions were
`constructed to flank the coding sequences of the mRNA.
`LNP Characterization. To calculate the nucleic acid
`encapsulation efficiency, a modified Quant-iT RiboGreen
`RNA assay (Invitrogen) was used as previously described.38
`The size and polydispersity (PDI) of the LNPs were measured
`using dynamic light scattering (ZetaPALS, Brookhaven Instru-
`ments). Zeta potential was measured using the same instrument
`in a 0.1× PBS solution. Size data is reported as the largest
`intensity mean peak average, which constituted >95% of the
`nanoparticles present in the sample. The pKa was determined
`using a TNS assay as previously described.38 To prepare LNPs
`for Transmission Electron Microscopy (TEM), LNPs were
`dialyzed against water and negative staining was performed with
`2% uranyl acetate. LNPs were then imaged with a Tecnai Spirit
`transmission electron microscope (FEI, Hillsboro, OR).
`Animal Experiments. All animal studies were approved by
`the M.I.T. Institutional Animal Care and Use Committee and
`were consistent with local, state, and federal regulations as
`applicable. Female C57BL/6 mice (Charles River Laboratories,
`18−22 g) were intravenously injected with LNPs via the tail
`vein. After 6 or 72 h, blood was collected via the tail vein with
`serum separation tubes, and the serum was isolated by
`centrifugation. Serum EPO levels were measured using an
`ELISA assay (Human Erythropoietin Quantikine IVD ELISA
`Kit, R&D Systems, Minneapolis, MD). Serum FVII levels were
`measured using a chromogenic assay (Biophen FVII, Aniara
`Corporation, West Chester, OH) and compared with a
`
`Letter
`
`standard curve obtained from control mice. Six hours after
`administration of Luc mRNA LNPs, mice were administered an
`intraperitoneal injection of 130 μL of D-luciferin (30 mg/mL in
`PBS). After 15 min, the mice were sacrificed, and eight organs
`were collected (liver, spleen, pancreas, kidneys, uterus, ovaries,
`lungs, heart). The organs’ luminescence were analyzed using an
`IVIS imaging system (PerkinElmer, Waltham, MA) and
`quantified using LivingImage software (PerkinElmer) to
`measure the radiance of each organ in photons/sec.
`Statistics. Design of Experiment (DOE) was performed, and
`statistical data were analyzed using JMP software (SAS, Cary,
`N.C.). In this study, statistical significance was defined as p-
`values less than 0.05. Three mice per formulation/dose (n = 3)
`were used for all in vivo experiments. For Library A, a 34 × 22
`Definitive Screening Design30 was used with 4 three-level
`quantitative factors (C12-200 RNA weight ratio, C12-200 mol
`%, phospholipid mol %, and PEG mol %) and 2 two-level
`qualitative factors for phospholipid tail group (DS = 1,2-
`distearoyl-sn-glycero- and DO = 1,2-dioleoyl-sn-glycero-) and
`phospholipid headgroup (PC = 3-phosphocholine and PE = 3-
`phosphoethanolamine). For Library B, a 34 × 21 L-18 Taguchi
`Fractional Factorial Design29 was used with 4 three-level
`quantitative factors (C12-200 RNA weight ratio, C12-200 mol
`%, phospholipid mol %, and PEG mol %) and 1 two-level
`qualitative factor for phospholipid (DSPC or DOPE). To make
`the Standard Least Squares regression model for Library B, a
`full model with all orthogonal and second-order effects was
`generated and subsequently reduced until only statistically
`significant effects remained in the model as determined by
`ANOVA. A posthoc Tukey test was performed using JMP to
`verify that the two levels of phospholipid effect were statistically
`different (p < 0.0001). When comparing means between two
`groups, a Student’s t
`test was used assuming a Gaussian
`distribution and unequal variances. Further details about
`statistics and models used in this study,
`including ANOVA
`results, parameter estimates, residuals, etc., can be found in
`Table S2, Figure S1, and the Supplementary Methods section.
`
`■ ASSOCIATED CONTENT
`*S Supporting Information
`The Supporting Information is available free of charge on the
`ACS Publications website at DOI: 10.1021/acs.nano-
`lett.5b02497.
`Description of Library B statistical model, nanoparticle
`characterization for all LNP formulations (including
`formulation composition, encapsulation efficiency, size,
`polydispersity, and efficacy measurements), additional
`structure/function relationships for Library A, and a
`detailed description of the statistical methodologies used
`including a flowchart (PDF)
`
`■ AUTHOR INFORMATION
`
`Corresponding Author
`*E-mail (D.G.A.) dgander@mit.edu. 500 Main Street, David.
`H. Koch Institute for Integrative Cancer Research, Massachu-
`setts Institute of Technology, Cambridge, MA 02139.
`Author Contributions
`K.J.K. and J.R.D. contributed equally to this work.
`Notes
`The authors declare no competing financial interest.
`
`F
`
`DOI: 10.1021/acs.nanolett.5b02497
`Nano Lett. XXXX, XXX, XXX−XXX
`
`Moderna Ex 1019-p. 6
`Moderna v Arbutus
`
`

`

`Nano Letters
`
`■ ACKNOWLEDGMENTS
`
`This work was supported by Shire Pharmaceuticals (Lexington,
`MA) and the MIT Skoltech Initiative. The authors thank Nicki
`Watson at the W.M. Keck Microscopy Institute (Whitehead
`Institute, Cambridge, MA) for assistance in performing the
`TEM experiments. We also thank Prof. Sumona Mondal
`(Clarkson University, Potsdam, NY) for performing a statistical
`review of the manuscript.
`
`■ REFERENCES
`(1) Whitehead, K. A.; Langer, R.; Anderson, D. G. Nat. Rev. Drug
`Discovery 2009, 8 (2), 129−138.
`(2) Kanasty, R.; Dorkin, J. R.; Vegas, A.; Anderson, D. Nat. Mater.
`2013, 12 (11), 967−977.
`(3) Semple, S. C.; Akinc, A.; Chen, J.; Sandhu, A. P.; Mui, B. L.; Cho,
`C. K.; Sah, D. W. Y.; Stebbing, D.; Crosley, E. J.; Yaworski, E.; Hafez, I.
`M.; Dorkin, J. R.; Qin, J.; Lam, K.; Rajeev, K. G.; Wong, K. F.; Jeffs, L.
`B.; Nechev, L.; Eisenhardt, M. L.; Jayaraman, M.; Kazem, M.; Maier,
`M. a; Srinivasulu, M.; Weinstein, M. J.; Chen, Q.; Alvarez, R.; Barros,
`S. a; De, S.; Klimuk, S. K.; Borland, T.; Kosovrasti, V.; Cantley, W. L.;
`Tam, Y. K.; Manoharan,

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