throbber
l e t t e r s
`
`rational design of cationic lipids for sirNA delivery
`Sean C Semple1,6, Akin Akinc2,6, Jianxin Chen1,5, Ammen P Sandhu1, Barbara L Mui1,5, Connie K Cho1,
`Dinah W Y Sah2, Derrick Stebbing1, Erin J Crosley1, Ed Yaworski1, Ismail M Hafez3, J Robert Dorkin2, June Qin2,
`Kieu Lam1, Kallanthottathil G Rajeev2, Kim F Wong3, Lloyd B Jeffs1, Lubomir Nechev2, Merete L Eisenhardt1,
`Muthusamy Jayaraman2, Mikameh Kazem3, Martin A Maier2, Masuna Srinivasulu4, Michael J Weinstein2,
`Qingmin Chen2, Rene Alvarez2, Scott A Barros2, Soma De2, Sandra K Klimuk1, Todd Borland2,
`Verbena Kosovrasti2, William L Cantley2, Ying K Tam1,5, Muthiah Manoharan2, Marco A Ciufolini4,
`Mark A Tracy2, Antonin de Fougerolles2, Ian MacLachlan1, Pieter R Cullis3, Thomas D Madden1,5 & Michael J Hope1,5
`
`We adopted a rational approach to design cationic lipids for
`use in formulations to deliver small interfering RNA (siRNA).
`Starting with the ionizable cationic lipid 1,2-dilinoleyloxy-3-
`dimethylaminopropane (DLinDMA), a key lipid component of
`stable nucleic acid lipid particles (SNALP) as a benchmark,
`we used the proposed in vivo mechanism of action of ionizable
`cationic lipids to guide the design of DLinDMA-based lipids
`with superior delivery capacity. The best-performing lipid
`recovered after screening (DLin-KC2-DMA) was formulated
`and characterized in SNALP and demonstrated to have
`in vivo activity at siRNA doses as low as 0.01 mg/kg in rodents
`and 0.1 mg/kg in nonhuman primates. To our knowledge, this
`represents a substantial improvement over previous reports of
`in vivo endogenous hepatic gene silencing.
`
`A key challenge in realizing the full potential of RNA interference
`(RNAi) therapeutics is the efficient delivery of siRNA, the molecules
`that mediate RNAi. The physicochemical characteristics of siRNA—
`high molecular weight, anionic charge and hydrophilicity—prevent
`passive diffusion across the plasma membrane of most cell types.
`Therefore, delivery mechanisms are required that allow siRNA to
`enter cells, avoid endolysosomal compartmentalization and localize
`in the cytoplasm where it can be loaded into the RNA-induced
`
` silencing complex. To date, formulation in lipid nanoparticles (LNPs)
` represents one of the most widely used strategies for in vivo delivery
`of siRNA1,2. LNPs represent a class of particles comprised of different
`lipid compositions and ratios as well as different sizes and structures
`formed by different methods. A family of LNPs, SNALP3–6, is charac-
`terized by very high siRNA-encapsulation efficiency and small,
`uniformly sized particles, enabled by a controlled step-wise dilution
`methodology. LNPs, including SNALP, have been successfully used to
`silence therapeutically relevant genes in nonhuman primates6–8 and
`are currently being evaluated in several clinical trials.
`An empirical, combinatorial chemistry–based approach recently
`identified novel materials for use in LNP systems7. A key feature of
`this approach was the development of a one-step synthetic strategy
`that allowed the rapid generation of a diverse library of ~1,200 com-
`pounds. This library was then screened for novel materials capable
`of mediating efficient delivery of siRNA in vitro and in vivo. Here, we
`instead used a medicinal chemistry (that is, structure-activity relation-
`ship) approach, guided by the putative in vivo mechanism of action
`of ionizable cationic lipids, for rational lipid design. Specifically,
`we hypothesized that after endocytosis, the cationic lipid interacts
`with naturally occurring anionic phospholipids in the endosomal
`membrane, forming ion pairs that adopt nonbilayer structures and
`disrupt membranes (Fig. 1)9–12. We previously advanced the concept
`
`Figure 1 Proposed mechanism of action for membrane disruptive effects of
`cationic lipids and structural diagram of DLinDMA divided into headgroup,
`linker and hydrocarbon chain domains. In isolation, cationic lipids and
`endosomal membrane anionic lipids such as phosphatidylserine adopt a
`cylindrical molecular shape, which is compatible with packing in a bilayer
`configuration. However, when cationic and anionic lipids are mixed together,
`they combine to form ion pairs where the cross-sectional area of the combined
`headgroup is less than that of the sum of individual headgroup areas in
`isolation. The ion pair therefore adopts a molecular ‘cone’ shape, which
`promotes the formation of inverted, nonbilayer phases such as the hexagonal
`HII phase illustrated. Inverted phases do not support bilayer structure and are
`associated with membrane fusion and membrane disruption9,21.
`
`+
`
`–
`
`+ –
`
`Cylindrical shape supports
`bilayer structure
`
`Cone shape disrupts
`bilayer structure
`
`Headgroup
`
`Linker
`
`Hydrocarbon
`chains
`
`Bilayer
`
`Hexagonal HII
`
`DLinDMA
`
`1Tekmira Pharmaceuticals, Burnaby, British Columbia, Canada. 2Alnylam Pharmaceuticals, Cambridge, Massachusetts, USA. 3Department of Biochemistry and
`Molecular Biology and 4Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada. 5Present address: Alcana Technologies,
`Vancouver, British Columbia, Canada. 6These authors contributed equally to this work. Correspondence should be addressed to S.C.S. (ssemple@tekmirapharm.com)
`or A.A. (aakinc@alnylam.com).
`
`Received 16 September 2009; accepted 17 December 2009; published online 17 January 2010; doi:10.1038/nbt.1602
`
`172
`
`VOLUME 28 NUMBER 2 FEBRUARY 2010 nature biotechnology
`
`© 2010 Nature America, Inc. All rights reserved.
`
`ARBUTUS - EXHIBIT 2021
`Moderna Therapeutics, Inc. v. Arbutus Biopharma Corporation - IPR2019-00554
`
`

`

`l e t t e r s
`
`Figure 2 In vivo evaluation of novel cationic lipids. (a) Silencing
`activity of DLinDAP (), DLinDMA (), DLin-K-DMA () and DLin-
`KC2-DMA (•) screening formulations in the mouse Factor VII model.
`All LNP-siRNA systems were prepared using the preformed vesicle
`(PFV) method and were composed of ionizable cationic lipid, DSPC,
`cholesterol and PEG-lipid (40:10:40:10 mol/mol) with a Factor
`VII siRNA/total lipid ratio of ~0.05 (wt/wt). Data points are expressed
`as a percentage of PBS control animals and represent group mean
`(n = 5) ± s.d., and all formulations were compared within the same
`study. (b) Influence of headgroup extensions on the activity of
`DLin-K-DMA. DLin-K-DMA () had additional methylene groups added
`between the DMA headgroup and the ketal ring linker to generate
`DLin-KC2-DMA (•), DLin-KC3-DMA () and DLin-KC4-DMA (). The
`activity of PFV formulations of each lipid was assessed in the mouse
`Factor VII model. Data points are expressed as a percentage of PBS
`control animals and represent group mean (n = 4) ± s.d. (c) Chemical
`structures of novel cationic lipids.
`
`120
`100
`80
`60
`40
`20
`
`00
`
`b
`
`protein (%)
`
`Relative serum factor VII
`
`0.1
`.01
`1
`10
`Factor VII siRNA dose (mg/kg)
`
`120
`100
`80
`60
`40
`20
`
`00
`
`protein (%)
`
`a
`
`Relative serum factor VII
`
`0.1
`.01
`1
`10
`100
`Factor VII siRNA dose (mg/kg)
`
`O
`
`R
`
`Me2N
`
`O
`R
`
`O
`
`O
`DLinDAP
`
`RR
`
`O O
`
`Me2N
`
`R R
`
`O O
`
`Me2N
`
`R
`R
`
`O O
`
`Me2N
`
`O R
`R
`
`O
`
`DLinDMA
`
`Me2N
`
`R
`R
`
`O O
`
`c
`
`Me2N
`
`DLin-K-DMA
`
`DLin-KC2-DMA
`
`DLin-KC3-DMA
`
`DLin-KC4-DMA
`
`R =
`
`of using ionizable cationic lipids with pKas < pH 7.0 to efficiently
`formulate nucleic acids at low pH and maintaining a neutral or low
`cationic surface charge density at pH 7.4 (ref. 13). This strategy should
`provide better control of the circulation properties of these systems
`and reduce nonspecific disruption of plasma membranes. As posi-
`tive charge density is minimal in the blood but increases substan-
`tially in the acidic environment of the endosome, this should activate
`the membrane-destabilizing property of the LNP. Although these
`attributes may account for the activity of these systems upon inter-
`nalization by hepatocytes, they do not necessarily explain the high
`levels of hepatic biodistribution observed for many LNPs, including
`SNALP. Although these LNPs do not specifically include a targeting
`ligand to direct them to hepatocytes after systemic administration,
`it is possible that these LNPs associate with one or more proteins in
`plasma that may promote hepatocyte endocytosis.
`The ionizable cationic lipid DLinDMA has proven to be highly
`effective in SNALP, has been extensively tested in rodents and non-
`human primates, and is now being evaluated in human clinical
`trials. Therefore, we selected it as the starting point for the design
`and synthesis of novel lipids. We chose the mouse Factor VII model7,
`as the primary in vivo screening system to assess functional LNP-
`mediated delivery to hepatocytes. Briefly, C57BL/6 mice received
`a single dose of LNP-formulated Factor VII siRNA through bolus
`tail vein injection and serum was collected from animals 24 h after
`administration to analyze Factor VII protein level. The initial screen-
`ing of LNP-siRNA systems was conducted using LNPs prepared by
`a preformed vesicle method14 and composed of ionizable cationic
`lipid, distearoylphosphatidylcholine (DSPC), cholesterol and PEG-
`lipid (40:10:40:10 mol/mol), with a Factor VII siRNA/total lipid ratio
`of ~0.05 (wt/wt). Although not a bilayer-destabilizing lipid, a small
`amount of phosphatidylcholine was incorporated into the LNP to
`help stabilize the LNP both during formulation and while it was in
`circulation. A short acyl chain PEG-lipid was incorporated into the
`LNP to control particle size during formulation, but is designed to
`leave the LNP rapidly upon intravenous injection. As our goal was
`to identify novel ionizable cationic lipids for use in LNPs, we aimed
`to minimize other effects by using a single robust composition and
`set of formulation conditions suitable for all novel lipids tested. The
`preformed vesicle method employing the composition listed above
`provides a convenient platform for such testing, but uses a different
`formulation process, a different lipid composition and a different
`siRNA/lipid ratio than SNALP. The structure of DLinDMA can be
`divided into three main regions: the hydrocarbon chains, the linker
`and the headgroup (Fig. 1). A detailed structure-function study to
`
`investigate the impact of increasing the number of cis double bonds
`in the hydrocarbon chains found the linoleyl lipid containing two
`double bonds per hydrocarbon chain (DLinDMA) to be optimal15.
`We therefore maintained the linoleyl hydrocarbon chains present in
`DLinDMA as an element in our lipid design, and focused on optimiz-
`ing the linker and headgroup moieties.
`The linker region in a bilayer structure resides at the membrane
`interface, an area of transition between the hydrophobic membrane
`core and hydrophilic headgroup surface. Our approach to linker
`modification of DLinDMA involved introducing groups expected to
`exhibit different rates of chemical or enzymatic stability and to span
`a range of hydrophilicity. A variety of these rationally designed lipids
`were made, characterized and tested (Supplementary Syntheses 1 and
`Supplementary Table 1). LNPs based on the ester-containing lipid
`DLinDAP showed substantially reduced in vivo activity compared
`to LNPs based on the alkoxy-containing lipid DLinDMA (Fig. 2).
`Further, LNPs based on DLin-2-DMAP, a lipid with one alkoxy link-
`age and one ester linkage, yielded activity intermediate between
`DLinDAP- and DLinDMA-based LNPs (Supplementary Table 1).
`Although it is uncertain why the ester-containing lipids are consid-
`erably less active in vivo, we speculate that the diester lipid (DLinDAP)
`is relatively inactive because it is more readily hydrolyzed in vivo
`than the alkoxy analog (DLinDMA), and therefore, unable to either
`protect the siRNA adequately before release from the endosome
`and/or survive long enough in the endosome to disrupt the mem-
`brane. These hypotheses are being investigated. LNPs based on lipids
` containing carbamate or thioether linkages also resulted in dramati-
`cally reduced in vivo activity. Interestingly, the introduction of a ketal
`ring linker into DLinDMA resulted in LNPs that were ~2.5-fold more
`potent in reducing serum Factor VII protein levels relative to the
`DLinDMA benchmark, with an ED50 (that is, dose to achieve 50%
`gene silencing) of ~0.4 mg/kg versus 1 mg/kg, respectively (Fig. 2).
`Given the importance of positive charge in the mechanism-
`of-action hypothesis guiding the lipid design, the effects of structural
`changes in the amine-based headgroup were investigated in the con-
`text of DLin-K-DMA as the new benchmark lipid. A series of head-
`group modifications were made, characterized and tested to explore
`the effects of size, acid-dissociation constant and number of ionizable
`groups (Supplementary Syntheses 2 and Supplementary Table 2).
`Piperazino, morpholino, trimethylamino or bis-dimethylamino modifi-
`cations tested were not better than the benchmark dimethylamino
`headgroup of DLin-K-DMA. As an additional parameter, the distance
`between the dimethylamino group and the dioxolane linker was varied
`by introducing additional methylene groups. This parameter can
`
`nature biotechnology VOLUME 28 NUMBER 2 FEBRUARY 2010
`
`173
`
`© 2010 Nature America, Inc. All rights reserved.
`
`

`

`l e t t e r s
`
`Cationic lipid
`
`In vivo ED50
`(mg/kg)
`
`Table 1 Biophysical parameters and in vivo activities of
`LNPs containing novel lipids
`LII to HII phase transition
`Apparent
`temperature (°C)b
`a
`lipid pKa
`6.8 ± 0.10
`~1
`27
`DLinDMA
`6.2 ± 0.05
`40–50
`26
`DLinDAP
`5.9 ± 0.03
`~0.4
`19
`DLin-K-DMA
`6.7 ± 0.08
`~0.1
`20
`DLin-KC2-DMA
`7.2 ± 0.05
`~0.6
`18
`DLin-KC3-DMA
`7.3 ± 0.07
`>3
`18
`DLin-KC4-DMA
`apKa values ± s.d. (n = 3 to 9). bLII to HII phase transition was measured at pH 4.8 in equi-
`molar mixtures with DSPS, using differential scanning calorimetric, repeat scans reproducible
`to within 0.1 °C.
`
`affect both the pKa of the amine headgroup as well as the distance
`and flexibility of the charge presentation relative to the lipid bilayer
`interface. Inserting a single additional methylene group into the head-
`group (DLin-KC2-DMA) produced a dramatic increase in potency
`relative to DLin-K-DMA. The ED50 for this lipid was ~0.1 mg/kg,
`making it fourfold more potent than DLin-K-DMA and tenfold more
`potent than the DLinDMA benchmark when compared head-to-head
`in the Factor VII model (Fig. 2a). Further extension of the tether with
`additional methylene groups, however, substantially decreased activ-
`ity, with an ED50 of ~0.6 mg/kg for DLin-KC3-DMA and >3 mg/kg
`for DLin-KC4-DMA (Fig. 2b).
`As changes in lipid design and chemistry may affect the pharmaco-
`kinetics, target tissue accumulation and intracellular delivery of LNP
`formulations, we investigated the relative importance of these para-
`meters on LNP activity at an early stage in this research program.
`Several of the novel lipids were incorporated into LNP-siRNA for-
`mulations containing cyanine dye (Cy3)-labeled siRNA. Plasma,
`liver and spleen levels of siRNA were determined at 0.5 and 3 h after
`injection at siRNA doses of 5 mg/kg, and the results are presented in
`Supplementary Table 3. In general, formulations that were the most
`active in the mouse Factor VII screens achieved the highest liver levels
`of siRNA at 0.5 h; however, delivery of siRNA to the target tissue was
`not the primary factor responsible for activity. This is supported by
`the observations that most formulations accumulated in the liver and
`spleen quite quickly and that some formulations with similar liver lev-
`els of siRNA had large differences in activity. Moreover, plasma phar-
`macokinetics alone did not predict activity. For example, although
`DLin-KC2-DMA and DLinDMA had virtually indistinguishable
`blood pharmacokinetic profiles in mice (data not shown), the activity
`of DLin-KC2-DMA in LNPs is approximately tenfold greater than the
`
`same formulation with DLinDMA. Taken together, these results led
`us to conclude that rapid target tissue accumulation was important,
`but not sufficient, for activity. Moreover, other parameters were more
`critical for maximizing the activity of LNP-siRNA formulations.
`Two important parameters underlying lipid design for SNALP-
`mediated delivery are the pKa of the ionizable cationic lipid and the
`abilities of these lipids, when protonated, to induce a nonbilayer
`(hexagonal HII) phase structure when mixed with anionic lipids.
`The pKa of the ionizable cationic lipid determines the surface
`charge on the LNP under different pH conditions. The charge state
`at physiologic pH (e.g., in circulation) can influence plasma pro-
`tein adsorption, blood clearance and tissue distribution behavior16,
`whereas the charge state at acidic pH (e.g., in endosomes) can influ-
`ence the ability of the LNP to combine with endogenous anionic
`lipids to form endosomolytic nonbilayer structures9. Consequently,
`the ability of these lipids to induce HII phase structure in mixtures
`with anionic lipids is a measure of their bilayer-destabilizing capacity
`and relative endosomolytic potential.
`The fluorescent probe 2-(p-toluidino)-6-napthalene sulfonic
`acid (TNS), which exhibits increased fluorescence in a hydrophobic
`environment, can be used to assess surface charge on lipid bilayers.
`Titrations of surface charge as a function of pH can then be used
`to determine the apparent pKa of the lipid in the bilayer (hereafter
`referred to as pKa) of constituent lipids17. Using this approach, the
`pKa values for LNPs containing DLinDAP, DLinDMA, DLin-K-DMA,
`DLin-KC2-DMA, DLin-KC3-DMA and DLin-KC4-DMA were deter-
`mined (Table 1). The relative ability of the protonated form of the
`ionizable cationic lipids to induce HII phase structure in anionic lipids
`was ascertained by measuring the bilayer-to-hexagonal HII transition
`temperature (TBH) in equimolar mixtures with distearoylphosphati-
`dylserine (DSPS) at pH 4.8, using 31P NMR18 and differential scan-
`ning calorimetric analyses19. Both techniques gave similar results.
`The data presented in Table 1 indicate that the highly active lipid
`DLin-KC2-DMA has pKa and TBH values that are theoretically favo-
`rable for use in siRNA delivery systems. The pKa of 6.4 indicates
`that LNPs based on DLin-KC2-DMA have limited surface charge in
`circulation, but will become positively charged in endosomes. Further,
`the TBH for DLin-KC2-DMA is 7 °C lower than that for DLinDMA,
`suggesting that this lipid has improved capacity for destabilizing
` bilayers. However, the data also demonstrate that pKa and TBH do
`not fully account for the in vivo activity of lipids used in LNPs. For
` example, although DLin-KC3-DMA and DLin-KC4-DMA have
`identical pKa and TBH values, DLin-KC4-DMA requires a more than
`fivefold higher dose to achieve the same activity in vivo. Moreover,
`
`*
`
`*
`
`**
`
`1
`mg/kg
`siApoB
`
`0.03
`mg/kg
`
`PBS
`
`0.3
`0.1
`mg/kg
`mg/kg
`siTTR
`
`1
`mg/kg
`
`1.4
`
`1.2
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0
`
`b
`
`Relative liver TTR/GAPDH mRNA levels
`
`Figure 3 Efficacy of KC2-SNALP in rodents
`and nonhuman primates. (a) Improved
`efficacy of KC2-SNALP relative to the
`initial screening formulation tested in
`mice. The in vivo efficacy of KC2-SNALP
`() was compared to that of the unoptimized
`DLin-KC2-DMA screening (that is, PFV)
`formulation (•) in the mouse Factor VII model.
`Data points are expressed as a percentage
`of PBS control animals and represent group
`mean (n = 5) ± s.d. (b) Efficacy of KC2-SNALP
`in nonhuman primates. Cynomolgus monkeys
`(n = 3 per group) received a total dose of either
`0.03, 0.1, 0.3 or 1 mg/kg siTTR, or 1 mg/kg
`siApoB formulated in KC2-SNALP or PBS
`as 15-min intravenous infusions (5 ml/kg)
`through the cephalic vein. Animals were euthanized 48 h after administration. TTR mRNA levels relative to GAPDH mRNA levels were determined in
`liver samples. Data points represent group mean ± s.d. *, P < 0.05; **, P < 0.005.
`
`0.1
`0.01
`1
`Factor VII siRNA dose (mg/kg)
`
`10
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`00
`
`.001
`
`a
`
`Relative serum factor VII protein (%)
`
`174
`
`VOLUME 28 NUMBER 2 FEBRUARY 2010 nature biotechnology
`
`© 2010 Nature America, Inc. All rights reserved.
`
`

`

`Table 2 Clinical chemistry and hematology parameters for KC2-SNALP–treated rats
`siRNA dose
`Total Bilirubin
`BUN
`(mg/kg)a
`(mg/dl)
`(mg/dl)
`
`Vehicle
`
`ALT (U/L)
`
`AST (U/L)
`
`l e t t e r s
`
`RBC
`(× 106/µl)
`
`Hemoglobin
`(g/dl)
`
`WBC
`(× 103/µl)
`
`PLT (× 103/µl)
`
`1,166 ± 177
`11 ± 3
`11.3 ± 0.4
`5.5 ± 0.3
`4.8 ± 0.8
`2 ± 0
`109 ± 31
` 56 ± 16
`PBS
`1,000 ± 272
`13 ± 2
`11.6 ± 0.6
`5.6 ± 0.2
`4.4 ± 0.6
`2 ± 0
`100 ± 14
` 58 ± 22
`1
`KC2-SNALP
`1,271 ± 269
`13 ± 4
`11.6 ± 0.3
`5.9 ± 0.3
`4.3 ± 0.6
`2.2 ± 0.4
` 81 ± 10
`73 ± 9
`2
`KC2-SNALP
` 958 ± 241
`15 ± 2
`11.9 ± 0.4
`6.0 ± 0.2
`5.0 ± 0.8
`2 ± 0
`100 ± 30
` 87 ± 19
`3
`KC2-SNALP
`aNontargeting, luciferase siRNA. Sprague-Dawley rats (n = 5) received 15-min intravenous infusions of KC2-SNALP formulated siRNA at different dose levels. Blood samples were taken 24 h
`after administration. ALT, alanine aminotransferase; AST, aspartate aminotransferase; BUN, blood urea nitrogen; RBC, red blood cells; WBC, white blood cells; PLT, platelets.
`
`DLin-KC2-DMA and DLin-KC4-DMA, which have very similar pKa
`and TBH values, exhibit a >30-fold difference in in vivo activity. This
`result suggests that other parameters, such as the distance and flex-
`ibility of the charged group relative to the lipid bilayer interface, may
`also be important. Thus, although the biophysical parameters of pKa
`and TBH are useful for guiding lipid design, the results presented in
`Table 1 support the strategy of testing variants of lead lipids, even
`ones with very similar pKa and TBH values.
`The lipid composition chosen for the initial formulation and
`screening of novel ionizable cationic lipids (cationic lipid/DSPC/
`cholesterol/PEG-lipid = 40:10:40:10 mol/mol, siRNA/total lipid
`~ 0.05 wt/wt) was useful for determining the rank-order potency
`of novel lipids, but is not necessarily optimal for in vivo delivery. In
`addition, the in vivo activity of resultant LNP-siRNA formulations
`is affected by the formulation process employed and the resulting
`particle structure. Improvements in activity were possible with the
`preformed vesicle process by modifying and optimizing lipid ratios
`and formulation conditions (results not shown). However, we chose
`to further validate DLin-KC2-DMA activity specifically in the context
`of the SNALP platform, currently the most advanced LNP formula-
`tion for delivery of siRNA in vivo. We therefore tested in vivo a ver-
`sion of SNALP (termed KC2-SNALP), which uses less PEG lipid than
`reported previously6 and in which DLinDMA was replaced with DLin-
`KC2-DMA. The incorporation of DLin-KC2-DMA into SNALP led
`to a marked improvement in potency in the mouse Factor VII model;
`the measured ED50 decreased from ~0.1 mg/kg for the unoptimized
`screening formulation to ~0.02 mg/kg for the KC2-SNALP formu-
`lation (Fig. 3a). KC2-SNALP also exhibited similar potency in rats
`(data not shown). Furthermore, after a single administration in rats,
`KC2-SNALP–mediated gene silencing was found to persist for over
`10 d (Supplementary Fig. 1).
`In addition to efficacy, tolerability is another critical attribute of
`a suitable LNP-siRNA delivery system for human use. We therefore
`studied the single-dose tolerability of KC2-SNALP in rats—a popular
`rodent model for assessing the toxicology of siRNA and nucleic acid–
`based therapeutics. As doses near the efficacious dose level were found
`to be very well tolerated (data not shown), single-dose escalation
`studies were conducted starting at doses ~50-fold higher (1 mg/kg)
`than the observed ED50 of the formulation. To understand formula-
`tion toxicity in the absence of any toxicity or pharmacologic effects
`resulting from target silencing, we conducted the experiments using
`a nontargeting control siRNA sequence directed against luciferase.
`KC2-SNALP containing luciferase siRNA was prepared in the exact
`same manner as that containing Factor VII siRNA, and the resultant
`size, lipid composition and entrapped siRNA/lipid ratio were similar.
`Clinical signs were observed daily and body weights, serum chem-
`istry and hematology parameters were measured 72 h after dosing.
`KC2-SNALP was very well tolerated at the high dose levels exam-
`ined (relative to the observed ED50 dose) with no dose-dependent,
`clinically significant changes in key serum chemistry or hematology
`parameters (Table 2).
`
`Given the promising activity and safety profile observed in rodents,
`studies were initiated in nonhuman primates to investigate the trans-
`lation of DLin-KC2-DMA activity in higher species. For these stud-
`ies, we chose to target transthyretin (TTR), a hepatic gene of high
`therapeutic interest20. TTR is a serum protein synthesized primarily
`in the liver, and although amyloidogenic TTR mutations are rare,
`they are endemic to certain populations and can affect the periph-
`eral nerves, leading to familial amyloidotic polyneuropathy, and the
`heart, leading to familial amyloid cardiomyopathy. Currently, the
`only disease-modifying therapy is liver transplantation. We treated
`cynomolgus monkeys with a single 15-min intravenous infusion of
`KC2-SNALP–formulated siTTR at siRNA doses of 0.03, 0.1, 0.3 and
`1 mg/kg. Control animals received a single 15-min intravenous infu-
`sion of PBS or KC2-SNALP–formulated ApoB siRNA at a dose of
`1 mg/kg. Tissues were harvested 48 h after administration and
`liver mRNA levels of TTR were determined. A clear dose response
`was obtained with an apparent ED50 of ~0.3 mg/kg (Fig. 3b).
`A toxicological analysis indicated that the treatment was well tolerated
`at the dose levels tested, with no treatment-related changes in animal
`appearance or behavior. No dose-dependent, clinically significant
`alterations in key clinical chemistry or hematological parameters
`were observed (Supplementary Table 4).
`In summary, we applied a rational approach to the design of novel
`cationic lipids, which were screened for use in LNP-based siRNA
`delivery systems. Lipid structure was divided into three main func-
`tional elements: alkyl chain, linker and headgroup. With DLinDMA
`as a starting point, the effect of each of these elements was inves-
`tigated in a systematic fashion, by holding the other two constant.
`First, the alkyl chains were established, then linker was varied and,
`finally, different headgroup structures were explored. Using this
`approach, important structure-activity considerations for ioniz-
`able cationic lipids were described and lipids with improved activ-
`ity relative to the DLinDMA benchmark were identified. A SNALP
`formulation of the best-performing lipid (DLin-KC2-DMA) was
`well-tolerated in both rodent and nonhuman primates and exhib-
`ited in vivo activity at siRNA doses as low as 0.01 mg/kg in rodents,
`as well as silencing of a therapeutically significant gene (TTR) in
`nonhuman primates. Although the scope of the current work has
`been limited to hepatic delivery in vivo, the TTR silencing achieved
`in this work (ED50 ~ 0.3 mg/kg) represents a substantial improve-
`ment in activity relative to previous reports of LNP-siRNA mediated
`silencing in nonhuman primates.
`
`MeThoDS
`Methods and any associated references are available in the online
`version of the paper at http://www.nature.com/naturebiotechnology/.
`
`Note: Supplementary information is available on the Nature Biotechnology website.
`
`ACKNoWLEDGMENTS
`The authors thank K. McClintock for assistance with animal studies. The authors
`also thank the Centre for Drug Research and Development at the University
`
`nature biotechnology VOLUME 28 NUMBER 2 FEBRUARY 2010
`
`175
`
`© 2010 Nature America, Inc. All rights reserved.
`
`

`

`l e t t e r s
`
`of British Columbia for use of the NMR facilities and M. Heller for his expert
`assistance in setting up the 31P-NMR experiments.
`
`AUTHoR CoNTRIBUTIoNS
`J.C., M.A.C., P.R.C., T.D.M., M.J.H. and K.F.W. designed and advised on novel
`lipids. J.C., K.F.W. and M.S. synthesized novel lipids. M.J.H., T.D.M., J.C., K.F.W.,
`M.M., K.G.R., M.A.M., M.T. and M.J. analyzed and interpreted lipid data. T.D.M.,
`M.J.H. and M.A.T. co-directed novel lipid synthesis and screening program. S.C.S.
`designed and directed rodent in vivo studies. S.C.S., S.K.K., B.L.M., K.L., M.L.E.,
`M.K., A.P.S., Y.K.T., S.A.B., W.L.C., M.J.W. and E.J.C. generated rodent in vivo
`data, including Factor VII and tolerability analyses. L.N., V.K., T.B., R.A., Q.C.
`and D.W.Y.S. developed novel siRNAs targeting TTR. R.A. and A.A. designed and
`directed NHP in vivo studies. S.C.S., S.K.K., A.A., B.L.M., I.M., A.P.S., Y.K.T., R.A.,
`T.B., D.W. Y. S., S.A.B., J.Q., J.R.D. and A.d.F. analyzed and interpreted in vivo data.
`B.L.M., K.L., A.P.S., S.K.K., S.C.S. and E.J.C. generated and characterized preformed
`vesicle formulations with novel lipids. D.S. and C.K.C. developed methods and
`designed and conducted HPLC lipid analyses of preformed vesicle formulations.
`E.Y. and L.B.J. prepared SNALP formulations. P.R.C. directed biophysical studies
`and advised on methods. A.P.S., I.M.H., S.D. and K.W. performed biophysical
`characterization studies (pKa, NMR, differential scanning calorimetric) of novel
`lipids and formulations. M.J.H., P.R.C., T.D.M., A.P.S., I.M.H. and K.F.W. analyzed
`biophysical data. S.C.S., M.J.H., A.A. and P.R.C. co-wrote the manuscript. T.D.M.,
`M.M., M.A.M., M.A.T. and A.D.F. reviewed and edited the manuscript. S.C.S.,
`M.J.H., A.A., P.R.C., I.M. and A.D.F. were responsible for approval of the final draft.
`
`CoMPETING INTERESTS STATEMENT
`The authors declare competing financial interests: details accompany the full-text
`HTML version of the paper at http://www.nature.com/naturebiotechnology/.
`
`
`Published online at http://www.nature.com/naturebiotechnology/.
`reprints and permissions information is available online at http://npg.nature.com/
`reprintsandpermissions/.
`
`1. de Fougerolles, A.R. Delivery vehicles for small interfering RNA in vivo. Hum. Gene
`Ther. 19, 125–132 (2008).
`2. Whitehead, K.A., Langer, R. & Anderson, D.G. Knocking down barriers: advances
`in siRNA delivery. Nat. Rev. Drug Discov. 8, 129–138 (2009).
`3. Judge, A.D. et al. Confirming the RNAi-mediated mechanism of action of
`siRNA-based cancer therapeutics in mice. J. Clin. Invest. 119, 661–673 (2009).
`
`4. Judge, A.D. et al. Sequence-dependent stimulation of the mammalian innate
`immune response by synthetic siRNA. Nat. Biotechnol. 23, 457–462 (2005).
`5. Morrissey, D.V. et al. Potent and persistent in vivo anti-HBV activity of chemically
`modified siRNAs. Nat. Biotechnol. 23, 1002–1007 (2005).
`6. Zimmermann, T.S. et al. RNAi-mediated gene silencing in non-human primates.
`Nature 441, 111–114 (2006).
`7. Akinc, A. et al. A combinatorial library of lipid-like materials for delivery of RNAi
`therapeutics. Nat. Biotechnol. 26, 561–569 (2008).
`8. Frank-Kamenetsky, M. et al. Therapeutic RNAi targeting PCSK9 acutely lowers
`plasma cholesterol in rodents and LDL cholesterol in nonhuman primates. Proc.
`Natl. Acad. Sci. USA 105, 11915–11920 (2008).
`9. Hafez, I.M., Maurer, N. & Cullis, P.R. On the mechanism whereby cationic lipids
`promote intracellular delivery of polynucleic acids. Gene Ther. 8, 1188–1196
`(2001).
`10. Xu, Y. & Szoka, F.C. Jr. Mechanism of DNA release from cationic liposome/DNA
`complexes used in cell transfection. Biochemistry 35, 5616–5623 (1996).
`11. Zelphati, O. & Szoka, F.C. Jr. Mechanism of oligonucleotide release from cationic
`liposomes. Proc. Natl. Acad. Sci. USA 93, 11493–11498 (1996).
`12. Torchilin, V.P. Recent approaches to intracellular delivery of drugs and DNA and
`organelle targeting. Annu. Rev. Biomed. Eng. 8, 343–375 (2006).
`13. Semple, S.C. et al. Efficient encapsulation of antisense oligonucleotides in lipid
`vesicles using ionizable aminolipids: formation of novel small multilamellar vesicle
`structures. Biochim. Biophys. Acta 1510, 152–166 (2001).
`14. Maurer, N. et al. Spontaneous entrapment of polynucleotides upon electrostatic
`liposomes. Biophys. J. 80,
`interaction with ethanol-destabilized cationic
`2310–2326 (2001).
`15. Heyes, J., Palmer, L., Bremner, K. & Maclachlan, I. Cationic lipid saturation
`influences intracellular delivery of encapsulated nucleic acids. J. Control. Release
`107, 276–287 (2005).
`16. Semple, S.C., Chonn, A. & Cullis, P.R. Interactions of liposomes and lipid-based
`carrier systems with blood proteins: Relation to clearance behaviour in vivo. Adv.
`Drug Deliv. Rev. 32, 3–17 (1998).
`17. Bailey, A.L. & Cullis, P.R. Modulation of membrane fusion by asymmetric transbilayer
`distributions of amino lipids. Biochemistry 33, 12573–12580 (1994).
`18. Cullis, P.R. & de Kruijff, B. The polymorphic phase behaviour of phosphatidyl-
`ethanolamines of natural and synthetic origin. A 31P NMR study. Biochim. Biophys.
`Acta 513, 31–42 (1978

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