throbber
doi:10.1038/nature04688
`
`LETTERS
`
`RNAi-mediated gene silencing in non-human
`primates
`Tracy S. Zimmermann1, Amy C. H. Lee2, Akin Akinc1, Birgit Bramlage3, David Bumcrot1, Matthew N. Fedoruk2,
`Jens Harborth1, James A. Heyes2, Lloyd B. Jeffs2, Matthias John3, Adam D. Judge2, Kieu Lam2,
`Kevin McClintock2, Lubomir V. Nechev1, Lorne R. Palmer2, Timothy Racie1, Ingo Ro¨hl3, Stephan Seiffert3,
`Sumi Shanmugam1, Vandana Sood2, Ju¨rgen Soutschek3, Ivanka Toudjarska1, Amanda J. Wheat2, Ed Yaworski2,
`William Zedalis1, Victor Koteliansky1, Muthiah Manoharan1, Hans-Peter Vornlocher3 & Ian MacLachlan2
`
`The opportunity to harness the RNA interference (RNAi) pathway
`to silence disease-causing genes holds great promise for the
`development of therapeutics directed against targets that are
`otherwise not addressable with current medicines1,2. Although
`there are numerous examples of in vivo silencing of target
`genes after local delivery of small interfering RNAs (siRNAs)3–5,
`there remain only a few reports of RNAi-mediated silencing in
`response to systemic delivery of siRNA6–8, and there are no
`reports of systemic efficacy in non-rodent species. Here we show
`that siRNAs, when delivered systemically in a liposomal formu-
`lation, can silence the disease target apolipoprotein B (ApoB) in
`non-human primates. APOB-specific siRNAs were encapsulated in
`stable nucleic acid lipid particles (SNALP) and administered by
`intravenous injection to cynomolgus monkeys at doses of 1 or
`2.5 mg kg21. A single siRNA injection resulted in dose-dependent
`silencing of APOB messenger RNA expression in the liver 48 h
`after administration, with maximal silencing of >90%. This
`silencing effect occurred as a result of APOB mRNA cleavage at
`precisely the site predicted for the RNAi mechanism. Significant
`reductions in ApoB protein, serum cholesterol and low-density
`lipoprotein levels were observed as early as 24 h after treatment
`and lasted for 11 days at the highest siRNA dose, thus demonstrat-
`ing an immediate, potent and lasting biological effect of siRNA
`treatment. Our findings show clinically relevant RNAi-mediated
`
`gene silencing in non-human primates, supporting RNAi thera-
`peutics as a potential new class of drugs.
`ApoB is expressed predominantly in the liver and jejunum, and is
`an essential protein for the assembly and secretion of very-low-
`density lipoprotein (VLDL) and low-density lipoprotein (LDL),
`which are required for the transport and metabolism of cholesterol9.
`As a large, lipid-associated protein, ApoB is not accessible to target-
`ing with conventional therapies, but it is a highly relevant and
`validated disease target. Elevated ApoB and LDL levels are correlated
`with increased risk of coronary artery disease, and inadequate control
`of LDL–cholesterol after acute coronary syndromes results in
`increased risk of recurrent cardiac events or death10,11. Approaches
`targeting ApoB with second-generation antisense oligonucleotides
`have progressed to pre-clinical and clinical studies12. Despite progress
`in the management of hypercholesterolaemia using HMG-CoA
`reductase inhibitors and other drugs that affect dietary cholesterol,
`there remains a significant need for new therapeutic approaches.
`We have previously demonstrated silencing of Apob in rodents
`using cholesterol-conjugated siRNAs6. In the current study, we used
`a liposomal formulation of SNALP to evaluate systemic delivery
`of siRNA directed towards APOB. Preliminary evaluations were
`conducted in mice. Whereas administration of the Apob-specific
`siRNA siApoB-1, without formulation or chemical conjugation, at
`doses higher than 50 mg kg21 was previously shown to have no
`
`Figure 1 | SNALP–siRNA-mediated silencing of murine Apob is potent,
`specific, dose-dependent and long-lasting. a, Liver Apob mRNA levels
`normalized to Gapdh mRNA and serum ApoB-100 protein levels measured
`two days after single i.v. injections of saline, SNALP–siApoB-1 (1 mg kg21),
`mismatched SNALP–siApoB-MM (1 mg kg21) or empty SNALP vesicles
`(25 mg kg21) (n ¼ 5 per group). b, Liver Apob mRNA levels normalized to
`1Alnylam Pharmaceuticals Inc., 300 Third Street, Cambridge, Massachusetts 02142, USA. 2Protiva Biotherapeutics Inc., 100-3480 Gilmore Way, Burnaby, British Columbia
`V5G 4YI, Canada. 3Alnylam Europe AG, Fritz-Hornschuch-Str. 9, 95326 Kulmbach, Germany.
`© 2006 Nature Publishing Group
`
`Gapdh mRNA, assessed three days after i.v. administration of saline or 5, 2.5,
`1 or 0.5 mg kg21 SNALP–siApoB-2 (n ¼ 4 per group). c, Serum ApoB-100
`levels after i.v. administration of either saline or 2.5 mg kg21 SNALP–
`siApoB-2 (n ¼ 6 per group). Serum ApoB-100 levels for SNALP–siApoB-2-
`treated animals are relative to the saline-treated group for the same time
`point. Data show mean ^ s.d.
`
`1
`
`Moderna Ex 1027-p. 1
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`LETTERS
`
`NATURE
`
`Figure 2 | Systemic silencing of APOB mRNA in non-human primates.
`a, b, Liver APOB mRNA levels for 12 biopsies (three isolated from each of
`four liver lobes) were quantified relative to GAPDH mRNA either 48 h
`(a, n ¼ 4 animals per group) or 11 days (b, n ¼ 2) after treatment with
`SNALP–siApoB-2. Data shown are mean APOB/GAPDH mRNA
`
`levels ^ s.d. for each animal. Mean values (^s.d.) of the per cent APOB
`mRNA reduction relative to the saline treatment group are shown above
`each group. Asterisks indicate statistical significance compared with the
`saline-treated group (P , 0.005; ANOVA).
`
`0
`
`5
`rapid amplification of cDNA ends (RACE) analysis and identifi-
`cation of the predicted cleavage site, exactly ten nucleotides from the
`0
`end of the antisense strand of siApoB-2 (Supplementary Fig. 5).
`5
`Notably, APOB mRNA silencing was maintained for 11 days after the
`single 2.5 mg kg21 treatment, with APOB mRNA levels still reduced
`by 91 ^ 1.5% (Fig. 2b). Monkeys treated with the 1 mg kg21 dose
`showed varying degrees of recovery from ApoB silencing at the day 11
`time point. Although APOB mRNA was efficiently silenced in the
`liver, SNALP–siApoB-2 showed no silencing of APOB expressed in
`the jejunum (Supplementary Fig. 6), consistent with the absence of
`significant biodistribution of SNALP-formulated siRNAs to intesti-
`nal tissues in mice (Supplementary Fig. 3b).
`The degree and persistence of RNAi-mediated silencing observed
`in cynomolgus monkeys far exceeds the results obtained with
`rodents. The lasting RNAi-mediated effects in vivo are consistent
`with observed long-lasting silencing by siRNAs in other studies13,14,
`and the longer duration observed in primates may relate to species
`differences in the efficiency and stability of the RNA-induced
`
`in vivo silencing activity6, ,80% silencing of liver Apob mRNA and
`ApoB-100 protein was achieved with a single 1 mg kg21 dose of
`SNALP-formulated siApoB-1 (Fig. 1a). In contrast, no detectable
`reduction was observed with a SNALP-formulated mismatched
`siRNA (siApoB-MM) or empty SNALP vesicles, indicating that
`silencing is specific to the siRNA and is not caused by the lipo-
`somal carrier. This silencing effect of SNALP-formulated siRNA
`represents more than a 100-fold improvement in potency compared
`with systemic administration of cholesterol-conjugated siApoB-1
`(chol–siApoB-1) (Supplementary Fig. 1). Moreover, liposomal for-
`mulation of siRNA seems to be a general strategy for silencing
`hepatocyte targets, as demonstrated in mice for coagulation factor
`VII, green fluorescent protein and cyclophilin B (A.A., R. Constien
`and M.N.F., unpublished results).
`As siApoB-1 was originally designed to be cross-reactive to both
`mouse and human ApoB genes, and we planned to conduct RNAi
`studies in non-human primates, a second ApoB-specific siRNA,
`siApoB-2, was designed to be cross-reactive with mouse, human
`and cynomolgus monkey ApoB genes. siApoB-2 was also selected on
`the basis of in vitro gene silencing activity and the absence of
`immunostimulatory activity (data not shown). Murine studies
`showed that encapsulated siApoB-2 showed a dose-dependent
`reduction in Apob mRNA, with .90% silencing achieved at the
`highest (5 mg kg21) dose (Fig. 1b). After a single 2.5 mg kg21 dose of
`SNALP–siApoB-2, 80% silencing of liver Apob mRNA was associated
`with a 72% reduction in serum ApoB-100 protein. The silencing
`effect was detected for up to nine days, and was followed by recovery
`to normal protein levels by day 13 after treatment (Fig. 1c).
`To address the therapeutic potential of this systemic RNAi
`approach, we evaluated the pharmacokinetics, efficacy and safety
`of SNALP-formulated siApoB-2 in cynomolgus monkeys. We first
`determined the circulating half-life of SNALP–siApoB-2 in plasma
`samples collected from cynomolgus monkeys (n ¼ 2) receiving a
`single 2.5 mg kg21 intravenous (i.v.) injection of the siRNA. An
`elimination half-life of 72 min was measured for the siRNA (Sup-
`plementary Fig. 2), compared with a 38-min half-life in mice
`(Supplementary Fig. 3a).
`To evaluate efficacy, cynomolgus monkeys were treated with saline
`or SNALP-formulated siApoB-2 at doses of 1 or 2.5 mg kg21 (n ¼ 6
`Figure 3 | Phenotypic effects of RNAi-mediated silencing of APOB mRNA in
`per group). siApoB-2 treatment was associated with a clear and
`non-human primates. a–d, Serial plasma samples were obtained from
`statistically significant dose-dependent gene-silencing effect on
`cynomolgus monkeys treated with saline or 1 or 2.5 mg kg21 SNALP–
`cynomolgus liver APOB mRNA. Forty-eight hours after treatment,
`APOB mRNA was reduced by 68 ^ 12% (mean ^ s.d., n ¼ 4,
`siApoB-2, and measured for ApoB-100 (a), total serum cholesterol (b), LDL
`P ¼ 0.004) and 90 ^ 1% (n ¼ 4, P ¼ 0.002) for the 1 mg kg21 and
`(c) and HDL (d) levels. Data show levels as a percentage of pre-dose values
`and are expressed as mean ^ s.d. Data sets collected at 0, 12, 24 and 48 h
`2.5 mg kg21 groups, respectively (Fig. 2a). Gene silencing was found
`have a group size of six, and data sets collected at later time points have a
`to be consistent across the liver and correlated with detectable tissue
`group size of two. Data points marked with asterisks are statistically
`levels of siApoB-2 (Supplementary Fig. 4). We also confirmed this
`significant compared with saline-treated animals (*P , 0.05, **P , 0.005;
`APOB mRNA silencing to be mediated by RNAi, as demonstrated by
`ANOVA).
`© 2006 Nature Publishing Group
`
`2
`
`Moderna Ex 1027-p. 2
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`NATURE
`
`LETTERS
`
`after 11 days, which prevented full evaluation of the time course for
`RNAi-mediated effects. Although further optimization of treatment
`regimen and safety profile characterization may be required, our data
`suggest that systemic delivery of siRNAs for targeting hepatocyte-
`specific genes in a higher species is possible. Furthermore, the rapid
`and long-lasting silencing of APOB using RNAi may represent a new
`strategy for reducing LDL–cholesterol in several relevant clinical
`settings.
`
`METHODS
`Additional details of the methods used are provided in the Supplementary
`Information.
`siRNA formulation. The SNALP formulation contained the lipids 3-N-[(q -
`methoxypoly(ethylene glycol)2000)carbamoyl]-1,2-dimyristyloxy-propylamine
`(PEG-C-DMA), 1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DLinDMA),
`1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) and cholesterol, in a
`2:40:10:48 molar per cent ratio.
`In vivo experiments. Saline and siRNA preparations were administered by tail
`vein injection under normal pressure and low volume (0.01 ml g21) for all
`rodent experiments. Cynomolgus monkeys (n ¼ 6 per group) received either
`2 ml kg21 phosphate buffered saline or 1 or 2.5 mg kg21 SNALP–siApoB-2 at a
`dose volume of 1.25 ml kg21 as bolus i.v. injections via the saphenous vein. For
`mRNA measurements, three liver biopsies per lobe were collected 48 h (n ¼ 4)
`or 264 h (n ¼ 2) after siRNA administration.
`Bioanalytical methods. The QuantiGene assay (Genospectra) was used to
`quantify reduction in APOB mRNA levels relative to the housekeeping gene
`GAPDH in lysates prepared from mouse liver or cynomolgus monkey liver and
`jejunum as previously described6 but with minor variations. Mouse6 and
`cynomolgus monkey ApoB-100 protein levels were quantified by enzyme-linked
`immunosorbent assay (ELISA). LDL and HDL lipoprotein content were
`determined for plasma samples (250 m l) as described previously6.
`Statistical analysis. P-values were calculated for comparison of SNALP–siApoB-
`2-treated animals with saline-treated animals using analysis of variance
`(ANOVA, two-factor without replication) with an alpha value of 0.05. P-values
`less than 0.05 were considered significant.
`
`Received 12 January; accepted 6 March 2006.
`Published online 26 March 2006.
`
`2.
`
`3.
`
`4.
`
`silencing complex (RISC), the mitotic state of hepatocytes and/or the
`tissue stability of the siRNA.
`The expected biological effects resulting from APOB mRNA
`silencing include reduction in the blood levels of ApoB-100 protein,
`total cholesterol and LDL. To evaluate the kinetics of these down-
`stream effects, we analysed plasma sampled serially from individual
`monkeys before and during the 11-day time course of the single-dose
`siApoB-2 study. Plasma ApoB-100 protein levels were reduced as
`early as 12 h after administration of 1 or 2.5 mg kg21 SNALP–
`siApoB-2, reaching nadirs of 35 ^ 2% and 22 ^ 9% of pre-treat-
`ment levels, respectively, 72 h after treatment (Fig. 3a). Animals that
`received the higher siRNA dose maintained a marked reduction in
`ApoB protein between 2 and 11 days after treatment, consistent with
`the lasting effect on mRNA silencing. Monkeys that received the
`lower siRNA dose showed an intermediate degree of ApoB protein
`reduction that returned to pre-dose levels by day 11, consistent with
`the observed recovery in APOB mRNA.
`Serum cholesterol
`levels were similarly reduced, in a dose-
`dependent manner and with comparable kinetics (Fig. 3b). The
`maximum cholesterol reduction of 62 ^ 5.5% (n ¼ 2, P ¼ 0.006)
`observed for the high dose siRNA group would be considered
`clinically significant for patients with hypercholesterolaemia,
`and exceeds levels of cholesterol reduction reported clinically for
`currently approved cholesterol-lowering drugs.
`Administration of SNALP–siApoB-2 also resulted in dramatic and
`rapid dose-dependent reduction in the ApoB-containing lipoprotein
`particle LDL. Reduction in LDL relative to pre-dose levels was
`observed as early as 24 h after treatment for both doses of SNALP–
`siApoB-2 (Fig. 3c). In contrast, there were no significant changes in
`circulating levels of the non-ApoB-containing high-density lipopro-
`tein particle (HDL, Fig. 3d). The reduction in LDL persisted over the
`11-day study for both siApoB-2 treatment groups, with a maximum
`82 ^ 7% decrease compared to pre-treatment levels observed for the
`high-dose group at day 11 (n ¼ 2, P ¼ 0.003). The time required for
`the biological effects to return to pre-dose levels was not determined
`for the high-dose group because the endpoint for this study was
`defined using rodent data, which indicated a faster rate of recovery.
`The rapid onset and lasting effect on lipoprotein metabolism suggest
`that siRNAs targeting APOB may be a valuable therapeutic strategy
`for achieving plaque stabilization in acute coronary syndromes10,11,
`as HMG-CoA reductase inhibitors can require up to 4–6 weeks to
`have the desired clinical effects15.
`An important consideration for the therapeutic application of
`siRNA relates to its general safety, as well as to the safety profile
`associated with specific delivery technologies. General tolerability
`as well as specific toxicities (such as activation of complement,
`coagulation and cytokines) were evaluated for all monkeys in this
`study. We observed no treatment-related effects on the appearance or
`behaviour of animals treated with SNALP–siApoB-2 compared with
`saline-treated animals. There was no evidence for complement
`activation, delayed coagulation, pro-inflammatory cytokine pro-
`duction (Supplementary Table 1) or changes in haematology pa-
`rameters (data not shown), toxicities that have been observed
`previously with treatments using related approaches16–19. Across a
`systematic evaluation, the only detected change in primates treated
`with SNALP–siApoB-2 was a transient increase in liver enzymes in
`monkeys that received the high dose of SNALP–siApoB-2. The
`observed transaminosis peaked 48 h after treatment and was highly
`variable across individual animals. These effects, which were
`observed only at the highest dose of SNALP–siApoB-2, were com-
`pletely reversible, with normalization by day 6 notwithstanding
`continued biological efficacy.
`Our study highlights the potential for therapeutic gene silencing
`using systemic RNAi in non-human primates. A single, low dose of
`APOB-specific siRNA resulted in rapid and lasting RNAi-mediated
`gene silencing, with associated and profound phenotypic changes.
`The study was limited by the premature termination of the protocol
`© 2006 Nature Publishing Group
`
`5.
`
`6.
`
`7.
`
`9.
`
`1. Novina, C. D. & Sharp, P. A. The RNAi revolution. Nature 430, 161–-164
`(2004).
`Shankar, P., Manjunath, N. & Lieberman, J. The prospect of silencing disease
`using RNA interference. J. Am. Med. Assoc. 293, 1367–-1373 (2005).
`Thakker, D. R. et al. Neurochemical and behavioral consequences of
`widespread gene knockdown in the adult mouse brain by using nonviral RNA
`interference. Proc. Natl Acad. Sci. USA 101, 17270–-17275 (2004).
`Bitko, V., Musiyenko, A., Shulyayeva, O. & Barik, S. Inhibition of respiratory
`viruses by nasally administered siRNA. Nature Med. 11, 50–-55 (2005).
`Palliser, D. et al. An siRNA-based microbicide protects mice from lethal herpes
`simplex virus 2 infection. Nature 439, 89–-94 (2006).
`Soutschek, J. et al. Therapeutic silencing of an endogenous gene by systemic
`administration of modified siRNAs. Nature 432, 173–-178 (2004).
`Song, E. et al. Antibody mediated in vivo delivery of small interfering RNAs via
`cell-surface receptors. Nature Biotechnol. 23, 709–-717 (2005).
`8. Morrissey, D. V. et al. Potent and persistent in vivo anti-HBV activity of
`chemically modified siRNAs. Nature Biotechnol. 23, 1002–-1007 (2005).
`Brown, M. S. & Goldstein, J. L. A receptor-mediated pathway for cholesterol
`homeostasis. Science 232, 34–-47 (1986).
`10. Cannon, C. P. et al. Intensive versus moderate lipid lowering with statins after
`acute coronary syndromes. N. Engl. J. Med. 350, 1495–-1504 (2004).
`11. Ridker, P. M. et al. C-reactive protein levels and outcomes after statin therapy.
`N. Engl. J. Med. 352, 20–-28 (2005).
`12. Crooke, R. M. et al. An apolipoprotein B antisense oligonucleotide lowers LDL
`cholesterol in hyperlipidemic mice without causing hepatic steatosis. J. Lipid
`Res. 46, 872–-884 (2005).
`13. Song, E. et al. Sustained small interfering RNA-mediated human
`immunodeficiency virus type 1 inhibition in primary macrophages. J. Virol. 77,
`7174–-7181 (2003).
`14. Bartlett, D. W. & Davis, M. E. Insights into the kinetics of siRNA-mediated gene
`silencing from live-cell and live-animal bioluminescent imaging. Nucleic Acids
`Res. 34, 322–-333 (2006).
`15. Lennernas, H. & Fager, G. Pharmacodynamics and pharmacokinetics of the
`HMG-CoA reductase inhibitors. Similarities and differences. Clin.
`Pharmacokinet. 32, 403–-425 (1997).
`16. Levin, A. A. A review of the issues in the pharmacokinetics and toxicology of
`phosphorothioate antisense oligonucleotides. Biochim. Biophys. Acta 1489,
`69–-84 (1999).
`
`3
`
`Moderna Ex 1027-p. 3
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`LETTERS
`
`NATURE
`
`17. Chonn, A., Cullis, P. R. & Devine, D. V. The role of surface charge in the
`activation of the classical and alternative pathways of complement by
`liposomes. J. Immunol. 146, 4234–-4241 (1991).
`18. Hornung, V. et al. Sequence-specific potent induction of IFN-a by short
`interfering RNA in plasmacytoid dendritic cells through TLR7. Nature Med. 11,
`263–-270 (2005).
`Judge, A. D. et al. Sequence-dependent stimulation of the mammalian innate
`immune response by synthetic siRNA. Nature Biotechnol. 23, 457–-462 (2005).
`
`19.
`
`Supplementary Information is linked to the online version of the paper at
`www.nature.com/nature.
`
`Acknowledgements We are grateful to P. Sharp, J. Maraganore and
`N. Mahanthappa for their assistance and support in this study. We would also
`like to thank W. J. Schneider, J. Frohlich, M. Hayden and J. E. Vance for
`
`discussions. We acknowledge the technical assistance of C. Woppmann and
`A. Wetzel, and thank V. Kesavan and G. Wang for preparation of the
`cholesterol-conjugated siRNA used in this study. Finally, we thank S. Young for
`providing anti-ApoB antibodies. This work was supported by grants from the
`National Science and Engineering Research Council of Canada (to A.J.W. and
`M.N.F.).
`
`Author Contributions This work represents the outcome of a collaboration
`between scientists at Alnylam Pharmaceuticals and Protiva Biotherapeutics Inc.
`
`Author Information Reprints and permissions information is available at
`npg.nature.com/reprintsandpermissions. The authors declare competing
`financial interests: details accompany the paper at www.nature.com/nature.
`Correspondence and requests for materials should be addressed to T.S.Z.
`(tzimmermann@alnylam.com) or I.M. (ian@protivabio.com).
`
`4
`
`© 2006 Nature Publishing Group
`
`Moderna Ex 1027-p. 4
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`1
`
`
`
`
`
`
`
`
`
`
`
`
`
`Supplementary Information: Methods
`“RNAi-mediated Gene Silencing in Non-Human Primates”
`Zimmermann. T.S. et al.
`
`Moderna Ex 1027-p. 5
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`2
`
`Supplementary Methods
`
`Synthesis of siRNAs. The siRNAs used in this study each consisted of a 21 nucleotide
`
`(nt) sense strand and a 23 nt antisense strand resulting in a single 2 nt overhang at the 3'-
`
`end of the antisense strand of the annealed duplex. siApoB-1 (position 10167-10187,
`
`NM_000384) sense: 5'-GUCAUCACACUGAAUACCAAU-3', antisense: 5'-
`
`AUUGGUAUUCAGUGUGAUGACAC-3', siApoB-MM sense: 5’-
`
`GUGAUCAGACUCAAUACGAAU-3’, antisense: 5’-
`
`AUUCGUAUUGAGUCUGAUCACAC-3’. The sequence and synthesis of Chol-
`siApoB-1 are as previously described1. siApoB-2 (position 2098-2118, NM_000384)
`sense: 5’-GGAAUCuuAuAuuuGAUCcA*A-3’, antisense: 5’-
`
`uuGGAUcAAAuAuAAGAuUCc*c*U-3’. 2’O-Methyl modified nucleotides are in
`
`lower case and phosphorothioate linkages are represented by asterisks. Sense and
`antisense strands for siApoB-2 were synthesized as described previously1 except that
`N4-Acetylcytidine phosphoramidites were used for cytidine residues. These
`oligonucleotides were characterized by ESMS and anion-exchange HPLC. siRNAs
`
`were generated by annealing equimolar amounts of complementary sense and antisense
`
`strands.
`
`Encapsulation of siRNA. siRNAs were encapsulated by an adaptation of the method
`of Jeffs et al2. The SNALP formulation contained the lipids PEG-C-
`DMA:DLinDMA:DSPC:Cholesterol (2:40:10:48 molar percent). 3-N-[(ω-Methoxy
`poly(ethylene glycol)2000)carbamoyl]-1,2-dimyristyloxy-propylamine (PEG-C-DMA)
`MW 2524 and 1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane (DLinDMA) MW 616
`were prepared by Protiva Biotherapeutics3, 1,2-Distearoyl-sn-glycero-3-phosphocholine
`(DSPC) MW 790 was obtained from Avanti Polar Lipids (Alabaster, AL), and
`
`cholesterol MW 387 was obtained from Sigma (Oakville, ON). The particle sizes of the
`
`SNALP samples used in this study were 77-83 nm with a polydispersity range of 0.09-
`
`Moderna Ex 1027-p. 6
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`3
`
`0.15. Nucleic acid encapsulation efficiencies were 92-97%. Particle size was
`
`determined using a Malvern Instruments Zetasizer 3000HSA (Malvern, UK) and
`
`nucleic acid encapsulation efficiency was determined using the Ribogreen assay as
`described elsewhere3.
`
`In vivo rodent experiments. All siRNAs were administered via tail vein injection
`
`under normal pressure and at a dosing volume of 0.01 ml/g. Female 8-10 week old
`
`C57BL/6 mice (Charles River Laboratories, MA) were used for the Chol-siApoB-1 and
`
`SNALP siApoB-1 dose response experiments. For the Chol-siApoB-1 experiment mice
`
`received either saline or Chol-siApoB-1 at doses of 100, 50, 25 or 12.5 mg/kg and for
`
`the SNALP siApoB-1 dose response mice received either saline or SNALP siApoB-1 at
`
`siRNA doses of 1, 0.5, 0.25 or 0.1 mg/kg. Liver mRNA levels were assessed 72 h after
`
`injection. For determination of specificity of apoB knockdown, five week old female
`
`BALB/c mice (Harlan Labs, IN) received either saline, SNALP siApoB-1 or SNALP
`
`siApoB-MM at a siRNA dose of 1 mg/kg or empty SNALP vehicle at an equivalent
`
`lipid dosage (25 mg/kg). Animals were sacrificed two days after treatment for liver
`
`mRNA and serum apoB-100 protein measures. For the dose response and duration of
`
`effect experiments, eight week old female C57BL/6 mice (Charles River Laboratories,
`
`MA) received either saline or SNALP siApoB-2. Liver mRNA levels were determined
`
`72 h after injection of 5, 2.5, 1 or 0.5 mg/kg SNALP siApoB-2 for the dose response
`
`experiment. For the duration of effect experiment, animals received either saline or 2.5
`
`mg/kg SNALP siApoB-2 and were anesthetized using isofluorine for collection of
`
`serum samples by retro-orbital bleed pre-dose (d0) and 3, 6, 9 and 13 days post-dose for
`
`measurement of apoB-100 protein. All procedures used in animal studies conducted at
`
`Alnylam were approved by the Institutional Animal Care and Use Committee (IACUC)
`
`and were consistent with local, state, and federal regulations as applicable. Animal
`
`studies conducted at Protiva were performed under the oversight of Protiva’s
`
`Moderna Ex 1027-p. 7
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`4
`
`Institutional Animal Care and Use Committee (IACUC) in accordance with the
`
`Canadian Council on Animal Care guidelines.
`
`In vivo non-human primate experiment. For determination of the rate of plasma
`
`clearance of SNALP siApoB-2 in cynomolgus monkeys, two animals received 2.5
`
`mg/kg SNALP siApoB-2 via bolus i.v. injection in the saphenous vein and plasma
`
`samples were collected 0.25, 0.5, 1, 2, 4, 6, 8, 12 and 24 h post-dose. In addition, blood
`
`samples were taken pre-dose, 6 and 24 h post-dose for Interferon-γ and IL-6 measures
`
`and liver samples (three punch biopsies per lobe, twelve total) were collected 24 h post-
`
`dose for determination of siRNA distribution and apoB mRNA levels. Eight jejunum
`
`sections were isolated from animals treated with saline, 1 or 2.5 mg/kg SNALP siApoB-
`
`2 48 h (n = 4) or 264 h (n = 2) post-dose for mRNA measurements. Plasma samples
`
`from animals treated with saline, 1 or 2.5 mg/kg SNALP siApoB-2 were collected pre-
`
`dose, 12, 24 and 48 h post-dose for all animals and 72, 96, 144, 192 and 264 h post-dose
`
`for two animals per group for apoB-100 protein measurements. Animals were fasted for
`
`16 h prior to blood sampling for total serum cholesterol and lipoprotein collections pre-
`
`dose, 24 and 48 h post-dose for all animals and 144 and 264 h post-dose for two animals
`
`per group. Blood samples were collected pre-dose, 0.25 and 48 h post-dose for
`
`determination of complement Bb, CH50 and activated partial thromboplastin time
`
`(APTT) (n = 6 per treatment group) and pre-dose, 24, 48 (n = 6 per treatment group),
`
`144 and 264 h post-dose (n = 2 per treatment group) for determination of alanine
`
`aminotransferase (ALT), aspartate aminotransferase (AST), bilirubin and urea nitrogen
`
`levels. All procedures using cynomolgus monkeys were conducted by a certified
`
`contract research organization using protocols consistent with local, state, and federal
`
`regulations as applicable and approved by the Institutional Animal Care and Use
`
`Committee (IACUC).
`
`Moderna Ex 1027-p. 8
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`5
`
`In vivo bioanalytical methods for non-human primate experiments. For
`
`determination of the circulation half-life of SNALP siApoB-2 in primates, 30 µl plasma
`(one per animal and time point) was incubated at 42oC for 20 min in a buffer containing
`1.7 mg/ml proteinase K, 0.1 M Tris-Cl pH 7.5, 12.2 mM EDTA, 0.15 M NaCl, 1%
`
`SDS, 4.2 µM 40mer RNA internal standard in a 96-well plate. Samples were spin-
`
`filtered using a 0.2 µm filter plate and the filter was washed two times with 30 µl water
`
`(Varian, CA). Washes were combined with the initial filtrate and analyzed by ion
`
`exchange HPLC under denaturing conditions at pH 8 and 80°C with detection at 260
`
`nm. Under these conditions, the siRNA eluted as two well-separated single strands.
`
`The internal standard, used for normalization of small differences in recovered filtrate
`
`volume, eluted at a later time than the two single strands of the siRNA. The amount of
`
`full length sense and antisense strand was determined relative to an external calibration
`
`curve derived from SNALP-formulated siApoB-2 and was calibrated over a linear range
`
`of 5-200 pmoles RNA. An estimated constant of 40 ml plasma per kg animal weight
`
`was used for calculation of the total amount of siRNA and the percent injected dose for
`
`each time point was calculated as the ratio of the amount of the sense or antisense strand
`
`to the amount of injected siRNA. Each data point represents the group mean ± s.d.
`
`The QuantiGene® assay (Genospectra, CA) was performed to quantitate
`
`reduction of apoB mRNA relative to the house keeping gene GAPDH in lysates
`prepared from mouse liver or cynomolgus liver and jejunum as previously described1
`with minor variations. For detection of cynomolgus mRNA, the apoB probe set was
`
`specific to human apoB (positions 13870 to 14110, NM_000384) and cross-reactive to
`
`the Macaque fascicularis apoB sequence (positions 380 to 615, CO775384) and the
`
`GAPDH probe set was specific for human GAPDH (positions 224 to 444, NM_002046)
`
`and cross-reactive to M. fasciularis GAPDH (positions 142 to 362, AB158631).
`
`Moderna Ex 1027-p. 9
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`6
`
`siRNA distribution in cynomolgus liver samples was detected using a
`
`ribonuclease protection assay with a radiolabeled probe complementary to the antisense
`strand of siApoB-2 using methods previously described1. The assay was performed on
`total RNA isolated from each of the twelve liver biopsies.
`
`5’ RACE analysis was performed using total RNA (5 µg) from pooled liver
`biopsies (4 biopsies, 1 per lobe) as described previously1. Ligated RNA was reverse
`transcribed using a gene specific primer (GSP: 5’-
`
`TGGAAGAAGTTGGTGTTCATCTGGA-3’). Two rounds of PCR were performed.
`
`First round PCR using primers complementary to the RNA adaptor (GeneRacer,
`
`Invitrogen, CA) and apoB mRNA (Rev1: 5’-TCTTTGGTATAGCCAAAGTGGTCCA-
`
`3’). Second round PCR using nested primers complementary to the RNA adaptor and
`
`apoB mRNA (Rev2: 5’-AAAGCTTTGTTGACACTGTCTGGGAA-3’). The cleavage
`
`site at 2108/2109 of the apoB mRNA (NM_000384) was confirmed by sequencing of
`
`the PCR products.
`
`Cynomolgus apoB-100 was detected from plasma samples using a sandwich
`
`ELISA consisting of a polyclonal goat anti-human apoB capture antibody (Chemicon
`
`International, CA) and a horseradish peroxidase-conjugated goat anti-human apoB-100
`
`polyclonal detection antibody (Academy Bio-Medical Company, TX).
`
`In vivo rodent PK experiment. Radiolabeled SNALP was prepared for plasma
`
`clearance and tissue distribution studies by incorporation of 2.7 µCi/mg total lipid of the
`non-exchangeable lipid label 3H-CHE4. SNALP was administered at a siRNA dose of 2
`mg/kg via lateral tail vein injection in eight week old female BALB/c mice (Harlan
`
`Labs, IN) and blood was collected via tail vein nick over a 24 h period. At 24 h after
`
`injection, mice were euthanized and harvested tissues were homogenized in FastPrep
`
`Lysing Matrix Tubes (MP Biomedicals, CA) containing distilled water. Tissue
`
`homogenates were assayed for radioactivity by liquid scintillation counting with
`
`Moderna Ex 1027-p. 10
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`7
`
`Picofluor 40 and whole blood was assayed using Picofluor 15 (Perkin-Elmer, Boston,
`
`MA).
`
`1. Soutschek, J. et al. Therapeutic silencing of an endogenous gene by systemic
`
`administration of modified siRNAs. Nature 432, 173-8 (2004).
`
`2. Jeffs, L. B. et al. A scalable, extrusion-free method for efficient liposomal
`
`encapsulation of plasmid DNA. Pharm. Res. 22, 362-72 (2005).
`
`3. Heyes, J., Palmer, L., Bremner, K. & MacLachlan, I. Cationic lipid saturation
`
`influences intracellular delivery of encapsulated nucleic acids. J. Control Release 107,
`
`276-87 (2005).
`
`4. Stein, Y., Halperin, G. & Stein, O. Biological stability of [3H]cholesteryl oleyl ether
`
`in cultured fibroblasts and intact rat. FEBS Lett. 111, 104-6 (1980).
`
`
`
`Moderna Ex 1027-p. 11
`Moderna v Arbutus
`IPR2019-00554
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Supplementary Information: Figures 1-6 and Table 1
`“RNAi-Mediated Gene Silencing in Non-Human Primate”
`Zimmermann, T.

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