Sei sulla pagina 1di 10

Articles

Postexposure protection of non-human primates against


a lethal Ebola virus challenge with RNA interference:
a proof-of-concept study
Thomas W Geisbert, Amy C H Lee*, Marjorie Robbins*, Joan B Geisbert, Anna N Honko, Vandana Sood, Joshua C Johnson, Susan de Jong,
Iran Tavakoli, Adam Judge, Lisa E Hensley, Ian MacLachlan

Summary
Lancet 2010; 375: 1896905 Background We previously showed that small interfering RNAs (siRNAs) targeting the Zaire Ebola virus (ZEBOV)
See Comment page 1850 RNA polymerase L protein formulated in stable nucleic acid-lipid particles (SNALPs) completely protected guineapigs
*Contributed equally to the when administered shortly after a lethal ZEBOV challenge. Although rodent models of ZEBOV infection are useful
report for screening prospective countermeasures, they are frequently not useful for prediction of ecacy in the more
National Emerging Infectious stringent non-human primate models. We therefore assessed the ecacy of modied non-immunostimulatory
Diseases Laboratories Institute siRNAs in a uniformly lethal non-human primate model of ZEBOV haemorrhagic fever.
(Prof T W Geisbert PhD,
J B Geisbert), Department of
Microbiology Methods A combination of modied siRNAs targeting the ZEBOV L polymerase (EK-1 mod), viral protein (VP)
(Prof T W Geisbert), and 24 (VP24-1160 mod), and VP35 (VP35-855 mod) were formulated in SNALPs. A group of macaques (n=3) was given
Department of Medicine these pooled anti-ZEBOV siRNAs (2 mg/kg per dose, bolus intravenous infusion) after 30 min, and on days 1, 3, and
(Prof T W Geisbert), Boston
University School of Medicine,
5 after challenge with ZEBOV. A second group of macaques (n=4) was given the pooled anti-ZEBOV siRNAs after
Boston, MA, USA; Virology 30 min, and on days 1, 2, 3, 4, 5, and 6 after challenge with ZEBOV.
Division, US Army Medical
Research Institute of Infectious
Findings Two (66%) of three rhesus monkeys given four postexposure treatments of the pooled anti-ZEBOV siRNAs
Diseases, Fort Detrick, MD, USA
(A N Honko PhD, J C Johnson BSc, were protected from lethal ZEBOV infection, whereas all macaques given seven postexposure treatments were
L E Hensley PhD); and Tekmira protected. The treatment regimen in the second study was well tolerated with minor changes in liver enzymes that
Pharmaceuticals, Burnaby, BC, might have been related to viral infection.
Canada (A C H Lee MSc,
M Robbins PhD, V Sood MSc,
S de Jong PhD, I Tavakoli BSc, Interpretation This complete postexposure protection against ZEBOV in non-human primates provides a model for
A Judge PhD, I MacLachlan PhD) the treatment of ZEBOV-induced haemorrhagic fever. These data show the potential of RNA interference as an
Correspondence to: eective postexposure treatment strategy for people infected with Ebola virus, and suggest that this strategy might
Prof Thomas W Geisbert, Boston also be useful for treatment of other emerging viral infections.
University School of Medicine,
National Emerging Infectious
Diseases Laboratories Institute,
Funding Defense Threat Reduction Agency.
620 Albany Street, Room 401B,
Boston, MA 02118, USA Introduction polymerase activity of the complex. This protein is an
geisbert@bu.edu
For more than 30 years, Ebola virus (EBOV) has been ideal target for antiviral interventions not only because its
associated with periodic episodes of haemorrhagic fever suppression should lead to a nearly complete loss of all
in Central Africa that produce severe disease in infected RNA synthesis, but also because of the absence of similar
patients. Mortality rates during outbreaks have ranged proteins in mammalian cells. VP24 and VP35 are also
from 50% with the Sudan species of EBOV (SEBOV) to promising targets for antiviral interventions since both
up to 90% with the Zaire species (ZEBOV).1 An outbreak have been shown to have inhibitory eects on the host
towards the end of 2007, caused by an apparently new type 1 interferon response. Specically, VP35 functions as
species of EBOV in Uganda, seemed to be less a type 1 interferon antagonist by blocking the activation of
pathogenic than SEBOV or ZEBOV with a case fatality interferon regulatory factor 3 and possibly preventing
rate of about 25%.2 transcription of interferon .4 Consistent with these
EBOV particles contain a non-infectious RNA genome ndings, mutations of ZEBOV VP35 rendered an adapted
of roughly 19 kilobases that encodes seven structural virus avirulent in guineapigs.5 VP24 expression interferes
proteins and one non-structural protein. The gene order with type 1 interferon signalling,6 and mutations in
is 3leader, nucleoprotein, virion protein (VP) 35 (VP35), VP24 have been linked to adaptation by ZEBOV to
VP40, glycoprotein, VP30, VP24, polymerase L protein, produce a lethal infection in mice7 and guineapigs.8
and 5 trailer.3 Four of these proteinsnucleoprotein, Although there are no vaccines or postexposure
VP30, VP35, and the polymerase L proteinare associated treatments for prevention or management of EBOV
with the viral genomic RNA in the ribonucleoprotein infections, progress has been made over the past few years
complex. The L protein and VP35 make up the polymerase in the development of candidate preventive vaccines that
complex, which transcribes and replicates the EBOV can protect non-human primates against infection with
genome. The L protein provides the RNA-dependent RNA EBOV.1 Progress in the development of antiviral drugs and

1896 www.thelancet.com Vol 375 May 29, 2010


Articles

other postexposure interventions has been much slower. siRNA at 08 nmol/L, 40 nmol/L, and 200 nmol/L)
RNA interference represents a promising intervention for were added to a 96-well plate before addition of 80 L of
the treatment of diseases, including those caused by HepG2 cells (15 000 cells per well). Cells were lysed 48 h
viruses. Indeed, RNA interference has been used in cell after transfection. The dual-luciferase reporter assay
culture systems and rodents to inhibit the replication of system (Promega) and a Berthold luminometer (Berthold
several pathogens that cause disease in human beings. Detection Systems, Pforzheim, Germany) were used to
However, its usefulness as an eective treatment in non- measure Renilla luciferase (fused to ZEBOV VP24 or
human primate models of human infectious diseases has ZEBOV VP35 transgene) and rey luciferase signals.
been assessed in only two studies.9,10 Small interfering The Renilla luciferase signal was normalised to the rey
RNAs (siRNAs) inhibited the replication of hepatitis GB luciferase signal and expressed as percentage gene
virus B in a non-lethal marmoset surrogate model of expression relative to a plasmid-only control assigned a
human hepatitis C.9 siRNAs against severe acute value of 100%. Table 1 shows the sequences of siRNAs
respiratory syndrome (SARS) coronavirus inhibited the that were used.
virus replication in a non-lethal rhesus monkey model.10
However, unmodied immune stimulatory siRNAs were ZEBOV infection of Vero E6 cells
used in both of these studies, potentially confounding the To assess whether mRNAs for polymerase L, VP35, and
interpretation of the results. Until now, the usefulness of VP24 were cleaved by the specic mechanism of RNA
RNA interference as a postexposure treatment has not interference, we plated Vero E6 cells at 100 000 cells per mL
been assessed in a lethal model of a human infectious (2 mL per well in a six-well plate), and after 24 h treated
disease in non-human primates. them with stable nucleic acid-lipid particles (SNALPs;
In a previous study, we identied EK-1, an siRNA that 50 nmol/L) containing siRNAs modied by substitution of
targeted the ZEBOV polymerase L protein, and inhibited 2-O-methyl versions of guanines and uridines (EK-1-mod,
the replication of ZEBOV in vitro, and completely protected VP24-1160-mod, or VP35-855-mod), ZEBOV cocktail
ZEBOV-infected guineapigs.11 Although EK-1 was eective, (consisting of EK-1-mod, VP24-1160-mod, and VP35-855-
we chose to use a cocktail of three siRNAs in the present mod in a 1:1:1 ratio), or luciferase siRNA (Luc mod) for 16 h
study, one each for ZEBOV L protein, VP24, and VP35 genes before infection with ZEBOV (Kikwit strain, at
for further studies in non-human primates on the premise 01 multiplicity of infection). 24 h after infection, cell
that a combination of siRNAs will aid in targeting potential supernatants were harvested for quantitative real-time RT-
RNA-interference escape mutants, as has been shown for PCR, and cell monolayers were lysed with Trizol
HIV-1 and polio viruses.12 By targeting three dierent viral (Invitrogen) for isolation of total RNA and rapid
gene products, we also aimed to inactivate the virus in amplication of cDNA ends (5RACE; Invitrogen).
three dierent stages of its life cycle.
Quantitative RT-PCR
Methods We puried viral RNA using the Qiagen QIAmp viral RNA
Design and in-vitro screening of siRNAs mini kit (Qiagen, Valencia, CA, USA) according to the
We designed siRNAs to target regions of the ZEBOV manufacturer's protocol. One-step quantitative real-time
(Mayinga strain) polymerase L, VP24, and VP35 genes RT-PCR reactions were done on a LightCycler 480 (Roche,
using a conventional siRNA design algorithm. We Indianapolis, IN, USA) in 20 L volumes with 5 L of For algorithm see http://www.
excluded siRNAs with sequence homology to any human puried RNA and the Superscript III One-Step RT-PCR rockefeller.edu/labheads/tuschl/
sirna.html
reference mRNAs of 16 or more contiguous bases within System (Invitrogen). Primers (forward 5-CGGACCT
the core duplex using the Basic Local Alignment Search GGTTTGGTTGTG-3; reverse 5-GCTGCAGTGTCGC For the Basic Local Alignment
Tool (version 2.2.13). The siRNA duplexes were ATCTGA-3) and TaqMan probe (6-carboxyuorescein-5- Search Tool see http://jura.wi.
mit.edu/bioc/siRNAext/
synthesised by Dharmacon (Chicago, IL, USA) or CCCTTGCCACAATCT-minor groove binder non-
Integrated DNA Technologies BVBA (Leuven, Belgium). uorescent quencher-3) from Applied Biosystems (Foster
EK-1 was previously described.11 siRNAs for targeting the City, CA, USA) were specic for the ZEBOV glycoprotein
VP24 and VP35 genes were identied by screening in gene. Cycling conditions were reverse transcription at 50C
vitro for reduction in expression of the ZEBOV VP24 or for 20 min, and initial denaturation at 95C for 5 min;
ZEBOV VP35 viral transgene inserted in the psiCHECK2 followed by 45 cycles of denaturation at 95C for 5 s, and
dual-luciferase plasmid system (Promega, Madison, WI, annealing, synthesis, and single acquisition at 60C for
USA) under the control of the SV40 promoter in HepG2 20 s; and nal cooling at 40C for 30 s. Absolute
cells. 10 L of Lipofectamine 2000 (Invitrogen, Life quantication of viral gene expression was based on a viral
Technologies, Carlsbad, CA, USA) complexes containing RNA standard by use of the LC480 software (version 1.50).
075 g of plasmid and 10 L of siRNA (VP24-775, VP24-
978, VP24-1160, or a non-specic negative control siRNA 5 RACE assays
at 13 nmol/L, 25 nmol/L, 50 nmol/L, 100 nmol/L, RNA extraction and 5 RACE were done as described11
200 nmol/L, and 400 nmol/L; or VP35-219, VP35-349, except we used gene-specic primers (GSP) for cDNA
VP35-687, VP35-855, or a non-specic negative control production: EK-1 GSP (5-TTTGTGATTCGTCCTTTT

www.thelancet.com Vol 375 May 29, 2010 1897


Articles

ZEBOV target Sequence


EK-1 mod L polymerase Sense 5-GmUACGAAGCUmGUAUAmUAAAUU-3, antisense 5-UUUAmUAUACAGCUUCGmUACAA-3
VP24-775 VP24 Sense 5-GCUGAUUGACCAGUCUUUGAU-3, antisense 5-CAAAGACUGGUCAAUCAGCUG-3
VP24-978 VP24 Sense 5-ACGGAUUGUUGAGCAGUAUUG-3, antisense 5-AUACUGCUCAACAAUCCGUUG3
VP24-1160 VP24 Sense 5-UCCUCGACACGAAUGCAAAGU-3, antisense 5-UUUGCAUUCGUGUCGAGGAUC-3
VP24-1160 mod VP24 Sense 5-UCCmUCGACACGAAmUGCAAAGU-3, antisense 5-UUmUGCAUUCGUGUCmGAGmGAUC-3
VP35-219 VP35 Sense 5-GCGACAUCUUCUGUGAUAUUG-3, antisense 5-AUAUCACAGAAGAUGUCGCUU-3
VP35-349 VP35 Sense 5-GGAGGUAGUACAAACAUUGdTdT-3, antisense 5-CAAUGUUUGUACUACCUCCdTdT-3
VP35-687 VP35 Sense 5-GGGAGGCAUUCAACAAUCUAG-3, antisense 5-AGAUUGUUGAAUGCCUCCCUA-3
VP35-855 VP35 Sense 5-GCAACUCAUUGGACAUCAUUC-3, antisense 5-AUGAUGUCCAAUGAGUUGCUA-3
VP35-855 mod VP35 Sense 5-GCAACmUCAUUGmGrArCrAmUCAUUC-3, antisense 5-AUGAUmGUCCAAUGAmGUmUGCUA-3
Luc NA Sense 5-GAUUAUGUCCGGUUAUGUAAA-3, antisense 5-UACAUAACCGGACAUAAUCAU-3
Luc mod NA Sense 5-GAmUmUAmUGmUCCGGmUmUAmUGmUAAA-3, antisense 5-UACAmUAACCGGACAmUAAmUCAU-3

Sequences used in the non-human primate studies contain an m in front of the base that designates a 2-O-methyl modication (unmodied versions do not have any
2-O-methyl modied bases). mod=modied. Luc mod=modied luciferase. NA=not applicable.

Table 1: Targets and target sequences in Zaire Ebola virus (ZEBOV) of small interfering RNAs

GCAGT3-), VP24-1160 GSP (5-AGCAATTCTATGATG replaced every 3 days. To prime cells for dierentiation
TTGTCTTGGA-3), or VP35-855 GSP (5-CATCACTTT into those positive for CD11c, CD11b, and CD14, we
TGGTTTGGGTTACTT-3); and reverse-2 (Rev2) primers supplemented them with recombinant human interleukins
for the 5 RACE PCR: EK-1 Rev2 (5-TGAGAT 3 (20 ng/mL) and 6 (20 ng/mL; both R&D Systems,
GGTTTTGGTGTGGCATCT-3), VP24-1160 Rev2 (5-CCTG Minneapolis, MN, USA) on day 6. On day 10, cells were
GTTTTTGTAAGGGTGTCAACT-3), or VP35-855 Rev2 dierentiated into a mixed myeloid dendritic cell and
(5-TTTCTGGCAAGCTCGGGGAATGT-3). Sequencing macrophage culture by the addition of recombinant
primers (seq) were EK-1 3seq (5-AGCCATAACATAC human macrophage colony-stimulating factor (50 ng/mL)
CCTCAGT-3), VP24-1160 3seq (5-ATGAAAGCAGAGA and recombinant human granulocyte-macrophage colony-
TGTCAAG-3), or VP35-855 3seq (5-ATTAGGGCACATT stimulating factor (20 ng/mL; both, Peprotech).
GAGGAG-3). The predicted size of the RACE PCR product Cultures of dierentiated cells were incubated with
for EK-1 and VP24-1160 was 282 base pairs, and that for SNALPs containing uorescein isothiocyanate (FITC,
VP35-855 was 205 base pairs. Samples were processed 150 nmol/L)-labelled Luc mod for 4 h and then harvested,
from two separate transfections with similar results. washed with 2% fetal bovine serum in phosphate-buered
saline (PBS), and stained with uorescent-labelled
In-vitro immune stimulation assays antibodies (BD Biosciences, San Jose, CA, USA) against
Peripheral blood mononuclear cells (PBMCs) were cell phenotype markers (CD11c, CD11b, CD14, and CD34).
isolated from whole blood from healthy donors by use of Cells with uptake of SNALPs containing FITC-labelled
a standard Ficoll-Hypaque density centrifugation Luc mod were separated according to phenotype and
technique. Immune stimulation assays were done as analysed on a three-laser, eight-colour LSRII ow
previously described.13 Native (unmodied) or cytometer with FACSDiva software (version 6.0).
2-O-methyl-modied siRNAs (100 nmol/L, 200 nmol/L,
or 400 nmol/L) were cultured with PBMCs for 24 h. The Mouse studies
lower limit of quantication of interferon was Mouse studies were completed in accordance with the
156 pg/mL. Mean (SD) was calculated for three separate Canadian Council on Animal Care guidelines following
culture wells in one representative experiment. approval by the local animal care and use committee at
Tekmira Pharmaceuticals. Animal research was done in
Dierentiation and SNALP uptake in CD34-positive cells compliance with the animal welfare act, and other federal
Human cord blood CD34-positive stem cells (Stem Cell statutes and regulations relating to animals and
Technologies, Vancouver, BC, Canada) were cultured for experiments involving animals, and adheres to the
5 days in Iscoves modied Dulbeccos medium with principles stated in the Guide for the Care and Use of
Glutamax (Invitrogen), 20% BIT9500 serum substitute Laboratory Animals, National Research Council, 1996.
(Stem Cell Technologies), human LDL (40 g/mL; The facilities used are fully accredited by the Association
Calbiochem, San Diego, CA, USA), mercaptoethanol for Assessment and Accreditation of Laboratory Animal
(55 mol/L), human peptide deformylase-related tyrosine Care International.
kinase-3 ligand (100 ng/mL), human stem-cell factor 68-week-old female CD1 ICR mice (n=16; immuno-
(100 ng/mL), and human thrombopoietin (10 ng/mL; all stimulation studies [ four mice per group]), and 68-week-
four Peprotech, Rocky Hill, NJ, USA). Cell media were old male (n=20) and female B6C3F1 mice (n=20; tolerability

1898 www.thelancet.com Vol 375 May 29, 2010


Articles

A
120

100

80
Gene expression (%)

60

40

20

0
1 3 25 50 100 200 400 13 25 50 100 200 400 13 25 50 100 200 400 1 3 25 50 100 200 400
VP24-775 VP24-978 VP24-1160 Negative control siRNA
siRNA (nmol/L)
B
120

100

80
Gene expression (%)

60

40

20

0
08 40 200 08 40 200 08 40 200 08 40 200 08 40 200 0
VP35-219 VP35-349 VP35-687 VP35-855 Negative control Plasmid
siRNA only
siRNA (nmol/L)

Figure 1: Eect of small interfering RNAs (siRNAs) targeting virion protein genes VP24 (A) and VP35 (B) of Zaire Ebola virus (ZEBOV) expressed in a non-viral
plasmid-based system in HepG2 cells
Error bars represent SD of triplicate tissue culture wells.

studies [ve mice per group]) were quarantined and were done. Clinical signs and organ weights were
acclimatised for 1 week before the studies. SNALPs were monitored in all studies.
administered by bolus injection in the lateral tail vein in
02 mL PBS. In the immune stimulation studies, plasma Lipid encapsulation of siRNAs
and livers were harvested 4 h after mice were injected with siRNAs were encapsulated in SNALPs by the process of
SNALPs containing a cocktail of siRNAs (5 mg/kg). spontaneous vesicle formation as reported by Jes and
Analysis of concentrations of cytokines in plasma, and colleagues.16 ZEBOV siRNAs were formulated as a
interferon-induced protein with tetratricopeptide repeats cocktail in the same lipid particles. SNALPs consisted
(IFIT1) mRNA (upregulation of this mRNA in tissues is a of synthetic cholesterol (Sigma-Aldrich, St Louis,
more sensitive measure of localised interferon production MO, USA), dipalmitoylphosphatidylcholine (Avanti
than is plasma interferon protein ELISA assay14) in liver Polar Lipids, Alabaster, AL, USA), 3-N-[(-methoxy
was done as previously reported.15 The lower limit of poly(ethylene glycol)2000)carbamoyl]-1,2-dimyrestyloxy-
quantication of interferon or interleukin 6 with ELISA propylamine,17 and cationic 1,2-dilinoleyloxy-3-N,N-
was 156 pg/mL. As an initial assessment of the tolerability dimethylaminopropane.17 The resulting SNALPs were
in animals, we injected SNALPs into uninfected mice dialysed against PBS and lter sterilised through a
(20 mg/kg per day, 28 mg/kg per day, or 43 mg/kg per 02 m lter before use. Particle sizes for all batches
day for 7 days), and then euthanised them 48 h after the were 8185 nm (eg, the average size of SNALPs containing
nal dose. The cumulative doses (14 mg/kg, 20 mg/kg, ZEBOV SNALPs used in the non-human primate study 1
and 30 mg/kg) equalled and exceeded the expected dosing was 817 nm) with a low polydispersity value (<015),
intensity in the non-human primate ecacy model. Blood indicating particle homogeneity and no aggregation.
serum was analysed for clinical chemistry and activity of Typically 9095% of siRNAs were encapsulated within
alanine aminotransferase, aspartate aminotransferase, the lipid particles. The amount of endotoxin was less
and sorbitol dehydrogenase; and complete blood counts than 15 EU/mg siRNA for all batches.

www.thelancet.com Vol 375 May 29, 2010 1899


Articles

A B C
1200 4000
50249
3500
1000

Mean IFIT:GAPDH mRNA ratio


3000
Interleukin 6 (pg/mL)

Inteferon (pg/mL)
800
2500

600 2000

1500
400 086
1000 053
043
200
500

0 0
PBS Luc Luc ZEBOV PBS Luc Luc ZEBOV PBS Luc Luc ZEBOV
mod cocktail mod cocktail mod cocktail

D
500
450
400
350
Interferon (pg/mL)

300
250
200
150
100
50
0
PBS 100 200 400 100 200 400 100 200 400 100 200 400 100 200 400 100 200 400 100 200 400 100 200 400
Luc Luc mod EK-1 EK-1 mod VP24-1160 VP24-1160 mod VP35-855 VP35-855 mod
siRNA (nmol/L)

Figure 2: Eect of small interfering RNAs (siRNAs) on synthesis of interleukin 6 (A) and interferon (B), induction of interferon-induced protein with
tetratricopeptide repeats (IFIT1) mRNA in mice (C), and induction of interferon in human peripheral blood mononuclear cell cultures (D)
Data are mean (SD). Error bars represent SD. Luc=luciferase. PBS=phosphate-buered saline. Luc mod=modied luciferase. GAPDH=glyceraldehyde 3-phosphate
dehydrogenase.

Non-human primate studies Scientic, Holliston, MA, USA). After 7 days, the
We investigated the protective ecacy of our lead anti- animals were inoculated intramuscularly with a target
ZEBOV siRNAs in an established rhesus macaque model dose of 1000 plaque-forming units (pfu) of ZEBOV
of ZEBOV-induced haemorrhagic fever.18 A combination (Kikwit strain; actual dose 1100 pfu in study 1 and
of modied siRNAs that targeted the ZEBOV genes for 1325 pfu in study 2).
polymerase L (EK-1 mod), VP24 (VP24-1160 mod), and In the rst study, the pool of anti-ZEBOV siRNAs
VP35 (VP35-855 mod; table 1) were formulated in formulated in SNALPs (2 mg/kg total siRNA per dose,
SNALPs. The studies were done in biosafety-level-4 bolus intravenous infusion) was administered to three of
biocontainment at the US Army Medical Research four macaques 30 min after the ZEBOV challenge, and
Institute of Infectious Diseases and were approved by the the control animal was not given any treatment. The
US Army Medical Research Institute of Infectious three treated animals were given additional treatments of
Diseases laboratory animal care and use committee. the SNALP-formulated anti-ZEBOV siRNAs at the same
Nine healthy, lovirus-seronegative Chinese rhesus dose on days 1, 3, and 5 after the ZEBOV challenge, while
macaques (Macaca mulatta; 58 kg) were used for the the control animal was not given any treatment.
non-human primate studies. Hickman, Leonard, Broviac In the second study, we investigated whether increasing
central venous catheters (BARD Access Systems, Salt the frequency of treatments could improve outcome. The
Lake City, UT) were inserted into the macaques as pool of SNALP-formulated anti-ZEBOV siRNAs (2 mg/kg
previously described.19 The monkeys were then placed per dose, bolus intravenous infusion) was administered
in primate jackets (Lomir Biomedical, Malone, NY, to four of ve macaques 30 min after the ZEBOV
USA), returned to their cages, and tethered. A challenge, while the control animal was given an equal
continuous intravenous infusion of saline (2 mL/h) was dose of SNALP-formulated non-specic modied siRNAs
provided to all animals during the entire study with a (Luc mod) 30 min after the ZEBOV challenge. The
basic single syringe KDS100 infusion pump (KDS animals were given additional treatments of the SNALP-

1900 www.thelancet.com Vol 375 May 29, 2010


Articles

formulated anti-ZEBOV siRNAs or the SNALP-


A
formulated non-specic siRNA (control) on days 1, 2, 3,

Viral genome (plaque forming units per mL)


810
4, 5, and 6 after the ZEBOV challenge. Animals were
given physical examinations, and blood was collected in
610
tubes containing EDTA at the time of challenge and on
days 3, 6, 10, 14, 22, and 4043 after lovirus challenge.
410
Haematology and serum biochemistry
In the non-human primate studies, we used a laser-based 210
haematological analyser (Coulter Electronics, Hialeah,
FL, USA) to obtain total white blood cell counts, white
blood cell dierentials, red blood cell counts, platelet PBS Luc mod ZEBOV
counts, haematocrit values, total haemoglobin, mean cell siRNA in SNALP (50 nmol/L)
B
volume, mean corpuscular volume, and mean corpuscular Lane 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17
haemoglobin concentration. We used a Piccolo point-of- 400 bp
care blood analyser (Abaxis, Sunnyvale, CA, USA) to 300 bp 282 bp
measure concentrations of albumin, amylase, alanine
200 bp 205 bp
aminotransferase, aspartate aminotransferase, alkaline
phosphatase, glutamyltransferase, glucose, cholesterol,
100 bp
total protein, total bilirubin, blood urea nitrogen, and
creatinine in serum samples. EK-1 RACE VP24-1160 RACE VP35-855 RACE

Virus detection by plaque assay Figure 3: Rapid amplication of cDNA ends (RACE)-PCR of small interfering RNA (siRNA)-mediated cleavage
For virus titration, we used a conventional plaque assay of Zaire Ebola virus (ZEBOV) L polymerase, virion protein (VP) 24 (VP24), and VP35 mRNAs in ZEBOV-
infected Vero E6 cells
on Vero E6 cells from cell culture uids of blood samples Vero E6 cells were treated with stable nucleic acid-lipid particles (SNALPs) containing ZEBOV siRNA cocktail, modied
collected from rhesus monkeys as previously described.20 luciferase (Luc mod), or phosphate-buered saline (PBS). (B) Vero E6 cells were treated with SNALPs containing
EK-1-mod, VP24-1160-mod, VP35-855-mod, ZEBOV siRNA cocktail, Luc mod, or PBS. The order of samples for each
Role of the funding source RACE PCR shown in the gel is SNALPs containing PBS, single gene-specic siRNAs (EK-1-mod, VP24-1160-mod, or
VP35-855-mod), ZEBOV cocktail, and Luc mod. Lanes 1 and 17 are the 100 base pair (bp) ladder, lanes 25 are EK-1
The sponsor provided nancial support for the study but RACE PCR, lanes 710 are VP24-1160 RACE, and lanes 1215 are VP35-855 RACE. Lanes 6, 11, and 16 are empty.
had no role in study design, data collection, data analysis,
data interpretation, or writing of the report. All authors
had access to the primary data, take responsibility for
Cells positive for uptake of SNALPs (%)

53%
accuracy and completeness of data reporting, and had nal
responsibility for the decision to submit for publication. 39%
34%
Results
VP24-1160 was the most ecacious siRNA at silencing
ZEBOV VP24 gene in HepG2 cells, and VP35-855 was
most ecacious at silencing ZEBOV VP35 gene (gure 1).
These sequences, and EK-1,11 were selected as the siRNAs 0
of the ZEBOV SNALP cocktail. Since non-selective
chemical modication might attenuate the potency of the CD34+ CD14+ CD11b+ CD11c+
Cell marker
siRNA, we selectively modied lead siRNAs by substituting
2-O-methyl guanosine or uridines in both strands to Figure 4: Proportion of dierentiated cells with uptake of stable nucleic acid-lipid particles (SNALPs)
eliminate the immune stimulatory capacity of the siRNA containing uorescein-isothiocyanate-labelled modied luciferase small interfering RNAs
duplex,1315 and conrmed the gene silencing activity of the
modied versions using the in-vitro plasmid-based high levels of IFIT1 mRNA (1169-fold greater than the
ZEBOV gene expression system (data not shown). PBS control; gure 2). No dierences were noted in
SNALPs containing modied ZEBOV siRNAs did not clinical signs and organ weights of mice treated with
induce interferon or interleukin 6 in the plasma of SNALPs containing siRNAs or PBS. Native (chemically
mice, whereas SNALPs containing chemically unmodied unmodied) versions of Luc, EK-1, VP24-1160, and VP35-
Luc siRNA induced high concentrations of both plasma 855 siRNAs also induced high levels of interferon in
cytokines (gure 2A and B). Analysis of liver IFIT1 cultures of human PBMCs, whereas the 2-O-methyl-
mRNA showed no dierences between SNALPs modied versions of siRNAs at up to 400 nmol/L did not
containing PBS (negative control) and those containing induce any interferon (gure 2).
the 2-O-methyl-modied Luc or ZEBOV siRNA cocktail, Figure 3A shows that SNALPs containing ZEBOV
whereas the SNALPs containing unmodied Luc induced siRNAs substantially reduced ZEBOV produced in

www.thelancet.com Vol 375 May 29, 2010 1901


Articles

A
Alanine aminotransferase
Aspartate aminotransferase
Sorbitol dehydrogenase
Enzyme activity (IU/mL)

70
43 51 45 43
37 30 37 35 41 27 28 28
40
28
37
19 21 26 23 24 24 20 22

PBS siRNAs siRNAs siRNAs PBS siRNAs siRNAs siRNAs


20 mg/kg 285 mg/kg 43 mg/kg 20 mg/kg 285 mg/kg 43 mg/kg
Male Female
SNALPs
B
6 White blood cells
Neutrophils
5 Lymphocytes

4
Cells (10 per mL)

0
PBS siRNAs siRNAs siRNAs PBS siRNAs siRNAs siRNAs
20 mg/kg 285 mg/kg 43 mg/kg 20 mg/kg 285 mg/kg 43 mg/kg
Male Female
SNALPs

Figure 5: Eect of daily administration of stable nucleic acid-lipid particles (SNALPs) containing Zaire Ebola virus (ZEBOV) small interfering RNAs on activities
(siRNAs) of alanine aminotransferase, aspartate aminotransferase, and sorbitol dehydrogenase (A), and blood cell counts (B) in mice
PBS=phosphate-buered saline.

supernatants of Vero E6 cells 48 h after infection. myeloid cells that were positive for CD14, CD11b, and
Figure 3B shows that SNALPs containing EK-1-mod, CD11c (gure 4). These data showed that SNALPs were
VP35-855-mod, or VP24-1160-mod induced specic actively taken up by the reticuloendothelial cell
mRNA cleavage only of their target mRNA in the cells, populations most relevant to EBOV infection.
whereas all three PCR products of the correct predicted In the tolerability studies, activities of alanine
size were present in cells treated with SNALPs containing aminotransferase, aspartate aminotransferase, and sorbitol
the ZEBOV cocktail of siRNAs (gure 3B). No 5RACE dehydrogenase remained unchanged 48 h after the mice
PCR products of the predicted size were noted in cells were given the nal dose of SNALPs containing ZEBOV
treated with SNALPs containing PBS or Luc mod siRNAs, even at the highest cumulative dose (gure 5A).
(gure 3B), further showing the specicity of the assay. Complete blood cell and dierential counts were also
The sequences of the 5RACE PCR products (data not unaected at the doses tested in the mice (gure 5B).
shown) corresponded to the specic cleavage products of All three macaques given four treatments of SNALPs
polymerase L protein, VP24, and VP35 mRNAs, thereby containing pooled siRNAs, and the control, developed
conrming the specic mechanism of RNA interference. clinical symptoms that were consistent with ZEBOV-
To show that our anti-ZEBOV siRNAs targeted induced haemorrhagic fever by day 6 (table 2). The
populations of cells that were relevant to lovirus control animal succumbed to ZEBOV infection before
infection, we assessed the uptake of the SNALPs in livers blood collection on day 6 (gure 6). One treated animal
of mice and in human CD34-derived myeloid cell (number 3) developed a high ZEBOV viraemia on
cultures. The SNALPs enabled entry of siRNAs into cells day 6 (table 3) and succumbed on day 10 (gure 6).
of the reticuloendothelial system as shown by the uptake ZEBOV was detected in the plasma of one of the
of uorescent SNALPs into Kuper cells in the murine remaining treated animals (number 2) on day 6, but we
liver (data not shown), and into subsets of human were unable to detect ZEBOV in the plasma of the other

1902 www.thelancet.com Vol 375 May 29, 2010


Articles

Adverse events on days 135 after ZEBOV challenge Status

Study 1
Animal 1 Anorexia (days 79), lymphopenia (day 6), >5-fold increase in AST (day 10) Survived
Animal 2 Fever (day 6), mild rash (days 8,11, and 12), moderate rash (days 9 and 10), depression (days 711), anorexia (days 7-11), diarrhoea (day 12), Survived
lymphopenia (days 6 and 14), thrombocytopenia (day 6), 23-fold increase in ALP (day 10), 23-fold increase in ALT (day 10), 23-fold increase in AST
(day 6), >5-fold increase in AST (day 10), >5-fold increase in GGT (day 10)
Animal 3 Mild rash (days 610), depression (days 6-10), anorexia (days 610), bleeding at venepuncture site (day 10), recumbency (day 10), thrombocytopenia Died on day 10
(day 6), 23-fold increase in ALP (day 6), >5-fold increase in ALP (day 10), 45-fold increase in ALT (day 10), 23-fold increase in AST (day 3), >5-fold
increase in AST (days 6 and 10), >5-fold increase in BUN (day 10), >5-fold increase in creatinine (day 10), >5-fold increase in GGT (day 10)
Control 1* Mild rash, anorexia, and depression on day 5 Died day 6
Study 2
Animal 4 Thrombocytopenia (days 6 and 10), 45-fold increase in AST (day 6) Survived
Animal 5 >5-fold increase in AST (day 6), 23-fold increase in AST (day 10) Survived
Animal 6 Fever (day 10), lymphopenia (day 6), thrombocytopenia (days 6, 10, and 14), 23-fold increase in AST (day 10) Survived
Animal 7 Fever (day 10), lymphopenia (day 6), thrombocytopenia (day 6), >5-fold increase in AST (day 10) Survived
Control 2 Fever (day 6), moderate rash (day 10), recumbency (day 10), thrombocytopenia (day 6) 23-fold increase in ALT (day 6), >5-fold increase in ALT Died on day 10
(day 10), >5-fold increase in AST (days 6 and 10), >5-fold increase in BUN (day 10), 23-fold increase in creatinine (day 10)

Fever was dened as a temperature that was 25F greater than baseline temperature or at least 15F greater than baseline temperature, and at least 1035F. Mild rash was dened as focal petechiae covering less
than 10% of skin. Moderate rash was dened as petechiae covering between 10% and 40% of skin. Severe rash was dened as petechiae or ecchymoses covering more than 40% of skin. Lymphopenia was dened
as at least a 35% reduction in numbers of lymphocytes; and thrombocytopenia was dened as at least a 35% reduction in numbers of platelets. AST=aspartate aminotransferase. ALP=alkaline phosphatase.
ALT=alanine aminotransferase. GGT= glutamyltransferase. BUN=blood urea nitrogen. *Not given any siRNAs. Given seven treatments with non-specic siRNAs.

Table 2: Macaques given four (study 1) or seven treatments (study 2) with stable nucleic acid-lipid particles containing Zaire Ebola virus (ZEBOV) small interfering RNAs (siRNAs) after
ZEBOV challenge

remaining treated animal (number 1; table 3). Both of was greater than 45 log10 pfu/mL, whereas animals
these animals survived. Seven doses of SNALPs treated therapeutically survived when the viraemia did
containing pooled siRNAs were more eective because not reach this level (Geisbert TW, unpublished).19,21 In the
clinical symptoms of ZEBOV infection in the treated current study, viraemia never exceeded 24 log10 pfu/mL
macaques were less severe than in those given four doses in any surviving animal.
(table 2). The control animal injected with SNALPs The development of treatments for EBOV-induced
containing Luc mod succumbed to haemorrhagic fever haemorrhagic fever has been slow and no previous
on day 10, whereas all four animals given the pooled anti- candidate treatment has shown complete protection
ZEBOV siRNAs survived (gure 6). ZEBOV was detected against ZEBOV haemorrhagic fever in non-human
in the plasma of the second control animal on days 3, 6, primates. Some success has been achieved with
and 10 (table 3). Low levels of ZEBOV were detected on interventions that mitigate the coagulation disorders
day 6 in the plasma of all four macaques injected with that characterise EBOV infection.19,21 We protected 50%
SNALPs containing ZEBOV siRNAs, and on day 10 in of non-human primates against ZEBOV by
one of these animals (animal 6), whereas we were unable administering a live-attenuated recombinant vesicular
to detect ZEBOV in any of these animals on day 14 (table 3). stomatitis virus vaccine vector that expressed the
Peak viraemia in surviving siRNA-treated macaques ZEBOV glycoprotein shortly after a lethal ZEBOV
never exceeded 24 log10 pfu/mL (table 3). challenge.22 Several new postexposure treatments based
Clinical assessments also showed that this aggressive on siRNA11 and antisense oligomers23,24 have shown
SNALP treatment regimen was well tolerated in promising results in rodent models, but there have
macaques with only minor changes in liver enzymes been no reports of the assessment of either treatment
(alanine aminotransferase <2-fold increase, aspartate strategy after EBOV challenge in non-human primates,
amino-transferase <6-fold increase), possibly related to which more faithfully reproduce human EBOV
viral infection. All six animals that survived the ZEBOV infections than do other animal models.1,18 This model
challenge in the two studies were healthy on day 40 and was rapid and uniformly lethal (death in all 21 naive
were euthanised on days 4043. rhesus macaques challenged with the same ZEBOV
seed stock by the same dose and route as in the present
Discussion study; Geisbert TW, unpublished) in which animals
Seven treatments with anti-ZEBOV siRNAs inhibited the typically succumbed 610 days after challenge with the
replication of the virus and completely protected rhesus virus (mean time to death 83 days). The ZEBOV target
monkeys against death from haemorrhagic fever. In in the siRNA postexposure treatment study in
previous studies, ZEBOV-infected rhesus monkeys guineapigs was the L polymerase gene, and treatment
succumbed when viraemia on days 610 after exposure initiated 1 h after ZEBOV challenge completely

www.thelancet.com Vol 375 May 29, 2010 1903


Articles

100 Plasma viral load (log10 plaque-froming units)

Day 3 Day 6 Day 10 Day 14


80
Study 1
Animal 1 0 0 0 0
Survival (%)

60
Animal 2 0 0 22 0
40 Animal 3 0 37 68 NA
Control 1* 0 NA NA NA
Study 1
20 Study 2 Study 2
Control
Animal 4 0 20 0 0
0 Animal 5 0 24 0 0
0 5 10 15 20 25 30
Days after challenge Animal 6 0 20 21 0
Animal 7 0 21 0 0
Figure 6: Survival curves for Zaire Ebola virus (ZEBOV)-infected rhesus Control 2 0 41 67 NA
macaques treated after challenge with stable nucleic acid-lipid particles
containing ZEBOV small interfering RNAs NA=not applicable. *Not given any siRNAs. Given seven treatments with non-
Study 1: four postexposure treatments. Study 2: seven post exposure treatments. specic siRNAs.

Table 3: Plasma viral load in rhesus monkeys infected with Zaire Ebola
protected all animals.11 Targeting only VP35 completely virus (ZEBOV) and given four (study 1) and seven (study 2)
protected mice against ZEBOV challenge when the postexposure treatments with anti-ZEBOV small interfering RNAs
antisense oligomers were administered 24 h after (siRNAs)
ZEBOV exposure.23 Targeting a combination of the
L polymerase, VP24, and VP35 genes also completely components (MacLachlan I, unpublished). Preliminary
protected mice and incompletely protected guineapigs tolerability studies in mice showed that seven daily
when given 24 h after ZEBOV challenge.24 doses of up to 43 mg/kg (300 mg/kg cumulative) of
Although RNA-interference-mediated treatment SNALPs containing ZEBOV siRNAs were well tolerated,
strategies show potential for combating EBOV corroborating the lack of hepatotoxicity (measured as
infections, systemic administration of synthetically increases in liver enzymes such as alanine amino-
manufactured siRNA duplexes can activate the innate transferase and aspartate aminotransferase) in the
immune response, inducing high levels of inammatory treated macaques that survived infection.
cytokines such as tumour necrosis factor , interleukin 6, Our investigation was designed as a proof-of-concept
and interferons, particularly interferon which might study. The rhesus macaque model that we used
contribute to antiviral activity in vivo.2527 O-target represents a worse-case scenario such as an accidental
eects can be toxic to the host and also confound needle stick exposure of a laboratory worker or rst
interpretation of preclinical data. siRNAs must therefore responder to a high infectious dose of ZEBOV, which
be modied to prevent detrimental immune activation has occurred several times during the past 5 years.2830
in vivo, particularly because immunostimulation in ZEBOV infection in human beings normally
human beings is much more toxic than in rodents.14,25 progresses slower than in macaques with case fatality
We therefore tested immune stimulatory activity of rates in man ranging from 7090%,1 suggesting that the
SNALPs containing anti-ZEBOV siRNAs in mice and in therapeutic window could be larger than in
human PBMC cultures. The ZEBOV cocktail was not experimentally infected macaques. Nonetheless, the
immune stimulatory at the limit of sensitivity of the focus in future studies will need to be on investigation
assays used. These data and the 5RACE PCR data of whether anti-ZEBOV siRNAs will be benecial if
suggest that the antiviral eects in non-human primates administered at the onset of symptoms. Here, we show
are the result of specic RNA interference in a substantial progress in the treatment of ZEBOV
reticuloendothelial cells and not due to immune infections in non-human primates compared with
stimulation or other o-target eects. Additionally, the previously described postexposure methods that
death of the macaque given control Luc mod on day 10, aorded only partial protection of non-human primates
which was within the normal range of expected time to against ZEBOV.19,21,22 To progress to license, studies will
death for historical untreated controls, also suggests be done in guineapig and non-human primate models
For more on the Code of Federal that the ZEBOV-mediated protection was specic to the of ZEBOV infection in accordance with the US Food
Regulations see http://www. siRNAs and not related to o-target eects. When the and Drug Administrations guidelines for the
accessdata.fda.gov/scripts/cdrh/
immune toxicities associated with the siRNA component assessment of treatments for Centers for Disease
cfdocs/cfcfr/CFRSearch.cfm?CFR
Part=314&showFR=1&subpartN of SNALPs were abrogated through the use of selective Control and Prevention category A threats (number
ode=21:5.0.1.1.4.9 chemical modication, the major remaining toxicity in 21CFR314.610). Additionally, studies will need to be
mouse and non-human primate models was done to obtain data about pharmacology, dosing, and
hepatotoxicity associated with the SNALP lipid toxicology in uninfected non-human primates.

1904 www.thelancet.com Vol 375 May 29, 2010


Articles

Contributors 12 Grimm D, Kay M. Combinatorial RNAi: a winning strategy for the


TWG, ACHL, MR, AJ, LEH, and IM conceived and designed the race against evolving targets? Mol Ther 2007; 15: 87888.
experiments. TWG wrote the research programme plan and the animal 13 Judge AD, Bola G, Lee ACH, MacLachlan I. Design of
protocol for the studies. LEH wrote modications for the animal noninammatory synthetic siRNA mediating potent gene silencing
protocol. TWG and LEH selected the ZEBOV L polymerase as a target. in vivo. Mol Ther 2006; 13: 494505.
TWG, ACHL, VS, MR, AJ, LEH, and IM selected ZEBOV VP24 and 14 Robbins MA, Judge AD, MacLachlan I. siRNA and innate
VP35 as targets. TWG co-designed the L polymerase siRNA, and ACHL immunity. Oligonucleotides 2009; 19: 89101.
and VS co-designed the VP24 and VP35 siRNAs. ACHL, VS, and AJ 15 Judge A, Robbins M, Tavakoli I, et al. Conrming the
designed the modications to the siRNAs. VS and ANH did the in vitro RNAi-mediated mechanism of action of siRNA-based cancer
siRNA assays. JBG and JCJ did the Ebola challenge experiments in non- therapeutics in mice. J Clin Invest 2009; 119: 66173.
human primates and the clinical pathology assays. JBG did the Ebola 16 Jes LB, Palmer LR, Ambegia EG, Giesbrecht C, Ewanick S,
virus infectivity assays. SDJ did the immunology and uptake assays. MR MacLachlan I. A scalable, extrusion-free method for ecient
liposomal encapsulation of plasmid DNA. Pharm Res 2005;
and IT did the 5RACE assays. TWG, ACHL, MR, JBG, ANH, JCJ, AJ,
22: 36272.
LEH, and IM analysed the data. TWG and MR co-wrote the report. LEH
17 Heyes J, Palmer L, Bremner K, MacLachlan I. Cationic lipid
and IM edited the report. All authors have seen and approved the nal
saturation inuences intracellular delivery of encapsulated nucleic
version of the report.
acids. J Control Release 2005; 107: 27687.
Conicts of interest 18 Geisbert TW, Jahrling PB, Larsen T, Davis KJ, Hensley LE. Filovirus
TWG, ACHL, MR, VS, AJ, LEH, and IM claim intellectual property pathogenesis in nonhuman primates. In: Klenk HD, Feldmann H,
regarding RNA interference for the treatment of loviral infections. ACHL, eds. Ebola and Marburg viruses: molecular and cellular biology.
MR, VS, SDJ, IT, AJ, and IM are employees of Tekmira Pharmaceuticals. Norfolk: Horizon Bioscience, 2004: 20338.
JCJ, ANH, and JBG declare that they have no conicts of interest. 19 Hensley LE, Stevens EL, Yan SB, et al. Recombinant human
activated protein C for the postexposure treatment of Ebola
Acknowledgments hemorrhagic fever. J Infect Dis 2007; 196 (suppl 2): S39099.
Work on loviruses at US Army Medical Research Institute of Infectious 20 Jahrling PB, Geisbert TW, Geisbert JB, et al. Evaluation of immune
Diseases was funded by the Defense Threat Reduction Agency (project globulin and recombinant interferon -2b for treatment of
number 04-4-7J-012). Opinions, interpretations, conclusions, and experimental Ebola virus infections. J Infect Dis 1999;
recommendations are those of the authors and are not necessarily 179 (suppl 1): S22434.
endorsed by Boston University or the US Army. 21 Geisbert TW, Hensley LE, Jahrling PB, et al. Treatment of Ebola
virus infection with a recombinant inhibitor of factor VIIa/tissue
References
factor: a study in rhesus monkeys. Lancet 2003; 362: 195358.
1 Sanchez A, Geisbert TW, Feldmann H. Filoviridae: Marburg and
Ebola viruses. In: Knipe DM, Howley PM, eds. Fields virology. 22 Feldmann H, Jones SM, Daddario-Dicaprio KM, et al. Eective
Philadelphia: Lippincott Williams and Wilkins, 2006: 140948. post-exposure treatment of Ebola infection. PLoS Pathog 2007; 3: e2.
2 Towner JS, Sealy TK, Khristova ML, et al. Newly discovered Ebola 23 Enterlein S, Wareld KL, Swenson DL, et al. VP35 knockdown
virus associated with hemorrhagic fever outbreak in Uganda. inhibits Ebola virus amplication and protects against lethal
PLoS Pathog 2008; 4: e1000212. infection in mice. Antimicrob Agents Chemother 2006; 50: 98493.
3 Sanchez A, Kiley MP, Holloway BP, Auperin DD. Sequence analysis 24 Wareld KL, Swenson DL, Olinger GG, et al. Gene-specic
of the Ebola virus genome: organization, genetic elements, and countermeasures against Ebola virus based on antisense
comparison with the genome of Marburg virus. Virus Res 1993; phosphorodiamidate morpholino oligomers. PLoS Pathog 2006; 2: e1.
29: 21540. 25 Robbins M, Judge A, Ambegia E, et al. Misinterpreting the
4 Basler CF, Mikulasova A, Martinez-Sobrido L, et al. The Ebola virus therapeutic eects of siRNA caused by immune stimulation.
VP35 protein inhibits activation of interferon regulatory factor 3. Hum Gene Ther 2008; 19: 99199.
J Virol 2003; 77: 794556. 26 Hornung V, Guenthner-Biller M, Bourquin C, et al. Sequence-
5 Prins KC, Delpeut S, Leung DW, et al. Mutations abrogating VP35 specic potent induction of IFN-alpha by short interfering RNA in
interaction with double stranded RNA render Ebola virus avirulent plasmacytoid dendritic cells through TLR7. Nat Med 2005;
in guinea pigs. J Virol 2010; 84: 300415. 11: 263270.
6 Reid SP, Leung DW, Hartman AL, et al. Ebola virus VP24 binds 27 Judge AD, Sood V, Shaw JR, Fang D, McClintock K, MacLachlan I.
karyopherin alpha1 and blocks STAT1 nuclear accumulation. J Virol Sequence-dependent stimulation of the mammalian innate
2006; 80: 515667. immune response by synthetic siRNA. Nat Biotechnol 2005;
23: 45762.
7 Ebihara H, Takada A, Kobasa D, et al. Molecular determinants of
Ebola virus virulence in mice. PLoS Pathog 2006; 2: e73. 28 Kortepeter MG, Martin JW, Rusnak JM, et al. Managing potential
laboratory exposure to Ebola virus by using a patient
8 Volchkov VE, Chepurnov AA, Volchkova VA, Ternovoj VA,
biocontainment care unit. Emerg Infect Dis 2008; 14: 88187.
Klenk HD. Molecular characterization of guinea pig-adapted
variants of Ebola virus. Virology 2000; 277: 14755. 29 International Society for Infectious Diseases. Ebolavirus,
needlestick injury Germany: (Hamburg). http://www.
9 Yokota T, Iijima S, Kubodera T, et al. Ecient regulation of viral
promedmail.org/pls/apex/f?p=2400:1001:::NO::F2400_P1001_
replication by siRNA in a non-human primate surrogate model for
BACK_PAGE,F2400_P1001_PUB_MAIL_ID:1000%2C76634
hepatitis C. Biochem Biophys Res Commun 2007; 361: 294300.
(accessed April 29, 2010).
10 Tang Q, Li B, Woodle M, Lu PY. Application if siRNA against SARS
30 International Society for Infectious Diseases. Ebola, lab accident
in the rhesus macaque model. Methods Mol Biol 2008; 442: 13958.
death Russia (Siberia). http://www.promedmail.org/pls/apex/
11 Geisbert TW, Hensley LE, Kagan E, et al. Postexposure protection of f?p=2400:1001:::NO::F2400_P1001_BACK_PAGE,F2400_P1001_
guinea pigs against a lethal Ebola virus challenge is conferred by PUB_MAIL_ID:1000%2C25465 (accessed April 29, 2010).
RNA interference. J Infect Dis 2006; 193: 165057.

www.thelancet.com Vol 375 May 29, 2010 1905

Potrebbero piacerti anche