Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amy C. H. Lee is active.

Publication


Featured researches published by Amy C. H. Lee.


Nature | 2006

RNAi-mediated gene silencing in non-human primates.

Tracy Zimmermann; Amy C. H. Lee; Akin Akinc; Birgit Bramlage; David Bumcrot; Matthew N. Fedoruk; Jens Harborth; James Heyes; Lloyd Jeffs; Matthias John; Adam Judge; Kieu Lam; Kevin McClintock; Lubomir Nechev; Lorne R. Palmer; Timothy Racie; Ingo Röhl; Stephan Seiffert; Sumi Shanmugam; Vandana Sood; Jürgen Soutschek; Ivanka Toudjarska; Amanda J. Wheat; Ed Yaworski; William Zedalis; Victor Koteliansky; Muthiah Manoharan; Hans-Peter Vornlocher; Ian Maclachlan

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 medicines. Although there are numerous examples of in vivo silencing of target genes after local delivery of small interfering RNAs (siRNAs), there remain only a few reports of RNAi-mediated silencing in response to systemic delivery of siRNA, and there are no reports of systemic efficacy in non-rodent species. Here we show that siRNAs, when delivered systemically in a liposomal formulation, 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 kg-1. 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 demonstrating 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 therapeutics as a potential new class of drugs.


The Lancet | 2010

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 Hensley; Ian Maclachlan

Summary Background We previously showed that small interfering RNAs (siRNAs) targeting the Zaire Ebola virus (ZEBOV) RNA polymerase L protein formulated in stable nucleic acid-lipid particles (SNALPs) completely protected guineapigs when administered shortly after a lethal ZEBOV challenge. Although rodent models of ZEBOV infection are useful for screening prospective countermeasures, they are frequently not useful for prediction of efficacy in the more stringent non-human primate models. We therefore assessed the efficacy of modified non-immunostimulatory siRNAs in a uniformly lethal non-human primate model of ZEBOV haemorrhagic fever. Methods A combination of modified siRNAs targeting the ZEBOV L polymerase (EK-1 mod), viral protein (VP) 24 (VP24-1160 mod), and VP35 (VP35-855 mod) were formulated in SNALPs. A group of macaques (n=3) was given these pooled anti-ZEBOV siRNAs (2 mg/kg per dose, bolus intravenous infusion) after 30 min, and on days 1, 3, and 5 after challenge with ZEBOV. A second group of macaques (n=4) was given the pooled anti-ZEBOV siRNAs after 30 min, and on days 1, 2, 3, 4, 5, and 6 after challenge with ZEBOV. Findings Two (66%) of three rhesus monkeys given four postexposure treatments of the pooled anti-ZEBOV siRNAs were protected from lethal ZEBOV infection, whereas all macaques given seven postexposure treatments were protected. The treatment regimen in the second study was well tolerated with minor changes in liver enzymes that might have been related to viral infection. Interpretation This complete postexposure protection against ZEBOV in non-human primates provides a model for the treatment of ZEBOV-induced haemorrhagic fever. These data show the potential of RNA interference as an effective postexposure treatment strategy for people infected with Ebola virus, and suggest that this strategy might also be useful for treatment of other emerging viral infections. Funding Defense Threat Reduction Agency.


The Journal of Infectious Diseases | 2006

Postexposure Protection of Guinea Pigs against a Lethal Ebola Virus Challenge Is Conferred by RNA Interference

Thomas W. Geisbert; Lisa E. Hensley; Elliott Kagan; Erik Z. Yu; Joan B. Geisbert; Kathleen M. Daddario-DiCaprio; Elizabeth A. Fritz; Peter B. Jahrling; Kevin McClintock; Janet R. Phelps; Amy C. H. Lee; Adam Judge; Lloyd Jeffs; Ian Maclachlan

Abstract BackgroundEbola virus (EBOV) infection causes a frequently fatal hemorrhagic fever (HF) that is refractory to treatment with currently available antiviral therapeutics. RNA interference represents a powerful, naturally occurring biological strategy for the inhibition of gene expression and has demonstrated utility in the inhibition of viral replication. Here, we describe the development of a potential therapy for EBOV infection that is based on small interfering RNAs (siRNAs) MethodsFour siRNAs targeting the polymerase (L) gene of the Zaire species of EBOV (ZEBOV) were either complexed with polyethylenimine (PEI) or formulated in stable nucleic acid–lipid particles (SNALPs). Guinea pigs were treated with these siRNAs either before or after lethal ZEBOV challenge ResultsTreatment of guinea pigs with a pool of the L gene–specific siRNAs delivered by PEI polyplexes reduced plasma viremia levels and partially protected the animals from death when administered shortly before the ZEBOV challenge. Evaluation of the same pool of siRNAs delivered using SNALPs proved that this system was more efficacious, as it completely protected guinea pigs against viremia and death when administered shortly after the ZEBOV challenge. Additional experiments showed that 1 of the 4 siRNAs alone could completely protect guinea pigs from a lethal ZEBOV challenge ConclusionsFurther development of this technology has the potential to yield effective treatments for EBOV HF as well as for diseases caused by other agents that are considered to be biological threats


Science Translational Medicine | 2014

Marburg virus infection in nonhuman primates: Therapeutic treatment by lipid-encapsulated siRNA.

Emily P. Thi; Chad E. Mire; Raul Ursic-Bedoya; Joan B. Geisbert; Amy C. H. Lee; Krystle N. Agans; Marjorie Robbins; Daniel J. Deer; Karla A. Fenton; Ian Maclachlan; Thomas W. Geisbert

Lipid nanoparticle delivery of anti-MARV nucleoprotein–targeting small interfering RNA can treat Marburg virus in nonhuman primates after symptom onset. Medicating Marburg Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever; however, there are currently no vaccines or drugs approved for human use to treat these devastating infections. Thi et al. now report that a lipid-encapsulated siRNA can treat MARV in nonhuman primates after symptom onset. They use lipid nanoparticles to deliver siRNA targeting the MARV nucleoprotein to treated animals at various time points after virus exposure. All animals that received the therapy survived MARV infection, including those that were treated 3 days after infection—a stage when animals are viremic and demonstrate the first clinical signs of disease. Therefore, this approach holds promise as a strategy to treat filovirus infection in humans. Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever (HF) in humans and nonhuman primates with mortality rates up to 90%. There are no vaccines or drugs approved for human use, and no postexposure treatment has completely protected nonhuman primates against MARV-Angola, the strain associated with the highest rate of mortality in naturally occurring human outbreaks. Studies performed with other MARV strains assessed candidate treatments at times shortly after virus exposure, before signs of disease are detectable. We assessed the efficacy of lipid nanoparticle (LNP) delivery of anti-MARV nucleoprotein (NP)–targeting small interfering RNA (siRNA) at several time points after virus exposure, including after the onset of detectable disease in a uniformly lethal nonhuman primate model of MARV-Angola HF. Twenty-one rhesus monkeys were challenged with a lethal dose of MARV-Angola. Sixteen of these animals were treated with LNP containing anti-MARV NP siRNA beginning at 30 to 45 min, 1 day, 2 days, or 3 days after virus challenge. All 16 macaques that received LNP-encapsulated anti-MARV NP siRNA survived infection, whereas the untreated or mock-treated control subjects succumbed to disease between days 7 and 9 after infection. These results represent the successful demonstration of therapeutic anti–MARV-Angola efficacy in nonhuman primates and highlight the substantial impact of an LNP-delivered siRNA therapeutic as a countermeasure against this highly lethal human disease.


Archive | 2005

siRNA silencing of apolipoprotein B

Ian Maclachlan; Lloyd Jeffs; Adam Judge; Amy C. H. Lee; Lorne R. Palmer; Vandana Sood


Archive | 2010

Compositions and methods for silencing apolipoprotein B

Amy C. H. Lee; Adam Judge; Marjorie Robbins; Ed Yaworski; Ian Maclachlan


Archive | 2010

Compositions and methods for silencing apolipoprotein C-III expression

Marcia Macdonald; Amy C. H. Lee; Ian Maclachlan


Archive | 2016

Compositions and methods for silencing hepatitis b virus gene expression

Jennifer L. Cross; Ammen P. Dhillon; Amy C. H. Lee; Ian Maclachlan; Nicholas M. Snead; Emily P. Thi


Archive | 2012

Compositions and methods for silencing aldehyde dehydrogenase

Ian Maclachlan; Amy C. H. Lee


Archive | 2016

Compositions et méthodes de silençage de l'expression du gène du virus de l'hépatite b

Amy C. H. Lee; Nicholas M. Snead; Emily P. Thi

Collaboration


Dive into the Amy C. H. Lee's collaboration.

Top Co-Authors

Avatar

Ian Maclachlan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Adam Judge

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Ed Yaworski

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Vandana Sood

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Joan B. Geisbert

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Lloyd Jeffs

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Thomas W. Geisbert

United States Department of the Army

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akin Akinc

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Anna N. Honko

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge