Network


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

Hotspot


Dive into the research topics where Ying K. Tam is active.

Publication


Featured researches published by Ying K. Tam.


Nature Biotechnology | 2010

Rational design of cationic lipids for siRNA delivery

Sean C. Semple; Akin Akinc; Jianxin Chen; Ammen Sandhu; Barbara L. Mui; Connie K Cho; Dinah Sah; Derrick Stebbing; Erin J Crosley; Ed Yaworski; Ismail Hafez; J. Robert Dorkin; June Qin; Kieu Lam; Kallanthottathil G. Rajeev; Kim F. Wong; Lloyd Jeffs; Lubomir Nechev; Merete L. Eisenhardt; Muthusamy Jayaraman; Mikameh Kazem; Martin Maier; Masuna Srinivasulu; Michael J Weinstein; Qingmin Chen; Rene Alvarez; Scott Barros; Soma De; Sandra K. Klimuk; Todd Borland

We adopted a rational approach to design cationic lipids for use in formulations to deliver small interfering RNA (siRNA). Starting with the ionizable cationic lipid 1,2-dilinoleyloxy-3-dimethylaminopropane (DLinDMA), a key lipid component of stable nucleic acid lipid particles (SNALP) as a benchmark, we used the proposed in vivo mechanism of action of ionizable cationic lipids to guide the design of DLinDMA-based lipids with superior delivery capacity. The best-performing lipid recovered after screening (DLin-KC2-DMA) was formulated and characterized in SNALP and demonstrated to have in vivo activity at siRNA doses as low as 0.01 mg/kg in rodents and 0.1 mg/kg in nonhuman primates. To our knowledge, this represents a substantial improvement over previous reports of in vivo endogenous hepatic gene silencing.


Angewandte Chemie | 2012

Maximizing the Potency of siRNA Lipid Nanoparticles for Hepatic Gene Silencing In Vivo

Muthusamy Jayaraman; Steven M. Ansell; Barbara L. Mui; Ying K. Tam; Jianxin Chen; Xinyao Du; David Butler; Laxman Eltepu; Shigeo Matsuda; Jayaprakash K. Narayanannair; Kallanthottathil G. Rajeev; Ismail Hafez; Akin Akinc; Martin Maier; Mark Tracy; Pieter R. Cullis; Thomas D. Madden; Muthiah Manoharan; Michael J. Hope

Special (lipid) delivery: The role of the ionizable lipid pK(a) in the in vivo delivery of siRNA by lipid nanoparticles has been studied with a large number of head group modifications to the lipids. A tight correlation between the lipid pK(a) value and silencing of the mouse FVII gene (FVII ED(50) ) was found, with an optimal pK(a) range of 6.2-6.5. The most potent cationic lipid from this study has ED(50) levels around 0.005 mg kg(-1) in mice and less than 0.03 mg kg(-1) in non-human primates.


Molecular therapy. Nucleic acids | 2012

Microfluidic Synthesis of Highly Potent Limit-size Lipid Nanoparticles for In Vivo Delivery of siRNA

Nathan M. Belliveau; Jens Huft; Paulo Jc Lin; Sam Chen; Alex K. K. Leung; Timothy Leaver; Andre Wild; Justin B. Lee; Robert James Taylor; Ying K. Tam; Carl L. Hansen; Pieter R. Cullis

Lipid nanoparticles (LNP) are the leading systems for in vivo delivery of small interfering RNA (siRNA) for therapeutic applications. Formulation of LNP siRNA systems requires rapid mixing of solutions containing cationic lipid with solutions containing siRNA. Current formulation procedures employ macroscopic mixing processes to produce systems 70-nm diameter or larger that have variable siRNA encapsulation efficiency, homogeneity, and reproducibility. Here, we show that microfluidic mixing techniques, which permit millisecond mixing at the nanoliter scale, can reproducibly generate limit size LNP siRNA systems 20 nm and larger with essentially complete encapsulation of siRNA over a wide range of conditions with polydispersity indexes as low as 0.02. Optimized LNP siRNA systems produced by microfluidic mixing achieved 50% target gene silencing in hepatocytes at a dose level of 10 µg/kg siRNA in mice. We anticipate that microfluidic mixing, a precisely controlled and readily scalable technique, will become the preferred method for formulation of LNP siRNA delivery systems.


Nature | 2017

Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination

Norbert Pardi; Michael J. Hogan; Rebecca S. Pelc; Hiromi Muramatsu; Hanne Andersen; Christina R. DeMaso; Kimberly A. Dowd; Laura L. Sutherland; Richard M. Scearce; Robert Parks; Wendeline Wagner; Alex Granados; Jack Greenhouse; Michelle Walker; Elinor Willis; Jae-Sung Yu; Charles E. McGee; Gregory D. Sempowski; Barbara L. Mui; Ying K. Tam; Yan-Jang Huang; Dana L. Vanlandingham; Veronica M. Holmes; Harikrishnan Balachandran; Sujata Sahu; Michelle A. Lifton; Stephen Higgs; Scott E. Hensley; Thomas D. Madden; Michael J. Hope

Zika virus (ZIKV) has recently emerged as a pandemic associated with severe neuropathology in newborns and adults. There are no ZIKV-specific treatments or preventatives. Therefore, the development of a safe and effective vaccine is a high priority. Messenger RNA (mRNA) has emerged as a versatile and highly effective platform to deliver vaccine antigens and therapeutic proteins. Here we demonstrate that a single low-dose intradermal immunization with lipid-nanoparticle-encapsulated nucleoside-modified mRNA (mRNA–LNP) encoding the pre-membrane and envelope glycoproteins of a strain from the ZIKV outbreak in 2013 elicited potent and durable neutralizing antibody responses in mice and non-human primates. Immunization with 30 μg of nucleoside-modified ZIKV mRNA–LNP protected mice against ZIKV challenges at 2 weeks or 5 months after vaccination, and a single dose of 50 μg was sufficient to protect non-human primates against a challenge at 5 weeks after vaccination. These data demonstrate that nucleoside-modified mRNA–LNP elicits rapid and durable protective immunity and therefore represents a new and promising vaccine candidate for the global fight against ZIKV.


Molecular Therapy | 2011

Influence of cationic lipid composition on gene silencing properties of lipid nanoparticle formulations of siRNA in antigen-presenting cells.

Genc Basha; Tatiana Novobrantseva; Nicole Rosin; Yuen Yi C. Tam; Ismail Hafez; Matthew Wong; Tsukasa Sugo; Vera M. Ruda; June Qin; Boris Klebanov; Marco A. Ciufolini; Akin Akinc; Ying K. Tam; Michael J. Hope; Pieter R. Cullis

Lipid nanoparticles (LNPs) are currently the most effective in vivo delivery systems for silencing target genes in hepatocytes employing small interfering RNA. Antigen-presenting cells (APCs) are also potential targets for LNP siRNA. We examined the uptake, intracellular trafficking, and gene silencing potency in primary bone marrow macrophages (bmMΦ) and dendritic cells of siRNA formulated in LNPs containing four different ionizable cationic lipids namely DLinDAP, DLinDMA, DLinK-DMA, and DLinKC2-DMA. LNPs containing DLinKC2-DMA were the most potent formulations as determined by their ability to inhibit the production of GAPDH target protein. Also, LNPs containing DLinKC2-DMA were the most potent intracellular delivery agents as indicated by confocal studies of endosomal versus cytoplamic siRNA location using fluorescently labeled siRNA. DLinK-DMA and DLinKC2-DMA formulations exhibited improved gene silencing potencies relative to DLinDMA but were less toxic. In vivo results showed that LNP siRNA systems containing DLinKC2-DMA are effective agents for silencing GAPDH in APCs in the spleen and peritoneal cavity following systemic administration. Gene silencing in APCs was RNAi mediated and the use of larger LNPs resulted in substantially reduced hepatocyte silencing, while similar efficacy was maintained in APCs. These results are discussed with regard to the potential of LNP siRNA formulations to treat immunologically mediated diseases.


Molecular Therapy | 2013

Biodegradable Lipids Enabling Rapidly Eliminated Lipid Nanoparticles for Systemic Delivery of RNAi Therapeutics

Martin Maier; Muthusamy Jayaraman; Shigeo Matsuda; Ju Liu; Scott Barros; William Querbes; Ying K. Tam; Steven M. Ansell; Varun Kumar; June Qin; Xuemei Zhang; Qianfan Wang; Sue Panesar; Renta Hutabarat; Mary Carioto; Julia Hettinger; Pachamuthu Kandasamy; David Butler; Kallanthottathil G. Rajeev; Bo Pang; Klaus Charisse; Kevin Fitzgerald; Barbara L. Mui; Xinyao Du; Pieter R. Cullis; Thomas D. Madden; Michael J. Hope; Muthiah Manoharan; Akin Akinc

In recent years, RNA interference (RNAi) therapeutics, most notably with lipid nanoparticle-based delivery systems, have advanced into human clinical trials. The results from these early clinical trials suggest that lipid nanoparticles (LNPs), and the novel ionizable lipids that comprise them, will be important materials in this emerging field of medicine. A persistent theme in the use of materials for biomedical applications has been the incorporation of biodegradability as a means to improve biocompatibility and/or to facilitate elimination. Therefore, the aim of this work was to further advance the LNP platform through the development of novel, next-generation lipids that combine the excellent potency of the most advanced lipids currently available with biodegradable functionality. As a representative example of this novel class of biodegradable lipids, the lipid evaluated in this work displays rapid elimination from plasma and tissues, substantially improved tolerability in preclinical studies, while maintaining in vivo potency on par with that of the most advanced lipids currently available.


International Journal of Cancer | 2012

Lipid nanoparticle siRNA systems for silencing the androgen receptor in human prostate cancer in vivo

Justin B. Lee; Kaixin Zhang; Yuen Yi C. Tam; Ying K. Tam; Nathan M. Belliveau; Vanessa Y.C. Sung; Paulo J.C. Lin; Eric Leblanc; Marco A. Ciufolini; Paul S. Rennie; Pieter R. Cullis

The androgen receptor (AR) plays a critical role in the progression of prostate cancer. Silencing this protein using short‐hairpin RNA (shRNA) has been correlated with tumor growth inhibition and decreases in serum prostate specific antigen (PSA). In our study, we have investigated the ability of lipid nanoparticle (LNP) formulations of small‐interfering RNA (siRNA) to silence AR in human prostate tumor cell lines in vitro and in LNCaP xenograft tumors following intravenous (i.v.) injection. In vitro screening studies using a panel of cationic lipids showed that LNPs containing the ionizable cationic lipid 2,2‐dilinoleyl‐4‐(2‐dimethylaminoethyl)‐[1,3]‐dioxolane (DLin‐KC2‐DMA) exhibited the most potent AR silencing effects in LNCaP cells. This is attributed to an optimized ability of DLin‐KC2‐DMA‐containing LNP to be taken up into cells and to release the siRNA into the cell cytoplasm following endocytotic uptake. DLin‐KC2‐DMA LNPs were also effective in silencing the AR in a wild‐type AR expressing cell line, LAPC‐4, and a variant AR expressing cell line, CWR22Rv1. Importantly, it is demonstrated that LNP AR‐siRNA systems containing DLin‐KC2‐DMA can silence AR gene expression in distal LNCaP xenograft tumors and decrease serum PSA levels following i.v. injection. To our knowledge, this is the first report demonstrating the feasibility of LNP delivery of siRNA for silencing AR gene expression in vivo.


Advanced Drug Delivery Reviews | 2009

Lipid-based delivery of CpG oligonucleotides enhances immunotherapeutic efficacy ☆

Kaley D. Wilson; Susan D. de Jong; Ying K. Tam

There has been significant interest in the potential of cytosine-guanine (CpG) containing oligodeoxynucleotides (ODN) as an immunotherapy for malignant, infectious and allergic diseases. While human trials have yielded promising results, clinical use of free CpG ODN still faces several challenges which limit their effectiveness. These include suboptimal in vivo stability, toxicity, unfavorable pharmacokinetic/biodistribution characteristics, lack of specificity for target cells and the requirement for intracellular uptake. To overcome these challenges, optimized lipid-based delivery systems have been developed to protect the CpG ODN payload, modify their circulation/distribution so as to enhance immune cell targeting and facilitate intracellular uptake. Ultimately, lipid-mediated delivery has the capacity to increase the immunopotency of CpG ODN and enhance their prophylactic or therapeutic efficacy in a range of diseases. Lipid-encapsulation provides a feasible strategy to optimize the immunostimulatory activity and immunotherapeutic efficacy of CpG ODN, thereby allowing their full clinical potential to be realized.


Journal of Controlled Release | 2015

Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes.

Norbert Pardi; Steven Tuyishime; Hiromi Muramatsu; Katalin Karikó; Barbara L. Mui; Ying K. Tam; Thomas D. Madden; Michael J. Hope; Drew Weissman

In recent years, in vitro transcribed messenger RNA (mRNA) has emerged as a potential therapeutic platform. To fulfill its promise, effective delivery of mRNA to specific cell types and tissues needs to be achieved. Lipid nanoparticles (LNPs) are efficient carriers for short-interfering RNAs and have entered clinical trials. However, little is known about the potential of LNPs to deliver mRNA. Here, we generated mRNA-LNPs by incorporating HPLC purified, 1-methylpseudouridine-containing mRNA comprising codon-optimized firefly luciferase into stable LNPs. Mice were injected with 0.005-0.250mg/kg doses of mRNA-LNPs by 6 different routes and high levels of protein translation could be measured using in vivo imaging. Subcutaneous, intramuscular and intradermal injection of the LNP-encapsulated mRNA translated locally at the site of injection for up to 10days. For several days, high levels of protein production could be achieved in the lung from the intratracheal administration of mRNA. Intravenous and intraperitoneal and to a lesser extent intramuscular and intratracheal deliveries led to trafficking of mRNA-LNPs systemically resulting in active translation of the mRNA in the liver for 1-4 days. Our results demonstrate that LNPs are appropriate carriers for mRNA in vivo and have the potential to become valuable tools for delivering mRNA encoding therapeutic proteins.


Molecular therapy. Nucleic acids | 2013

Influence of Polyethylene Glycol Lipid Desorption Rates on Pharmacokinetics and Pharmacodynamics of siRNA Lipid Nanoparticles.

Barbara L. Mui; Ying K. Tam; Muthusamy Jayaraman; Steven M. Ansell; Xinyao Du; Yuen Yi C. Tam; Paulo Jc Lin; Sam Chen; Jayaprakash K. Narayanannair; Kallanthottathil G. Rajeev; Muthiah Manoharan; Akin Akinc; Martin Maier; Pieter R. Cullis; Thomas D. Madden; Michael J. Hope

Lipid nanoparticles (LNPs) encapsulating short interfering RNAs that target hepatic genes are advancing through clinical trials, and early results indicate the excellent gene silencing observed in rodents and nonhuman primates also translates to humans. This success has motivated research to identify ways to further advance this delivery platform. Here, we characterize the polyethylene glycol lipid (PEG-lipid) components, which are required to control the self-assembly process during formation of lipid particles, but can negatively affect delivery to hepatocytes and hepatic gene silencing in vivo. The rate of transfer from LNPs to plasma lipoproteins in vivo is measured for three PEG-lipids with dialkyl chains 14, 16, and 18 carbons long. We show that 1.5 mol % PEG-lipid represents a threshold concentration at which the chain length exerts a minimal effect on hepatic gene silencing but can still modify LNPs pharmacokinetics and biodistribution. Increasing the concentration to 2.5 and 3.5 mol % substantially compromises hepatocyte gene knockdown for PEG-lipids with distearyl (C18) chains but has little impact for shorter dimyristyl (C14) chains. These data are discussed with respect to RNA delivery and the different rates at which the steric barrier disassociates from LNPs in vivo.

Collaboration


Dive into the Ying K. Tam's collaboration.

Top Co-Authors

Avatar

Pieter R. Cullis

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Michael J. Hope

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Thomas D. Madden

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Yuen Yi C. Tam

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sam Chen

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Norbert Pardi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Akin Akinc

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Justin B. Lee

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge