Network


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

Hotspot


Dive into the research topics where June Qin is active.

Publication


Featured researches published by June Qin.


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.


Nature Biotechnology | 2008

A combinatorial library of lipid-like materials for delivery of RNAi therapeutics

Akin Akinc; Andreas Zumbuehl; Michael Goldberg; Elizaveta S. Leshchiner; Valentina Busini; Naushad Hossain; Sergio Bacallado; David N. Nguyen; Jason Fuller; Rene Alvarez; Anna Borodovsky; Todd Borland; Rainer Constien; Antonin de Fougerolles; J. Robert Dorkin; K. Narayanannair Jayaprakash; Muthusamy Jayaraman; Matthias John; Victor Koteliansky; Muthiah Manoharan; Lubomir Nechev; June Qin; Timothy Racie; Denitza Raitcheva; Kallanthottathil G. Rajeev; Dinah Sah; Jürgen Soutschek; Ivanka Toudjarska; Hans-Peter Vornlocher; Tracy Zimmermann

The safe and effective delivery of RNA interference (RNAi) therapeutics remains an important challenge for clinical development. The diversity of current delivery materials remains limited, in part because of their slow, multi-step syntheses. Here we describe a new class of lipid-like delivery molecules, termed lipidoids, as delivery agents for RNAi therapeutics. Chemical methods were developed to allow the rapid synthesis of a large library of over 1,200 structurally diverse lipidoids. From this library, we identified lipidoids that facilitate high levels of specific silencing of endogenous gene transcripts when formulated with either double-stranded small interfering RNA (siRNA) or single-stranded antisense 2′-O-methyl (2′-OMe) oligoribonucleotides targeting microRNA (miRNA). The safety and efficacy of lipidoids were evaluated in three animal models: mice, rats and nonhuman primates. The studies reported here suggest that these materials may have broad utility for both local and systemic delivery of RNA therapeutics.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Lipid-like materials for low-dose, in vivo gene silencing

Kevin Love; Kerry P. Mahon; Christopher G. Levins; Kathryn A. Whitehead; William Querbes; J. Robert Dorkin; June Qin; William Cantley; Liu Liang Qin; Timothy Racie; Maria Frank-Kamenetsky; Ka Ning Yip; Rene Alvarez; Dinah Sah; Antonin de Fougerolles; Kevin Fitzgerald; Victor Koteliansky; Akin Akinc; Robert Langer; Daniel G. Anderson

Significant effort has been applied to discover and develop vehicles which can guide small interfering RNAs (siRNA) through the many barriers guarding the interior of target cells. While studies have demonstrated the potential of gene silencing in vivo, improvements in delivery efficacy are required to fulfill the broadest potential of RNA interference therapeutics. Through the combinatorial synthesis and screening of a different class of materials, a formulation has been identified that enables siRNA-directed liver gene silencing in mice at doses below 0.01 mg/kg. This formulation was also shown to specifically inhibit expression of five hepatic genes simultaneously, after a single injection. The potential of this formulation was further validated in nonhuman primates, where high levels of knockdown of the clinically relevant gene transthyretin was observed at doses as low as 0.03 mg/kg. To our knowledge, this formulation facilitates gene silencing at orders-of-magnitude lower doses than required by any previously described siRNA liver delivery system.


Molecular Therapy | 2010

Targeted Delivery of RNAi Therapeutics With Endogenous and Exogenous Ligand-Based Mechanisms

Akin Akinc; William Querbes; Soma De; June Qin; Maria Frank-Kamenetsky; K. Narayanannair Jayaprakash; Muthusamy Jayaraman; Kallanthottathil G. Rajeev; William Cantley; J. Robert Dorkin; James Butler; Liuliang Qin; Timothy Racie; Andrew Sprague; Eugenio Fava; Anja Zeigerer; Michael J. Hope; Marino Zerial; Dinah Sah; Kevin Fitzgerald; Mark Tracy; Muthiah Manoharan; Victor Koteliansky; Antonin de Fougerolles; Martin Maier

Lipid nanoparticles (LNPs) have proven to be highly efficient carriers of short-interfering RNAs (siRNAs) to hepatocytes in vivo; however, the precise mechanism by which this efficient delivery occurs has yet to be elucidated. We found that apolipoprotein E (apoE), which plays a major role in the clearance and hepatocellular uptake of physiological lipoproteins, also acts as an endogenous targeting ligand for ionizable LNPs (iLNPs), but not cationic LNPs (cLNPs). The role of apoE was investigated using both in vitro studies employing recombinant apoE and in vivo studies in wild-type and apoE-/- mice. Receptor dependence was explored in vitro and in vivo using low-density lipoprotein receptor (LDLR-/-)-deficient mice. As an alternative to endogenous apoE-based targeting, we developed a targeting approach using an exogenous ligand containing a multivalent N-acetylgalactosamine (GalNAc)-cluster, which binds with high affinity to the asialoglycoprotein receptor (ASGPR) expressed on hepatocytes. Both apoE-based endogenous and GalNAc-based exogenous targeting appear to be highly effective strategies for the delivery of iLNPs to liver.Lipid nanoparticles (LNPs) have proven to be highly efficient carriers of short-interfering RNAs (siRNAs) to hepatocytes in vivo; however, the precise mechanism by which this efficient delivery occurs has yet to be elucidated. We found that apolipoprotein E (apoE), which plays a major role in the clearance and hepatocellular uptake of physiological lipoproteins, also acts as an endogenous targeting ligand for ionizable LNPs (iLNPs), but not cationic LNPs (cLNPs). The role of apoE was investigated using both in vitro studies employing recombinant apoE and in vivo studies in wild-type and apoE(-/-) mice. Receptor dependence was explored in vitro and in vivo using low-density lipoprotein receptor (LDLR(-/-))-deficient mice. As an alternative to endogenous apoE-based targeting, we developed a targeting approach using an exogenous ligand containing a multivalent N-acetylgalactosamine (GalNAc)-cluster, which binds with high affinity to the asialoglycoprotein receptor (ASGPR) expressed on hepatocytes. Both apoE-based endogenous and GalNAc-based exogenous targeting appear to be highly effective strategies for the delivery of iLNPs to liver.


Molecular Therapy | 2009

Development of Lipidoid–siRNA Formulations for Systemic Delivery to the Liver

Akin Akinc; Michael Goldberg; June Qin; J. Robert Dorkin; Christina Gamba-Vitalo; Martin Maier; K. Narayanannair Jayaprakash; Muthusamy Jayaraman; Kallanthottathil G. Rajeev; Muthiah Manoharan; Victor Koteliansky; Ingo Röhl; Elizaveta S. Leshchiner; Robert Langer; Daniel G. Anderson

RNA interference therapeutics afford the potential to silence target gene expression specifically, thereby blocking production of disease-causing proteins. The development of safe and effective systemic small interfering RNA (siRNA) delivery systems is of central importance to the therapeutic application of siRNA. Lipid and lipid-like materials are currently the most well-studied siRNA delivery systems for liver delivery, having been utilized in several animal models, including nonhuman primates. Here, we describe the development of a multicomponent, systemic siRNA delivery system, based on the novel lipid-like material 98N(12)-5(1). We show that in vivo delivery efficacy is affected by many parameters, including the formulation composition, nature of particle PEGylation, degree of drug loading, and biophysical parameters such as particle size. In particular, small changes in the anchor chain length of poly(ethylene glycol) (PEG) lipids can result in significant effects on in vivo efficacy. The lead formulation developed is liver targeted (>90% injected dose distributes to liver) and can induce fully reversible, long-duration gene silencing without loss of activity following repeat administration.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates

Yizhou Dong; Kevin Love; J. Robert Dorkin; Sasilada Sirirungruang; Yunlong Zhang; Delai Chen; Roman L. Bogorad; Hao Yin; Yi Chen; Arturo Vegas; Christopher A. Alabi; Gaurav Sahay; Karsten Olejnik; Weiheng Wang; Avi Schroeder; Abigail K. R. Lytton-Jean; Daniel J. Siegwart; Akin Akinc; Carmen Barnes; Scott Barros; Mary Carioto; Kevin Fitzgerald; Julia Hettinger; Varun Kumar; Tatiana Novobrantseva; June Qin; William Querbes; Victor Koteliansky; Robert Langer; Daniel G. Anderson

Significance The safe, selective, and efficient delivery of siRNA is a key challenge to the broad application of siRNA therapeutics in humans. Motivated by the structure of lipoproteins, we developed lipopeptide nanomaterials for siRNA delivery. In vivo in mice, siRNA–lipopeptide particles provide the most potent delivery to hepatocytes (ED50 ∼ 0.002 mg/kg for FVII silencing), with the highest selectivity of delivery to hepatocytes over nontarget cell types (orders of magnitude), yet reported. These materials also show efficacy in nonhuman primates. siRNA therapeutics have promise for the treatment of a wide range of genetic disorders. Motivated by lipoproteins, we report lipopeptide nanoparticles as potent and selective siRNA carriers with a wide therapeutic index. Lead material cKK-E12 showed potent silencing effects in mice (ED50 ∼ 0.002 mg/kg), rats (ED50 < 0.01 mg/kg), and nonhuman primates (over 95% silencing at 0.3 mg/kg). Apolipoprotein E plays a significant role in the potency of cKK-E12 both in vitro and in vivo. cKK-E12 was highly selective toward liver parenchymal cell in vivo, with orders of magnitude lower doses needed to silence in hepatocytes compared with endothelial cells and immune cells in different organs. Toxicity studies showed that cKK-E12 was well tolerated in rats at a dose of 1 mg/kg (over 100-fold higher than the ED50). To our knowledge, this is the most efficacious and selective nonviral siRNA delivery system for gene silencing in hepatocytes reported to date.


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.


Nature Medicine | 2015

An RNAi therapeutic targeting antithrombin to rebalance the coagulation system and promote hemostasis in hemophilia

Alfica Sehgal; Scott Barros; Lacramioara Ivanciu; Brian C. Cooley; June Qin; Tim Racie; Julia Hettinger; Mary Carioto; Yongfeng Jiang; Josh Brodsky; Harsha Prabhala; Xuemei Zhang; Husain Attarwala; Renta Hutabarat; Don Foster; Klaus Charisse; Satya Kuchimanchi; Martin Maier; Lubo Nechev; Pachamuthu Kandasamy; Alexander V. Kelin; Jayaprakash K. Nair; Kallanthottathil G. Rajeev; Muthiah Manoharan; Rachel Meyers; Benny Sorensen; Amy Simon; Yesim Dargaud; Claude Negrier; Rodney M. Camire

Hemophilia A and B are inherited bleeding disorders characterized by deficiencies in procoagulant factor VIII (FVIII) or factor IX (FIX), respectively. There remains a substantial unmet medical need in hemophilia, especially in patients with inhibitory antibodies against replacement factor therapy, for novel and improved therapeutic agents that can be used prophylactically to provide effective hemostasis. Guided by reports suggesting that co-inheritance of prothrombotic mutations may ameliorate the clinical phenotype in hemophilia, we developed an RNA interference (RNAi) therapeutic (ALN-AT3) targeting antithrombin (AT) as a means to promote hemostasis in hemophilia. When administered subcutaneously, ALN-AT3 showed potent, dose-dependent, and durable reduction of AT levels in wild-type mice, mice with hemophilia A, and nonhuman primates (NHPs). In NHPs, a 50% reduction in AT levels was achieved with weekly dosing at approximately 0.125 mg/kg, and a near-complete reduction in AT levels was achieved with weekly dosing at 1.5 mg/kg. Treatment with ALN-AT3 promoted hemostasis in mouse models of hemophilia and led to improved thrombin generation in an NHP model of hemophilia A with anti-factor VIII inhibitors. This investigational compound is currently in phase 1 clinical testing in subjects with hemophilia A or B.


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.


Bioconjugate Chemistry | 2010

Combinatorial approach to determine functional group effects on lipidoid-mediated siRNA delivery.

Kerry P. Mahon; Kevin Love; Kathryn A. Whitehead; June Qin; Akin Akinc; Elizaveta S. Leshchiner; Ignaty Leshchiner; Robert Langer; Daniel G. Anderson

The application of RNA interference (RNAi), either in the clinic or in the laboratory, requires safe and effective delivery methods. Here, we develop a combinatorial approach to synthesize a library of delivery vectors based on two lipid-like substrates with known siRNA delivery capabilities. Members of this library have a mixture of lipid-like tails and feature appendages containing hydroxyl, carbamate, ether, or amine functional groups as well as variations in alkyl chain length and branching. Using a luciferase reporter system in HeLa cells, we studied the relationship between lipid chemical modification and delivery performance in vitro. The impact of the functional group was shown to vary depending on the overall amine content and tail number of the delivery vector. Additionally, in vivo performance was evaluated using a Factor VII knockdown assay. Two library members, each containing ether groups, were found to knock down the target protein at levels comparable to those of the parent delivery vector. These results demonstrate that small chemical changes to the delivery vector impact knockdown efficiency and cell viability both in vitro and in vivo. The work described here identifies new materials for siRNA delivery and provides new insight into the parameters for optimized chemical makeup of lipid-like siRNA delivery materials.

Collaboration


Dive into the June Qin's collaboration.

Top Co-Authors

Avatar

Akin Akinc

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Maier

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dinah Sah

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

J. Robert Dorkin

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Scott Barros

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge