Scott Barros
Alnylam Pharmaceuticals
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Scott Barros.
Nature Biotechnology | 2010
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.
Proceedings of the National Academy of Sciences of the United States of America | 2014
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.
Nature Communications | 2014
Kathryn A. Whitehead; J. Robert Dorkin; Arturo Vegas; Philip H. Chang; Omid Veiseh; Jonathan C. F. Matthews; Owen S. Fenton; Yunlong Zhang; Karsten Olejnik; Volkan Yesilyurt; Delai Chen; Scott Barros; Boris Klebanov; Tatiana Novobrantseva; Robert Langer; Daniel G. Anderson
One of the most significant challenges in the development of clinically-viable delivery systems for RNA interference therapeutics is to understand how molecular structures influence delivery efficacy. To this end, we synthesized 1400 degradable lipidoids and evaluated their transfection ability and structure function activity. Here we show that lipidoid nanoparticles mediate potent gene knockdown in hepatocytes and immune cell populations upon IV administration to mice (siRNA EC50 values as low as 0.01 mg/kg). Surprisingly, we identify four necessary and sufficient structural and pKa criteria that robustly predict the ability of nanoparticles to mediate greater than 95% protein silencing in vivo. Because these efficacy criteria can be dictated through chemical design, this discovery could eliminate our dependence on time-consuming and expensive cell culture assays and animal testing. Herein, we identify promising degradable lipidoids and describe new design criteria that reliably predict in vivo siRNA delivery efficacy without any prior biological testing.
Nature Medicine | 2015
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
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.
Molecular therapy. Nucleic acids | 2012
Tatiana Novobrantseva; Anna Borodovsky; Jamie Wong; Boris Klebanov; Mohammad Zafari; Kristina Yucius; William Querbes; Pei Ge; Vera M. Ruda; Rick Duncan; Scott Barros; Genc Basha; Pieter R. Cullis; Akin Akinc; Jessica S. Donahoe; K. Narayanannair Jayaprakash; Muthusamy Jayaraman; Roman L. Bogorad; Kevin Love; Katie Whitehead; Chris Levins; Muthiah Manoharan; Filip K. Swirski; Ralph Weissleder; Robert Langer; Daniel G. Anderson; Antonin de Fougerolles; Matthias Nahrendorf; Victor Koteliansky
Leukocytes are central regulators of inflammation and the target cells of therapies for key diseases, including autoimmune, cardiovascular, and malignant disorders. Efficient in vivo delivery of small interfering RNA (siRNA) to immune cells could thus enable novel treatment strategies with broad applicability. In this report, we develop systemic delivery methods of siRNA encapsulated in lipid nanoparticles (LNP) for durable and potent in vivo RNA interference (RNAi)-mediated silencing in myeloid cells. This work provides the first demonstration of siRNA-mediated silencing in myeloid cell types of nonhuman primates (NHPs) and establishes the feasibility of targeting multiple gene targets in rodent myeloid cells. The therapeutic potential of these formulations was demonstrated using siRNA targeting tumor necrosis factor-α (TNFα) which induced substantial attenuation of disease progression comparable to a potent antibody treatment in a mouse model of rheumatoid arthritis (RA). In summary, we demonstrate a broadly applicable and therapeutically relevant platform for silencing disease genes in immune cells.
Molecular Therapy | 2012
Tomoko Nakayama; James Butler; Alfica Sehgal; Mariano Severgnini; Tim Racie; Jennifer Sharman; Feng Ding; Svetlana Shulga Morskaya; Joshua Brodsky; Lubomir Tchangov; Verbena Kosovrasti; Mike Meys; Lubomir Nechev; Gang Wang; Chang Geng Peng; Yupang Fang; Martin Maier; Kallanthottathil G. Rajeev; Robert Li; Julia Hettinger; Scott Barros; Valerie Clausen; Xuemei Zhang; Qianfan Wang; Renta Hutabarat; Nikolay V. Dokholyan; Christian Wolfrum; Muthiah Manoharan; Victor Kotelianski; Markus Stoffel
Therapeutics based on RNA interference (RNAi) have emerged as a potential new class of drugs for treating human disease by silencing the target messenger RNA (mRNA), thereby reducing levels of the corresponding pathogenic protein. The major challenge for RNAi therapeutics is the development of safe delivery vehicles for small interfering RNAs (siRNAs). We previously showed that cholesterol-conjugated siRNAs (chol-siRNA) associate with plasma lipoprotein particles and distribute primarily to the liver after systemic administration to mice. We further demonstrated enhancement of silencing by administration of chol-siRNA pre-associated with isolated high-density lipoprotein (HDL) or low-density lipoprotein (LDL). In this study, we investigated mimetic lipoprotein particle prepared from recombinant apolipoprotein A1 (apoA) and apolipoprotein E3 (apoE) as a delivery vehicle for chol-siRNAs. We show that apoE-containing particle (E-lip) is highly effective in functional delivery of chol-siRNA to mouse liver. E-lip delivery was found to be considerably more potent than apoA-containing particle (A-lip). Furthermore, E-lip–mediated delivery was not significantly affected by high endogenous levels of plasma LDL. These results demonstrate that E-lip has substantial potential as delivery vehicles for lipophilic conjugates of siRNAs.
Molecular therapy. Nucleic acids | 2014
Varun Kumar; June Qin; Yongfeng Jiang; Richard G. Duncan; Benjamin Brigham; Shannon Fishman; Jayaprakash K. Nair; Akin Akinc; Scott Barros; Pia Kasperkovitz
Formulation of short interfering RNA (siRNA) into multicomponent lipid nanoparticles (LNP) is an effective strategy for hepatic delivery and therapeutic gene silencing. This study systematically evaluated the effect of polyethylene glycol (PEG) density on LNP physicochemical properties, innate immune response stimulation, and in vivo efficacy. Increased PEG density not only shielded LNP surface charge but also reduced hemolytic activity, suggesting the formation of a steric barrier. In addition, increasing the PEG density reduced LNP immunostimulatory potential as reflected in cytokine induction both in vivo and in vitro. Higher PEG density also hindered in vivo efficacy, presumably due to reduced association with apolipoprotein E (ApoE), a protein which serves as an endogenous targeting ligand to hepatocytes. This effect could be overcome by incorporating an exogenous targeting ligand into the highly shielded LNPs, thereby circumventing the requirement for ApoE association. Therefore, these studies provide useful information for the rational design of LNP-based siRNA delivery systems with an optimal safety and efficacy profile.
Gastroenterology | 2015
Alfica Sehgal; Keith Blomenkamp; Kun Qian; Amy Simon; Patrick Haslett; Scott Barros; Jeffrey Teckman
value <10-8), and between hepatic fibrosis and metabolic traits revealed a significant association between hepatic fibrosis and HbA1c (p-value <0.001). There was no significant shared gene effect between hepatic steatosis and hepatic fibrosis. Conclusions: Utilizing a wellcharacterized prospective cohort of community-dwelling twins, this study demonstrates that both hepatic steatosis and hepatic fibrosis are heritable traits. Although both are heritable, they appear to have distinct basis for their genetic susceptibility.
Gastroenterology | 2014
Alfica Sehgal; Keith Blomenkamp; Kun Qian; Shannon Fishman; James Butler; Satya Kuchimanch; Scott Barros; Brian Bettencourt; Klaus Charisse; Amy Simon; David Bumcrot; Kevin Fitzgerald; Jeffrey Teckman
The segmentation motor pattern of the intestine, the most dominant pattern in response to eating, designed to optimally absorb nutrients, was described in the late 19th century. How the intestine switches from propulsion to segmentation was revealed recently by us in Nature Communications (in press). Fatty acids induce a low frequency transient rhythmic pacemaker in a network of interstitial cells of Cajal (ICC) associated with the deepmuscular neural plexus (ICC-DMP; 1-4 cycles/min), second to the dominant omnipresent slow wave pacemaker that is generated by ICC associated with the myenteric plexus (ICC-MP; 30-40 cycles/min) in both mouse and rat colon. The introduction of the second pacemaker is associated with slow wave activity changing into a waxing and waning electrical pattern. Not unlike the interaction of waves caused by two stones dropped in water, the pacemaker activities interact with each other, changing a propulsive motor pattern into a non-propagating rhythmic pattern of short-lasting contractions. Our aim of the present study was to understand the mechanism of initiation and control of this trigger for segmentation, and the switch that can change segmentation into propulsion and vice versa. Using intracellular electrical recordings (n=21), we found that the ICC-DMP pacemaker is under tonic inhibitory control by enteric nitrergic nerves and can be evoked by substance P acting on NK1 receptors of ICCDMP at 3.6 ± 0.6 cycles/min. Furthermore, the switch can be activated by medium chain fatty acids through the blood stream at 2.8 ± 0.5 cycles/min, and by short chain fatty acids via the lumen. To further understand the nature of the interaction between the two pacemakers we have developed a model based on the electrophysiological characteristics of the electrical pacemaker activities and based on phase-amplitude coupling of the electrical activities. The motor patterns of the musculature develop because both pacemaker activities propagate into the musculature from opposite sides. The segmentation activity develops when the propagation velocities of the two pacemakers are significantly different from each other. When the propagation velocities are similar, the two pacemakers strengthen each other to create strong low frequency propulsive activity. Hence, the ICC-DMP play a critical role in initiation and the regulation of the nature of the motor pattern of the intestine. It is likely therefore that abnormalities in the ICC-DMP pacemaker activity or its neural or nutrient control may give rise to either the propulsive or segmentation motor pattern becoming too dominant resulting in diarrhea or constipation. The ICC-DMP pacemaker will therefore become a target for rectifying abnormal motor patterns.