Network


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

Hotspot


Dive into the research topics where Owen S. Fenton is active.

Publication


Featured researches published by Owen S. Fenton.


Nature Communications | 2014

Degradable lipid nanoparticles with predictable in vivo siRNA delivery activity

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.


Nano Letters | 2015

Optimization of Lipid Nanoparticle Formulations for mRNA Delivery in Vivo with Fractional Factorial and Definitive Screening Designs.

Kevin J. Kauffman; Dorkin; Junghoon Yang; Michael Heartlein; Frank Derosa; Mir Ff; Owen S. Fenton; Daniel G. Anderson

Intracellular delivery of messenger RNA (mRNA) has the potential to induce protein production for many therapeutic applications. Although lipid nanoparticles have shown considerable promise for the delivery of small interfering RNAs (siRNA), their utility as agents for mRNA delivery has only recently been investigated. The most common siRNA formulations contain four components: an amine-containing lipid or lipid-like material, phospholipid, cholesterol, and lipid-anchored polyethylene glycol, the relative ratios of which can have profound effects on the formulation potency. Here, we develop a generalized strategy to optimize lipid nanoparticle formulations for mRNA delivery to the liver in vivo using Design of Experiment (DOE) methodologies including Definitive Screening and Fractional Factorial Designs. By simultaneously varying lipid ratios and structures, we developed an optimized formulation which increased the potency of erythropoietin-mRNA-loaded C12-200 lipid nanoparticles 7-fold relative to formulations previously used for siRNA delivery. Key features of this optimized formulation were the incorporation of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and increased ionizable lipid:mRNA weight ratios. Interestingly, the optimized lipid nanoparticle formulation did not improve siRNA delivery, indicating differences in optimized formulation parameter design spaces for siRNA and mRNA. We believe the general method described here can accelerate in vivo screening and optimization of nanoparticle formulations with large multidimensional design spaces.


Nano Letters | 2017

Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer Immunotherapy

Matthias A. Oberli; Andreas M. Reichmuth; J. Robert Dorkin; Michael J. Mitchell; Owen S. Fenton; Ana Jaklenec; Daniel G. Anderson; Robert Langer; Daniel Blankschtein

The induction of a strong cytotoxic T cell response is an important prerequisite for successful immunotherapy against many viral diseases and tumors. Nucleotide vaccines, including mRNA vaccines with their intracellular antigen synthesis, have been shown to be potent activators of a cytotoxic immune response. The intracellular delivery of mRNA vaccines to the cytosol of antigen presenting immune cells is still not sufficiently well understood. Here, we report on the development of a lipid nanoparticle formulation for the delivery of mRNA vaccines to induce a cytotoxic CD 8 T cell response. We show transfection of dendritic cells, macrophages, and neutrophils. The efficacy of the vaccine was tested in an aggressive B16F10 melanoma model. We found a strong CD 8 T cell activation after a single immunization. Treatment of B16F10 melanoma tumors with lipid nanoparticles containing mRNA coding for the tumor-associated antigens gp100 and TRP2 resulted in tumor shrinkage and extended the overall survival of the treated mice. The immune response can be further increased by the incorporation of the adjuvant LPS. In conclusion, the lipid nanoparticle formulation presented here is a promising vector for mRNA vaccine delivery, one that is capable of inducing a strong cytotoxic T cell response. Further optimization, including the incorporation of different adjuvants, will likely enhance the potency of the vaccine.


Advanced Materials | 2016

Bioinspired Alkenyl Amino Alcohol Ionizable Lipid Materials for Highly Potent In Vivo mRNA Delivery

Owen S. Fenton; Kevin J. Kauffman; Rebecca L. Mcclellan; Eric A. Appel; J. Robert Dorkin; Mark W. Tibbitt; Michael Heartlein; Frank Derosa; Robert Langer; Daniel G. Anderson

Thousands of human diseases could be treated by selectively controlling the expression of specific proteins in vivo. A new series of alkenyl amino alcohol (AAA) ionizable lipid nanoparticles (LNPs) capable of delivering human mRNA with unprecedented levels of in vivo efficacy is demonstrated. This study highlights the importance of utilizing synthesis tools in tandem with biological inspiration to understand and improve nucleic acid delivery in vivo.


Angewandte Chemie | 2016

Polymer–Lipid Nanoparticles for Systemic Delivery of mRNA to the Lungs

James C. Kaczmarek; Asha K. Patel; Kevin J. Kauffman; Owen S. Fenton; Matthew J. Webber; Michael Heartlein; Frank Derosa; Daniel G. Anderson

Therapeutic nucleic acids hold great promise for the treatment of disease but require vectors for safe and effective delivery. Synthetic nanoparticle vectors composed of poly(β-amino esters) (PBAEs) and nucleic acids have previously demonstrated potential utility for local delivery applications. To expand this potential utility to include systemic delivery of mRNA, hybrid polymer-lipid nanoformulations for systemic delivery to the lungs were developed. Through coformulation of PBAEs with lipid-polyethylene glycol (PEG), mRNA formulations were developed with increased serum stability and increased in vitro potency. The formulations were capable of functional delivery of mRNA to the lungs after intravenous administration in mice. To our knowledge, this is the first report of the systemic administration of mRNA for delivery to the lungs using degradable polymer-lipid nanoparticles.


Advanced Materials | 2017

Synthesis and Biological Evaluation of Ionizable Lipid Materials for the In Vivo Delivery of Messenger RNA to B Lymphocytes

Owen S. Fenton; Kevin J. Kauffman; James C. Kaczmarek; Rebecca L. Mcclellan; Siddharth Jhunjhunwala; Mark W. Tibbitt; Manhao D. Zeng; Eric A. Appel; Joseph R. Dorkin; Faryal F. Mir; Jung H. Yang; Matthias A. Oberli; Michael Heartlein; Frank Derosa; Robert Langer; Daniel G. Anderson

B lymphocytes regulate several aspects of immunity including antibody production, cytokine secretion, and T-cell activation; moreover, B cell misregulation is implicated in autoimmune disorders and cancers such as multiple sclerosis and non-Hodgkins lymphomas. The delivery of messenger RNA (mRNA) into B cells can be used to modulate and study these biological functions by means of inducing functional protein expression in a dose-dependent and time-controlled manner. However, current in vivo mRNA delivery systems fail to transfect B lymphocytes and instead primarily target hepatocytes and dendritic cells. Here, the design, synthesis, and biological evaluation of a lipid nanoparticle (LNP) system that can encapsulate mRNA, navigate to the spleen, transfect B lymphocytes, and induce more than 60 pg of protein expression per million B cells within the spleen is described. Importantly, this LNP induces more than 85% of total protein production in the spleen, despite LNPs being observed transiently in the liver and other organs. These results demonstrate that LNP composition alone can be used to modulate the site of protein induction in vivo, highlighting the critical importance of designing and synthesizing new nanomaterials for nucleic acid delivery.


Advanced Healthcare Materials | 2016

Poly(Limonene Thioether) Scaffold for Tissue Engineering

Kristin M. Fischer; Kathy Ye Morgan; Keith Hearon; Demetra Sklaviadis; Zachary L. Tochka; Owen S. Fenton; Daniel G. Anderson; Robert Langer; Lisa E. Freed

A photocurable thiol-ene network polymer, poly(limonene thioether) (PLT32o), is synthesized, characterized, fabricated into tissue engineering scaffolds, and demonstrated in vitro and in vivo. Micromolded PLT32o grids exhibit compliant, elastomeric mechanical behavior similar to grids made of poly(glycerol sebacate) (PGS), an established biomaterial. Multilayered PL32o scaffolds with regular, geometrically defined pore architectures support heart cell seeding and culture in a manner similar to multilayered PGS scaffolds. Subcutaneous implantation of multilayered PLT32o scaffolds with cultured heart cells provides long-term 3D structural support and retains the exogenous cells, whereas PGS scaffolds lose both their structural integrity and the exogenous cells over 31 d in vivo. PLT32o membrane implants retain their dry mass, whereas PGS implants lose 70 percent of their dry mass by day 31. Macrophages are initially recruited to PLT32o and PGS membrane implants but are no longer present by day 31. Facile synthesis and processing in combination with the capability to support heart cells in vitro and in vivo suggest that PLT32o can offer advantages for tissue engineering applications where prolonged in vivo maintenance of 3D structural integrity and elastomeric mechanical behavior are required.


Advanced Materials | 2018

Advances in Biomaterials for Drug Delivery

Owen S. Fenton; Katy N. Olafson; Padmini S. Pillai; Michael J. Mitchell; Robert Langer

Advances in biomaterials for drug delivery are enabling significant progress in biology and medicine. Multidisciplinary collaborations between physical scientists, engineers, biologists, and clinicians generate innovative strategies and materials to treat a range of diseases. Specifically, recent advances include major breakthroughs in materials for cancer immunotherapy, autoimmune diseases, and genome editing. Here, strategies for the design and implementation of biomaterials for drug delivery are reviewed. A brief history of the biomaterials field is first established, and then commentary on RNA delivery, responsive materials development, and immunomodulation are provided. Current challenges associated with these areas as well as opportunities to address long-standing problems in biology and medicine are discussed throughout.


Nature Biomedical Engineering | 2018

Biomanufacturing for clinically advanced cell therapies

Ayesha Aijaz; Matthew Li; David Smith; Danika Khong; Courtney LeBlon; Owen S. Fenton; Ronke M. Olabisi; Steven Libutti; Jay Tischfield; Marcela V. Maus; Robert Deans; Rita N. Barcia; Daniel G. Anderson; Jerome Ritz; Robert Preti; Biju Parekkadan

The achievements of cell-based therapeutics have galvanized efforts to bring cell therapies to the market. To address the demands of the clinical and eventual commercial-scale production of cells, and with the increasing generation of large clinical datasets from chimeric antigen receptor T-cell immunotherapy, from transplants of engineered haematopoietic stem cells and from other promising cell therapies, an emphasis on biomanufacturing requirements becomes necessary. Robust infrastructure should address current limitations in cell harvesting, expansion, manipulation, purification, preservation and formulation, ultimately leading to successful therapy administration to patients at an acceptable cost. In this Review, we highlight case examples of cutting-edge bioprocessing technologies that improve biomanufacturing efficiency for cell therapies approaching clinical use.This Review discusses the manufacturing of cell products for clinically advanced cell therapies, and highlights potential manufacturing bottlenecks and solutions towards the cost-effective commercialization of the therapies.


Advanced Healthcare Materials | 2018

Poly(β‐amino ester)‐co‐poly(caprolactone) Terpolymers as Nonviral Vectors for mRNA Delivery In Vitro and In Vivo

Umberto Capasso Palmiero; James C. Kaczmarek; Owen S. Fenton; Daniel G. Anderson

The production of new proteins with messenger RNA (mRNA) has gained a broad interest due to its potential for addressing a wide range of diseases. Here, the design and characterization of novel ionizable poly(β-amino ester)-co-poly(caprolactone) terpolymers, synthesized via the combination of the ring opening polymerization and the Michael step-growth polymerization, are reported. The versatility of this method is demonstrated by varying the number of caprolactone units attached to each poly(β-amino ester) (PBAE) terpolymer. The ability of the novel poly-caprolactone (PCL)-based PBAE materials to deliver mRNA is shown to depend on the physiochemical characteristics of the material, such as lipophilicity, as well as the formulation method used to complex the polymer with the oligonucleotide. This latter variable represents a previously unstudied aspect of PBAE library screens that can play an important role in identifying true top candidates for nucleic acid delivery. The most stable terpolymer is injected intravenously (IV) in mice and shows a transfection efficacy several times higher than the polyethylenimine (PEI) which is focused in the spleen, opening the possibility to use these biodegradable carriers in the intravenous delivery of antigen-encoding mRNA for cancer immunotherapy and vaccination.

Collaboration


Dive into the Owen S. Fenton's collaboration.

Top Co-Authors

Avatar

Daniel G. Anderson

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Kevin J. Kauffman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Robert Langer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

James C. Kaczmarek

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Rebecca L. Mcclellan

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Joseph R. Dorkin

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Mark W. Tibbitt

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Robert Dorkin

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Mohammad Movassaghi

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge