Network


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

Hotspot


Dive into the research topics where Arturo Vegas is active.

Publication


Featured researches published by Arturo Vegas.


Nature Reviews Genetics | 2014

Non-viral vectors for gene-based therapy

Hao Yin; Rosemary Lynn Kanasty; Ahmed A. Eltoukhy; Arturo Vegas; J. Robert Dorkin; Daniel G. Anderson

Gene-based therapy is the intentional modulation of gene expression in specific cells to treat pathological conditions. This modulation is accomplished by introducing exogenous nucleic acids such as DNA, mRNA, small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides. Given the large size and the negative charge of these macromolecules, their delivery is typically mediated by carriers or vectors. In this Review, we introduce the biological barriers to gene delivery in vivo and discuss recent advances in material sciences, nanotechnology and nucleic acid chemistry that have yielded promising non-viral delivery systems, some of which are currently undergoing testing in clinical trials. The diversity of these systems highlights the recent progress of gene-based therapy using non-viral approaches.


Nature Materials | 2013

Delivery materials for siRNA therapeutics

Rosemary Lynn Kanasty; Joseph R. Dorkin; Arturo Vegas; Daniel G. Anderson

RNA interference (RNAi) has broad potential as a therapeutic to reversibly silence any gene. To achieve the clinical potential of RNAi, delivery materials are required to transport short interfering RNA (siRNA) to the site of action in the cells of target tissues. This Review provides an introduction to the biological challenges that siRNA delivery materials aim to overcome, as well as a discussion of the way that the most effective and clinically advanced classes of siRNA delivery systems, including lipid nanoparticles and siRNA conjugates, are designed to surmount these challenges. The systems that we discuss are diverse in their approaches to the delivery problem, and provide valuable insight to guide the design of future siRNA delivery materials.


Nature Materials | 2015

Size- and shape-dependent foreign body immune response to materials implanted in rodents and non-human primates

Omid Veiseh; Joshua C. Doloff; Minglin Ma; Arturo Vegas; Hok Hei Tam; Andrew Bader; Jie Li; Erin Langan; Jeffrey Wyckoff; Whitney S. Loo; Siddharth Jhunjhunwala; Alan Chiu; Sean Siebert; Katherine Tang; Jennifer Hollister-Lock; Stephanie Aresta-Dasilva; Matthew A. Bochenek; Joshua E. Mendoza-Elias; Yong Wang; Merigeng Qi; Danya M. Lavin; Michael Chen; Nimit Dholakia; Raj Thakrar; Igor Lacík; Gordon C. Weir; Jose Oberholzer; Dale L. Greiner; Robert Langer; Daniel G. Anderson

The efficacy of implanted biomedical devices is often compromised by host recognition and subsequent foreign body responses. Here, we demonstrate the role of the geometry of implanted materials on their biocompatibility in vivo. In rodent and non-human primate animal models, implanted spheres 1.5 mm and above in diameter across a broad spectrum of materials, including hydrogels, ceramics, metals, and plastics, significantly abrogated foreign body reactions and fibrosis when compared to smaller spheres. We also show that for encapsulated rat pancreatic islet cells transplanted into streptozotocin-treated diabetic C57BL/6 mice, islets prepared in 1.5 mm alginate capsules were able to restore blood-glucose control for up to 180 days, a period more than 5-fold longer than for transplanted grafts encapsulated within conventionally sized 0.5-mm alginate capsules. Our findings suggest that the in vivo biocompatibility of biomedical devices can be significantly improved by simply tuning their spherical dimensions.


Nature Medicine | 2016

Long-term glycemic control using polymer-encapsulated human stem cell–derived beta cells in immune-competent mice

Arturo Vegas; Omid Veiseh; Mads Gürtler; Jeffrey R. Millman; Felicia W. Pagliuca; Andrew Bader; Joshua C. Doloff; Jie Li; Michael Chen; Karsten Olejnik; Hok Hei Tam; Siddharth Jhunjhunwala; Erin Langan; Stephanie Aresta-Dasilva; Srujan Gandham; James J. McGarrigle; Matthew A. Bochenek; Jennifer Hollister-Lock; Jose Oberholzer; Dale L. Greiner; Gordon C. Weir; Douglas A. Melton; Robert Langer; Daniel G. Anderson

The transplantation of glucose-responsive, insulin-producing cells offers the potential for restoring glycemic control in individuals with diabetes. Pancreas transplantation and the infusion of cadaveric islets are currently implemented clinically, but these approaches are limited by the adverse effects of immunosuppressive therapy over the lifetime of the recipient and the limited supply of donor tissue. The latter concern may be addressed by recently described glucose-responsive mature beta cells that are derived from human embryonic stem cells (referred to as SC-β cells), which may represent an unlimited source of human cells for pancreas replacement therapy. Strategies to address the immunosuppression concerns include immunoisolation of insulin-producing cells with porous biomaterials that function as an immune barrier. However, clinical implementation has been challenging because of host immune responses to the implant materials. Here we report the first long-term glycemic correction of a diabetic, immunocompetent animal model using human SC-β cells. SC-β cells were encapsulated with alginate derivatives capable of mitigating foreign-body responses in vivo and implanted into the intraperitoneal space of C57BL/6J mice treated with streptozotocin, which is an animal model for chemically induced type 1 diabetes. These implants induced glycemic correction without any immunosuppression until their removal at 174 d after implantation. Human C-peptide concentrations and in vivo glucose responsiveness demonstrated therapeutically relevant glycemic control. Implants retrieved after 174 d contained viable insulin-producing cells.


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 | 2012

Action and Reaction: The Biological Response to siRNA and Its Delivery Vehicles

Rosemary Lynn Kanasty; Kathryn A. Whitehead; Arturo Vegas; Daniel G. Anderson

RNA interference (RNAi)-based therapeutics have significant potential for the treatment of human disease. Safe and effective delivery of RNA to target tissues remains a major barrier to realizing its clinical potential. Several factors can affect the in vivo performance of short interfering RNA (siRNA) delivery formulations, including siRNA sequence, structure, chemical modification, and delivery formulation. This review provides an introduction to the principles underlying the pharmacokinetics and pharmacodynamics of systemically administered siRNA and its delivery formulations, including the factors that lead to its degradation, clearance, and tissue uptake, as well as its potential for immunogenicity, toxicity, and off-target effects within the body.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Combinatorial synthesis of chemically diverse core-shell nanoparticles for intracellular delivery

Daniel J. Siegwart; Kathryn A. Whitehead; Lutz Nuhn; Gaurav Sahay; Hao Cheng; Shan Jiang; Minglin Ma; Abigail K. R. Lytton-Jean; Arturo Vegas; Patrick Fenton; Christopher G. Levins; Kevin Love; Haeshin Lee; Christina Cortez; Sean P. Collins; Ying Fei Li; Janice Jang; William Querbes; Christopher Zurenko; Tatiana Novobrantseva; Robert Langer; Daniel G. Anderson

Analogous to an assembly line, we employed a modular design for the high-throughput study of 1,536 structurally distinct nanoparticles with cationic cores and variable shells. This enabled elucidation of complexation, internalization, and delivery trends that could only be learned through evaluation of a large library. Using robotic automation, epoxide-functionalized block polymers were combinatorially cross-linked with a diverse library of amines, followed by measurement of molecular weight, diameter, RNA complexation, cellular internalization, and in vitro siRNA and pDNA delivery. Analysis revealed structure-function relationships and beneficial design guidelines, including a higher reactive block weight fraction, stoichiometric equivalence between epoxides and amines, and thin hydrophilic shells. Cross-linkers optimally possessed tertiary dimethylamine or piperazine groups and potential buffering capacity. Covalent cholesterol attachment allowed for transfection in vivo to liver hepatocytes in mice. The ability to tune the chemical nature of the core and shell may afford utility of these materials in additional applications.


Endocrine Reviews | 2011

Islets Transplanted in Immunoisolation Devices: A Review of the Progress and the Challenges that Remain

Esther O'Sullivan; Arturo Vegas; Daniel G. Anderson; Gordon C. Weir

The concept of using an immunoisolation device to facilitate the transplantation of islets without the need for immunosuppression has been around for more than 50 yr. Significant progress has been made in developing suitable materials that satisfy the need for biocompatibility, durability, and permselectivity. However, the search is ongoing for a device that allows sufficient oxygen transfer while maintaining a barrier to immune cells and preventing rejection of the transplanted tissue. Separating the islets from the rich blood supply in the native pancreas takes its toll. The immunoisolated islets commonly suffer from hypoxia and necrosis, which in turn triggers a host immune response. Efforts have been made to improve the supply of nutrients by using proangiogenic factors to augment the development of a vascular supply in the transplant site, by using small islet cell aggregates to reduce the barrier to diffusion of oxygen, or by creating scaffolds that are in close proximity to a vascular network such as the omental blood supply. Even if these efforts are successful, the shortage of donor islet tissue available for transplantation remains a major problem. To this end, a search for a renewable source of insulin-producing cells is ongoing; whether these will come from adult or embryonic stem cells or xenogeneic sources remains to be seen. Herein we will review the above issues and chart the progress made with various immunoisolation devices in small and large animal models and the small number of clinical trials carried out to date.


Nature Biotechnology | 2016

Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates

Arturo Vegas; Omid Veiseh; Joshua C. Doloff; Minglin Ma; Hok Hei Tam; Kaitlin M. Bratlie; Jie Li; Andrew Bader; Erin Langan; Karsten Olejnik; Patrick Fenton; Jeon Woong Kang; Jennifer Hollister-Locke; Matthew A. Bochenek; Alan Chiu; Sean Siebert; Katherine Tang; Siddharth Jhunjhunwala; Stephanie Aresta-Dasilva; Nimit Dholakia; Raj Thakrar; Thema Vietti; Michael Chen; Josh Cohen; Karolina Siniakowicz; Meirigeng Qi; James J. McGarrigle; Stephen Lyle; David M. Harlan; Dale L. Greiner

The foreign body response is an immune-mediated reaction that can lead to the failure of implanted medical devices and discomfort for the recipient. There is a critical need for biomaterials that overcome this key challenge in the development of medical devices. Here we use a combinatorial approach for covalent chemical modification to generate a large library of variants of one of the most widely used hydrogel biomaterials, alginate. We evaluated the materials in vivo and identified three triazole-containing analogs that substantially reduce foreign body reactions in both rodents and, for at least 6 months, in non-human primates. The distribution of the triazole modification creates a unique hydrogel surface that inhibits recognition by macrophages and fibrous deposition. In addition to the utility of the compounds reported here, our approach may enable the discovery of other materials that mitigate the foreign body response.

Collaboration


Dive into the Arturo Vegas's collaboration.

Top Co-Authors

Avatar

Daniel G. Anderson

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Robert Langer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Joshua C. Doloff

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Omid Veiseh

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Bader

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jie Li

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dale L. Greiner

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Michael Chen

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge