Network


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

Hotspot


Dive into the research topics where Julia Hettinger is active.

Publication


Featured researches published by Julia Hettinger.


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.


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.


Blood | 2013

An RNAi therapeutic targeting Tmprss6 decreases iron overload in Hfe−/− mice and ameliorates anemia and iron overload in murine β-thalassemia intermedia

Paul J. Schmidt; Iva Toudjarska; Anoop K. Sendamarai; Tim Racie; Brian Bettencourt; Julia Hettinger; David Bumcrot; Mark D. Fleming

Mutations in HFE lead to hereditary hemochromatosis (HH) because of inappropriately high iron uptake from the diet resulting from decreased hepatic expression of the iron-regulatory hormone hepcidin. -thalassemia is a congenital anemia caused by partial or complete loss of -globin synthesis causing ineffective erythropoiesis, anemia, decreased hepcidin production, and secondary iron overload. Tmprss6 is postulated to regulate hepcidin production by cleaving Hemojuvelin (Hjv), a key modulator of hepcidin expression, from the hepatocyte surface. On this basis, we hypothesized that treatment of mouse models of HH (Hfe(-/-)) and -thalassemia intermedia (Hbb(th3/+)) with Tmprss6 siRNA formulated in lipid nanoparticles (LNPs) that are preferentially taken up by the liver would increase hepcidin expression and lessen the iron loading in both models. In the present study, we demonstrate that LNP-Tmprss6 siRNA treatment of Hfe(-/-) and Hbb(th3/+) mice induces hepcidin and diminishes tissue and serum iron levels. Furthermore, LNP-Tmprss6 siRNA treatment of Hbb(th3/+) mice substantially improved the anemia by altering RBC survival and ineffective erythropoiesis. Our results indicate that pharmacologic manipulation of Tmprss6 with RNAi therapeutics isa practical approach to treating iron overload diseases associated with diminished hepcidin expression and may have efficacy in modifying disease-associated morbidities of -thalassemia intermedia.


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.


Molecular Therapy | 2012

Harnessing a physiologic mechanism for siRNA delivery with mimetic lipoprotein particles

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.


Journal of The American Society of Nephrology | 2017

An Investigational RNAi Therapeutic Targeting Glycolate Oxidase Reduces Oxalate Production in Models of Primary Hyperoxaluria

Abigail Liebow; Xingsheng Li; Timothy Racie; Julia Hettinger; Brian Bettencourt; Nader Najafian; Patrick Haslett; Kevin Fitzgerald; Ross P. Holmes; David V. Erbe; William Querbes; John Knight

Primary hyperoxaluria type 1 (PH1), an inherited rare disease of glyoxylate metabolism, arises from mutations in the enzyme alanine-glyoxylate aminotransferase. The resulting deficiency in this enzyme leads to abnormally high oxalate production resulting in calcium oxalate crystal formation and deposition in the kidney and many other tissues, with systemic oxalosis and ESRD being a common outcome. Although a small subset of patients manages the disease with vitamin B6 treatments, the only effective treatment for most is a combined liver-kidney transplant, which requires life-long immune suppression and carries significant mortality risk. In this report, we discuss the development of ALN-GO1, an investigational RNA interference (RNAi) therapeutic targeting glycolate oxidase, to deplete the substrate for oxalate synthesis. Subcutaneous administration of ALN-GO1 resulted in potent, dose-dependent, and durable silencing of the mRNA encoding glycolate oxidase and increased serum glycolate concentrations in wild-type mice, rats, and nonhuman primates. ALN-GO1 also increased urinary glycolate concentrations in normal nonhuman primates and in a genetic mouse model of PH1. Notably, ALN-GO1 reduced urinary oxalate concentration up to 50% after a single dose in the genetic mouse model of PH1, and up to 98% after multiple doses in a rat model of hyperoxaluria. These data demonstrate the ability of ALN-GO1 to reduce oxalate production in preclinical models of PH1 across multiple species and provide a clear rationale for clinical trials with this compound.


American Journal of Hematology | 2018

RNAi-mediated reduction of hepatic Tmprss6 diminishes anemia and secondary iron overload in a splenectomized mouse model of β-thalassemia intermedia

Paul J. Schmidt; Kaifeng Liu; Gary A. Visner; Kevin Fitzgerald; Shannon Fishman; Tim Racie; Julia Hettinger; James Butler; Mark D. Fleming

Diminished β‐globin synthesis in β‐thalassemia is associated with ineffective erythropoiesis, leading to secondary iron overload caused by inappropriately low levels of hepcidin and to splenomegaly in the symptomatic thalassemias. Splenectomy is often employed in patients with β‐thalassemia to reduce hemolysis. Expression of the iron regulatory peptide hormone hepcidin is repressed by the serine protease TMPRSS6. Hepcidin induction by RNAi‐mediated inhibition of TMPRSS6 expression reduces iron overload and mitigates anemia in murine models of β‐thalassemia intermedia. To interrogate the efficacy of RNAi‐mediated reduction of Tmprss6 in splenectomized β‐thalassemia, splenectomized β‐thalassemic Hbbth3/+ animals were treated with a GalNAc‐conjugated siRNA targeting Tmprss6 (GalNAc‐Tmprss6) and their hematological and iron parameters monitored. We demonstrate that treatment with GalNAc‐Tmprss6 significantly diminishes Tmprss6 expression and appropriately elevates hepcidin expression in splenectomized Hbbth3/+ animals. Similar to unsplenectomized animals, treated animals have markedly improved anemia due to diminished ineffective erythropoiesis and reduced iron loading in both serum and tissue. These results suggest that RNAi‐mediated reduction of Tmprss6 may have positive outcomes even in splenectomized β‐thalassemia patients.


Blood | 2011

Targeting the Hepcidin Pathway with RNAi Therapeutics for the Treatment of Anemia

Akin Akinc; Amy Chan-Daniels; Alfica Sehgal; Don Foster; Brian Bettencourt; Julia Hettinger; Tim Racie; Justin Aubin; Satya Kuchimanchi; Hila Epstein-Barash; Tomoko Nakayama


Blood | 2012

An RNAi Therapeutic Targeting Antithrombin Increases Thrombin Generation in Nonhuman Primates

Akin Akinc; Alfica Sehgal; Scott Barros; Julia Hettinger; Klaus Charisse; Josh Brodsky; June Qin; Tim Racie


Blood | 2011

RNAi-Mediated Inhibition of Activated Protein C- A New Approach for Hemophilia Treatment

Ivanka Toudjarska; Zuhua Cai; Tim Racie; Julia Hettinger; Brian Bettencourt; Dinah Sah; David Bumcrot; John Maraganore; Akshay Vaishnaw

Collaboration


Dive into the Julia Hettinger's collaboration.

Top Co-Authors

Avatar

Tim Racie

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Akin Akinc

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

June Qin

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Scott Barros

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Mary Carioto

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge