Network


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

Hotspot


Dive into the research topics where Lubomir Nechev is active.

Publication


Featured researches published by Lubomir Nechev.


Nature | 2006

RNAi-mediated gene silencing in non-human primates.

Tracy Zimmermann; Amy C. H. Lee; Akin Akinc; Birgit Bramlage; David Bumcrot; Matthew N. Fedoruk; Jens Harborth; James Heyes; Lloyd Jeffs; Matthias John; Adam Judge; Kieu Lam; Kevin McClintock; Lubomir Nechev; Lorne R. Palmer; Timothy Racie; Ingo Röhl; Stephan Seiffert; Sumi Shanmugam; Vandana Sood; Jürgen Soutschek; Ivanka Toudjarska; Amanda J. Wheat; Ed Yaworski; William Zedalis; Victor Koteliansky; Muthiah Manoharan; Hans-Peter Vornlocher; Ian Maclachlan

The opportunity to harness the RNA interference (RNAi) pathway to silence disease-causing genes holds great promise for the development of therapeutics directed against targets that are otherwise not addressable with current medicines. Although there are numerous examples of in vivo silencing of target genes after local delivery of small interfering RNAs (siRNAs), there remain only a few reports of RNAi-mediated silencing in response to systemic delivery of siRNA, and there are no reports of systemic efficacy in non-rodent species. Here we show that siRNAs, when delivered systemically in a liposomal formulation, can silence the disease target apolipoprotein B (ApoB) in non-human primates. APOB-specific siRNAs were encapsulated in stable nucleic acid lipid particles (SNALP) and administered by intravenous injection to cynomolgus monkeys at doses of 1 or 2.5 mg kg-1. A single siRNA injection resulted in dose-dependent silencing of APOB messenger RNA expression in the liver 48 h after administration, with maximal silencing of >90%. This silencing effect occurred as a result of APOB mRNA cleavage at precisely the site predicted for the RNAi mechanism. Significant reductions in ApoB protein, serum cholesterol and low-density lipoprotein levels were observed as early as 24 h after treatment and lasted for 11 days at the highest siRNA dose, thus demonstrating an immediate, potent and lasting biological effect of siRNA treatment. Our findings show clinically relevant RNAi-mediated gene silencing in non-human primates, supporting RNAi therapeutics as a potential new class of drugs.


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 | 2008

Therapeutic RNAi targeting PCSK9 acutely lowers plasma cholesterol in rodents and LDL cholesterol in nonhuman primates.

Maria Frank-Kamenetsky; Aldo Grefhorst; Norma N. Anderson; Timothy Racie; Birgit Bramlage; Akin Akinc; David Butler; Klaus Charisse; Robert Dorkin; Yupeng Fan; Christina Gamba-Vitalo; Philipp Hadwiger; Muthusamy Jayaraman; Matthias John; K. Narayanannair Jayaprakash; Martin Maier; Lubomir Nechev; Kallanthottathil G. Rajeev; Timothy Read; Ingo Röhl; Jürgen Soutschek; Pamela Tan; Jamie Wong; Gang Wang; Tracy Zimmermann; Antonin de Fougerolles; Hans Peter Vornlocher; Robert Langer; Daniel G. Anderson; Muthiah Manoharan

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates low density lipoprotein receptor (LDLR) protein levels and function. Loss of PCSK9 increases LDLR levels in liver and reduces plasma LDL cholesterol (LDLc), whereas excess PCSK9 activity decreases liver LDLR levels and increases plasma LDLc. Here, we have developed active, cross-species, small interfering RNAs (siRNAs) capable of targeting murine, rat, nonhuman primate (NHP), and human PCSK9. For in vivo studies, PCSK9 and control siRNAs were formulated in a lipidoid nanoparticle (LNP). Liver-specific siRNA silencing of PCSK9 in mice and rats reduced PCSK9 mRNA levels by 50–70%. The reduction in PCSK9 transcript was associated with up to a 60% reduction in plasma cholesterol concentrations. These effects were shown to be mediated by an RNAi mechanism, using 5′-RACE. In transgenic mice expressing human PCSK9, siRNAs silenced the human PCSK9 transcript by >70% and significantly reduced PCSK9 plasma protein levels. In NHP, a single dose of siRNA targeting PCSK9 resulted in a rapid, durable, and reversible lowering of plasma PCSK9, apolipoprotein B, and LDLc, without measurable effects on either HDL cholesterol (HDLc) or triglycerides (TGs). The effects of PCSK9 silencing lasted for 3 weeks after a single bolus i.v. administration. These results validate PCSK9 targeting with RNAi therapeutics as an approach to specifically lower LDLc, paving the way for the development of PCSK9-lowering agents as a future strategy for treatment of hypercholesterolemia.


Antiviral Research | 2008

Evaluation of the safety, tolerability and pharmacokinetics of ALN-RSV01, a novel RNAi antiviral therapeutic directed against respiratory syncytial virus (RSV).

John P. DeVincenzo; Jeffrey Cehelsky; Rene Alvarez; Sayda M. Elbashir; Jens Harborth; Iva Toudjarska; Lubomir Nechev; Veeravagu Murugaiah; Andre van Vliet; Akshay Vaishnaw; Rachel Meyers

Small interfering RNAs (siRNAs) work through RNA interference (RNAi), the natural RNA inhibitory pathway, to down-regulate protein production by inhibiting targeted mRNA in a sequence-specific manner. ALN-RSV01 is an siRNA directed against the mRNA encoding the N-protein of respiratory syncytial virus (RSV) that exhibits specific in vitro and in vivo anti-RSV activity. The results of two safety and tolerability studies with ALN-RSV01 involving 101 healthy adults (65 active, 36 placebo, single- and multiple dose, observer-blind, randomized dose-escalation) are described. Intranasal administration of ALN-RSV01 was well tolerated over a dose range up through 150mg as a single dose and for five daily doses. Adverse events were similar in frequency and severity to placebo (normal saline) and were transient, mild to moderate, with no dose-dependent trend. The frequency or severity of adverse events did not increase with increasing ALN-RSV01 exposure. All subjects completed all treatments and assessments with no early withdrawals or serious adverse events. Physical examinations, vital signs, ECGs and laboratory tests were normal. Systemic bioavailability of ALN-RSV01 was minimal. ALN-RSV01 appears safe and well tolerated when delivered intranasally and is a promising therapeutic candidate for further clinical development.


Antimicrobial Agents and Chemotherapy | 2009

RNA Interference-Mediated Silencing of the Respiratory Syncytial Virus Nucleocapsid Defines a Potent Antiviral Strategy

Rene Alvarez; Sayda M. Elbashir; Todd Borland; Ivanka Toudjarska; Philipp Hadwiger; Mathias John; Ingo Roehl; Svetlana Shulga Morskaya; Rick Martinello; Jeffrey S. Kahn; Mark Van Ranst; Ralph A. Tripp; John P. DeVincenzo; Rajendra K. Pandey; Martin Maier; Lubomir Nechev; Muthiah Manoharan; Victor Kotelianski; Rachel Meyers

ABSTRACT We describe the design and characterization of a potent human respiratory syncytial virus (RSV) nucleocapsid gene-specific small interfering RNA (siRNA), ALN-RSV01. In in vitro RSV plaque assays, ALN-RSV01 showed a 50% inhibitory concentration of 0.7 nM. Sequence analysis of primary isolates of RSV showed that the siRNA target site was absolutely conserved in 89/95 isolates, and ALN-RSV01 demonstrated activity against all isolates, including those with single-mismatch mutations. In vivo, intranasal dosing of ALN-RSV01 in a BALB/c mouse model resulted in potent antiviral efficacy, with 2.5- to 3.0-log-unit reductions in RSV lung concentrations being achieved when ALN-RSV01 was administered prophylactically or therapeutically in both single-dose and multidose regimens. The specificity of ALN-RSV01 was demonstrated in vivo by using mismatch controls; and the absence of an immune stimulatory mechanism was demonstrated by showing that nonspecific siRNAs that induce alpha interferon and tumor necrosis factor alpha lack antiviral efficacy, while a chemically modified form of ALN-RSV01 lacking measurable immunostimulatory capacity retained full activity in vivo. Furthermore, an RNA interference mechanism of action was demonstrated by the capture of the site-specific cleavage product of the RSV mRNA via rapid amplification of cDNA ends both in vitro and in vivo. These studies lay a solid foundation for the further investigation of ALN-RSV01 as a novel therapeutic antiviral agent for clinical use by humans.


Experimental Neurology | 2012

Widespread suppression of huntingtin with convection-enhanced delivery of siRNA

David K. Stiles; Zhiming Zhang; Pei Ge; Brian D. Nelson; Richard Grondin; Yi Ai; Peter A. Hardy; Peter T. Nelson; Andrei P. Guzaev; Mark T. Butt; Klaus Charisse; Verbena Kosovrasti; Lubomir Tchangov; Michael Meys; Martin Maier; Lubomir Nechev; Muthiah Manoharan; William F. Kaemmerer; Douglas Ulen Gwost; Gregory R. Stewart; Don M. Gash; Dinah Sah

Huntingtons disease is an autosomal dominant neurodegenerative disease caused by a toxic gain of function mutation in the huntingtin gene (Htt). Silencing of Htt with RNA interference using direct CNS delivery in rodent models of Huntingtons disease has been shown to reduce pathology and promote neuronal recovery. A key translational step for this approach is extension to the larger non-human primate brain, achieving sufficient distribution of small interfering RNA targeting Htt (siHtt) and levels of Htt suppression that may have therapeutic benefit. We evaluated the potential for convection enhanced delivery (CED) of siHtt to provide widespread and robust suppression of Htt in nonhuman primates. siHtt was infused continuously for 7 or 28 days into the nonhuman primate putamen to analyze effects of infusion rate and drug concentration on the volume of effective suppression. Distribution of radiolabeled siHtt and Htt suppression were quantified by autoradiography and PCR, respectively, in tissue punches. Histopathology was evaluated and Htt suppression was also visualized in animals treated for 28 days. Seven days of CED led to widespread distribution of siHtt and significant Htt silencing throughout the nonhuman primate striatum in an infusion rate and dose dependent manner. Htt suppression at therapeutic dose levels was well tolerated by the brain. A model developed from these results predicts that continuous CED of siHtt can achieve significant coverage of the striatum of Huntingtons disease patients. These findings suggest that this approach may provide an important therapeutic strategy for treating Huntingtons disease.


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 Hepatology | 2016

RNAi-nanoparticulate manipulation of gene expression as a new functional genomics tool in the liver

Hao Yin; Roman L. Bogorad; Carmen Barnes; Stephen Walsh; Iris Zhuang; Hidenori Nonaka; Vera M. Ruda; Satya Kuchimanchi; Lubomir Nechev; Akin Akinc; Wen Xue; Marino Zerial; Robert Langer; Daniel G. Anderson; Victor Koteliansky

BACKGROUND & AIMS The Hippo pathway controls organ size through a negative regulation of the transcription co-activator Yap1. The overexpression of hyperactive mutant Yap1 or deletion of key components in the Hippo pathway leads to increased organ size in different species. Analysis of interactions of this pathway with other cellular signals corroborating organ size control is limited in part due to the difficulties associated with development of rodent models. METHODS Here, we develop a new model of reversible induction of the liver size in mice using siRNA-nanoparticles targeting two kinases of the Hippo pathway, namely, mammalian Ste20 family kinases 1 and 2 (Mst1 and Mst2), and an upstream regulator, neurofibromatosis type II (Nf2). RESULTS The triple siRNAs nanoparticle-induced hepatomegaly in mice phenocopies one observed with Mst1(-/-)Mst2(-/-) liver-specific depletion, as shown by extensive proliferation of hepatocytes and activation of Yap1. The simultaneous co-treatment with a fourth siRNA nanoparticle against Yap1 fully blocked the liver growth. Hippo pathway-induced liver enlargement is associated with p53 activation, evidenced by its accumulation in the nuclei and upregulation of its target genes. Moreover, injections of the triple siRNAs nanoparticle in p53(LSL/LSL) mice shows that livers lacking p53 expression grow faster and exceed the size of livers in p53 wild-type animals, indicating a role of p53 in controlling Yap1-induced liver growth. CONCLUSION Our data show that siRNA-nanoparticulate manipulation of gene expression can provide the reversible control of organ size in adult animals, which presents a new avenue for the investigation of complex regulatory networks in liver.


Angewandte Chemie | 2011

Unique Gene‐Silencing and Structural Properties of 2′‐Fluoro‐Modified siRNAs

Muthiah Manoharan; Akin Akinc; Rajendra K. Pandey; June Qin; Philipp Hadwiger; Matthias John; Kathy Mills; Klaus Charisse; Martin Maier; Lubomir Nechev; Emily M. Greene; Pradeep S. Pallan; Eriks Rozners; Kallanthottathil G. Rajeev; Martin Egli

Collaboration


Dive into the Lubomir Nechev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Maier

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

Dinah Sah

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge