Network


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

Hotspot


Dive into the research topics where Klaus Charisse is active.

Publication


Featured researches published by Klaus Charisse.


Nature | 2004

Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs

Jürgen Soutschek; Akin Akinc; Birgit Bramlage; Klaus Charisse; Rainer Constien; Mary Donoghue; Sayda M. Elbashir; Anke Geick; Philipp Hadwiger; Jens Harborth; Matthias John; Venkitasamy Kesavan; Gary Lavine; Rajendra K. Pandey; Timothy Racie; Kallanthottathil G. Rajeev; Ingo Röhl; Ivanka Toudjarska; Gang Wang; Silvio Wuschko; David Bumcrot; Victor Koteliansky; Stefan Limmer; Muthiah Manoharan; Hans-Peter Vornlocher

RNA interference (RNAi) holds considerable promise as a therapeutic approach to silence disease-causing genes, particularly those that encode so-called ‘non-druggable’ targets that are not amenable to conventional therapeutics such as small molecules, proteins, or monoclonal antibodies. The main obstacle to achieving in vivo gene silencing by RNAi technologies is delivery. Here we show that chemically modified short interfering RNAs (siRNAs) can silence an endogenous gene encoding apolipoprotein B (apoB) after intravenous injection in mice. Administration of chemically modified siRNAs resulted in silencing of the apoB messenger RNA in liver and jejunum, decreased plasma levels of apoB protein, and reduced total cholesterol. We also show that these siRNAs can silence human apoB in a transgenic mouse model. In our in vivo study, the mechanism of action for the siRNAs was proven to occur through RNAi-mediated mRNA degradation, and we determined that cleavage of the apoB mRNA occurred specifically at the predicted site. These findings demonstrate the therapeutic potential of siRNAs for the treatment of disease.


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.


Nature Nanotechnology | 2012

Molecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNA delivery.

Hyukjin Lee; Abigail K. R. Lytton-Jean; Yi Chen; Kevin Love; Angela I. Park; Emmanouil D. Karagiannis; Alfica Sehgal; William Querbes; Christopher Zurenko; Muthusamy Jayaraman; Chang G. Peng; Klaus Charisse; Anna Borodovsky; Muthiah Manoharan; Jessica S. Donahoe; Jessica Truelove; Matthias Nahrendorf; Robert Langer; Daniel G. Anderson

Nanoparticles are employed for delivering therapeutics into cells1,2. However, size, shape, surface chemistry and the presentation of targeting ligands on the surface of nanoparticles can affect circulation half-life and biodistribution, cell specific internalization, excretion, toxicity, and efficacy3-7. A variety of materials have been explored for delivering small interfering RNAs (siRNAs) - a therapeutic agent that suppresses the expression of targeted genes8,9. However, conventional delivery nanoparticles such as liposomes and polymeric systems are heterogeneous in size, composition and surface chemistry, and this can lead to suboptimal performance, lack of tissue specificity and potential toxicity10-12. Here, we show that self-assembled DNA tetrahedral nanoparticles with a well-defined size can deliver siRNAs into cells and silence target genes in tumours. Monodisperse nanoparticles are prepared through the self-assembly of complementary DNA strands. Because the DNA strands are easily programmable, the size of the nanoparticles and the spatial orientation and density of cancer targeting ligands (such as peptides and folate) on the nanoparticle surface can be precisely controlled. We show that at least three folate molecules per nanoparticle is required for optimal delivery of the siRNAs into cells and, gene silencing occurs only when the ligands are in the appropriate spatial orientation. In vivo, these nanoparticles showed a longer blood circulation time (t1/2 ∼ 24.2 min) than the parent siRNA (t1/2 ∼ 6 min).


Journal of the American Chemical Society | 2014

Multivalent N‑Acetylgalactosamine-Conjugated siRNA Localizes in Hepatocytes and Elicits Robust RNAi-Mediated Gene Silencing

Jayaprakash K. Nair; Jennifer L. S. Willoughby; Amy Chan; Klaus Charisse; Md. Rowshon Alam; Qianfan Wang; Menno Hoekstra; Pachamuthu Kandasamy; Alexander V. Kel’in; Nate Taneja; Jonathan O′Shea; Sarfraz Shaikh; Ligang Zhang; Ronald J. van der Sluis; Michael E. Jung; Akin Akinc; Renta Hutabarat; Satya Kuchimanchi; Kevin Fitzgerald; Tracy Zimmermann; Theo J.C. van Berkel; Martin Maier; Kallanthottathil G. Rajeev; Muthiah Manoharan

Conjugation of small interfering RNA (siRNA) to an asialoglycoprotein receptor ligand derived from N-acetylgalactosamine (GalNAc) facilitates targeted delivery of the siRNA to hepatocytes in vitro and in vivo. The ligands derived from GalNAc are compatible with solid-phase oligonucleotide synthesis and deprotection conditions, with synthesis yields comparable to those of standard oligonucleotides. Subcutaneous (SC) administration of siRNA-GalNAc conjugates resulted in robust RNAi-mediated gene silencing in liver. Refinement of the siRNA chemistry achieved a 5-fold improvement in efficacy over the parent design in vivo with a median effective dose (ED50) of 1 mg/kg following a single dose. This enabled the SC administration of siRNA-GalNAc conjugates at therapeutically relevant doses and, importantly, at dose volumes of ≤1 mL. Chronic weekly dosing resulted in sustained dose-dependent gene silencing for over 9 months with no adverse effects in rodents. The optimally chemically modified siRNA-GalNAc conjugates are hepatotropic and long-acting and have the potential to treat a wide range of diseases involving liver-expressed genes.


Nature Nanotechnology | 2014

In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight

James E. Dahlman; Carmen Barnes; Omar F. Khan; Aude Thiriot; Siddharth Jhunjunwala; Taylor E. Shaw; Yiping Xing; Hendrik B. Sager; Gaurav Sahay; Andrew Bader; Roman L. Bogorad; Hao Yin; Tim Racie; Yizhou Dong; Shan Jiang; Danielle Seedorf; Apeksha Dave; Kamaljeet Singh Sandhu; Matthew J. Webber; Tatiana Novobrantseva; Vera M. Ruda; Abigail K. R. Lytton-Jean; Christopher G. Levins; Brian T. Kalish; Dayna K. Mudge; Mario Perez; Ludmila Abezgauz; Partha Dutta; Lynelle Smith; Klaus Charisse

Dysfunctional endothelium contributes to more diseases than any other tissue in the body. Small interfering RNAs (siRNAs) can help in the study and treatment of endothelial cells in vivo by durably silencing multiple genes simultaneously, but efficient siRNA delivery has so far remained challenging. Here, we show that polymeric nanoparticles made of low-molecular-weight polyamines and lipids can deliver siRNA to endothelial cells with high efficiency, thereby facilitating the simultaneous silencing of multiple endothelial genes in vivo. Unlike lipid or lipid-like nanoparticles, this formulation does not significantly reduce gene expression in hepatocytes or immune cells even at the dosage necessary for endothelial gene silencing. These nanoparticles mediate the most durable non-liver silencing reported so far and facilitate the delivery of siRNAs that modify endothelial function in mouse models of vascular permeability, emphysema, primary tumour growth and metastasis.


Nature Biotechnology | 2015

Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown.

Anders Wittrup; Angela Ai; Xing Liu; Péter Hamar; Radiana Trifonova; Klaus Charisse; Muthiah Manoharan; Tomas Kirchhausen; Judy Lieberman

A central hurdle in developing small interfering RNAs (siRNAs) as therapeutics is the inefficiency of their delivery across the plasma and endosomal membranes to the cytosol, where they interact with the RNA interference machinery. With the aim of improving endosomal release, a poorly understood and inefficient process, we studied the uptake and cytosolic release of siRNAs, formulated in lipoplexes or lipid nanoparticles, by live-cell imaging and correlated it with knockdown of a target GFP reporter. siRNA release occurred invariably from maturing endosomes within ∼5–15 min of endocytosis. Cytosolic galectins immediately recognized the damaged endosome and targeted it for autophagy. However, inhibiting autophagy did not enhance cytosolic siRNA release. Gene knockdown occurred within a few hours of release and required <2,000 copies of cytosolic siRNAs. The ability to detect cytosolic release of siRNAs and understand how it is regulated will facilitate the development of rational strategies for improving the cytosolic delivery of candidate drugs.


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.


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.


ChemBioChem | 2015

Hepatocyte‐Specific Delivery of siRNAs Conjugated to Novel Non‐nucleosidic Trivalent N‐Acetylgalactosamine Elicits Robust Gene Silencing in Vivo

Kallanthottathil G. Rajeev; Jayaprakash K. Nair; Muthusamy Jayaraman; Klaus Charisse; Nate Taneja; Jonathan O'Shea; Jennifer L. S. Willoughby; Kristina Yucius; Tuyen Nguyen; Svetlana Shulga-Morskaya; Abigail Liebow; William Querbes; Anna Borodovsky; Kevin Fitzgerald; Martin Maier; Muthiah Manoharan

We recently demonstrated that siRNAs conjugated to triantennary N‐acetylgalactosamine (GalNAc) induce robust RNAi‐mediated gene silencing in the liver, owing to uptake mediated by the asialoglycoprotein receptor (ASGPR). Novel monovalent GalNAc units, based on a non‐nucleosidic linker, were developed to yield simplified trivalent GalNAc‐conjugated oligonucleotides under solid‐phase synthesis conditions. Synthesis of oligonucleotide conjugates using monovalent GalNAc building blocks required fewer synthetic steps compared to the previously optimized triantennary GalNAc construct. The redesigned trivalent GalNAc ligand maintained optimal valency, spatial orientation, and distance between the sugar moieties for proper recognition by ASGPR. siRNA conjugates were synthesized by sequential covalent attachment of the trivalent GalNAc to the 3′‐end of the sense strand and resulted in a conjugate with in vitro and in vivo potency similar to that of the parent trivalent GalNAc conjugate design.


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.

Collaboration


Dive into the Klaus Charisse's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Maier

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

Akin Akinc

Alnylam Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge