Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Med Chem ; 66(4): 2506-2523, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36757090

ABSTRACT

Conjugation of synthetic triantennary N-acetyl-d-galactosamine (GalNAc) to small interfering RNA (siRNA) mediates binding to the asialoglycoprotein receptor (ASGPR) on the surface of hepatocytes, facilitating liver-specific uptake and siRNA-mediated gene silencing. The natural ß-glycosidic bond of the GalNAc ligand is rapidly cleaved by glycosidases in vivo. Novel GalNAc ligands with S-, and C-glycosides with both α- and ß-anomeric linkages, N-glycosides with ß-anomeric linkage, and the O-glycoside with α-anomeric linkage were synthesized and conjugated to siRNA either on-column during siRNA synthesis or through a high-throughput, post-synthetic method. Unlike natural GalNAc, modified ligands were resistant to glycosidase activity. The siRNAs conjugated to newly designed ligands had similar affinities for ASGPR and similar silencing activity in mice as the parent GalNAc-siRNA conjugate. These data suggest that other factors, such as protein-nucleic acid interactions and loading of the antisense strand into the RNA-induced silencing complex (RISC), are more critical to the duration of action than the stereochemistry and stability of the anomeric linkage between the GalNAc moiety of the ligand conjugated to the sense strand of the siRNA.


Subject(s)
Asialoglycoprotein Receptor , Galactosamine , RNA, Small Interfering , RNA-Induced Silencing Complex , Animals , Mice , Acetylgalactosamine/chemistry , Asialoglycoprotein Receptor/metabolism , Glycoside Hydrolases/metabolism , Glycosides/metabolism , Hepatocytes/metabolism , Ligands , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/metabolism
2.
Nucleic Acids Res ; 45(19): 10969-10977, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28981809

ABSTRACT

Covalent attachment of a synthetic triantennary N-acetylagalactosamine (GalNAc) ligand to chemically modified siRNA has enabled asialoglycoprotein (ASGPR)-mediated targeted delivery of therapeutically active siRNAs to hepatocytes in vivo. This approach has become transformative for the delivery of RNAi therapeutics as well as other classes of investigational oligonucleotide therapeutics to the liver. For efficient functional delivery of intact drug into the desired subcellular compartment, however, it is critical that the nucleic acids are stabilized against nucleolytic degradation. Here, we compared two siRNAs of the same sequence but with different modification pattern resulting in different degrees of protection against nuclease activity. In vitro stability studies in different biological matrices show that 5'-exonuclease is the most prevalent nuclease activity in endo-lysosomal compartments and that additional stabilization in the 5'-regions of both siRNA strands significantly enhances the overall metabolic stability of GalNAc-siRNA conjugates. In good agreement with in vitro findings, the enhanced stability translated into substantially improved liver exposure, gene silencing efficacy and duration of effect in mice. Follow-up studies with a second set of conjugates targeting a different transcript confirmed the previous results, provided additional insights into kinetics of RISC loading and demonstrated excellent translation to non-human primates.


Subject(s)
Acetylgalactosamine/pharmacokinetics , Kidney/metabolism , Liver/metabolism , RNA, Small Interfering/pharmacokinetics , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/metabolism , Animals , Area Under Curve , Drug Delivery Systems/methods , Humans , Liver/cytology , Male , Metabolic Clearance Rate , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/metabolism
3.
Mol Ther ; 25(1): 71-78, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-28129130

ABSTRACT

Advancement of RNAi-based therapeutics depends on effective delivery to the site of protein synthesis. Although intravenously administered, multi-component delivery vehicles have enabled small interfering RNA (siRNA) delivery and progression into clinical development, advances of single-component, systemic siRNA delivery have been challenging. In pre-clinical models, attachment of a triantennary N-acetylgalactosamine (GalNAc) ligand to an siRNA mediates hepatocyte uptake via the asialoglycoprotein receptor enabling RNAi-mediated gene silencing. In this phase 1 study, we assessed translation of this delivery approach by evaluating the safety, tolerability, pharmacokinetics, and pharmacodynamics of a GalNAc-siRNA conjugate, revusiran, targeting transthyretin (TTR). Subjects received a placebo or ascending doses of revusiran subcutaneously ranging from 1.25-10 mg/kg in the single and 2.5-10 mg/kg in the multiple ascending dose phases. Revusiran was generally well tolerated, with transient, mild to moderate injection site reactions the most common treatment-emergent adverse events. Doses of 2.5-10 mg/kg revusiran elicited a significant reduction of serum TTR versus the placebo (p < 0.01), with mean TTR reductions of approximately 90% observed with multiple dosing. These results demonstrate translation of this novel delivery platform, enabling clinical development of subcutaneously administered GalNAc-siRNAs for liver-based diseases.


Subject(s)
Acetylgalactosamine , Hepatocytes/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Acetylgalactosamine/chemistry , Adult , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Drug Monitoring , Female , Gene Silencing , Healthy Volunteers , Humans , Male , Middle Aged , Prealbumin/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Young Adult
4.
Nat Med ; 21(5): 492-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25849132

ABSTRACT

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.


Subject(s)
Antithrombins/chemistry , Blood Coagulation/drug effects , Factor IX/chemistry , Factor VIII/chemistry , Hemophilia A/drug therapy , RNA Interference , Animals , Dose-Response Relationship, Drug , Female , Hemophilia A/genetics , Hemostasis/drug effects , Homozygote , Humans , Male , Mice , Mutation
5.
J Am Chem Soc ; 136(49): 16958-61, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25434769

ABSTRACT

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.


Subject(s)
Acetylgalactosamine/chemistry , Gene Silencing , Hepatocytes/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Animals , Mice , Mice, Inbred C57BL , Molecular Structure
6.
Lancet ; 383(9911): 60-68, 2014 Jan 04.
Article in English | MEDLINE | ID: mdl-24094767

ABSTRACT

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to LDL receptors, leading to their degradation. Genetics studies have shown that loss-of-function mutations in PCSK9 result in reduced plasma LDL cholesterol and decreased risk of coronary heart disease. We aimed to investigate the safety and efficacy of ALN-PCS, a small interfering RNA that inhibits PCSK9 synthesis, in healthy volunteers with raised cholesterol who were not on lipid-lowering treatment. METHODS: We did a randomised, single-blind, placebo-controlled, phase 1 dose-escalation study in healthy adult volunteers with serum LDL cholesterol of 3·00 mmol/L or higher. Participants were randomly assigned in a 3:1 ratio by computer algorithm to receive one dose of intravenous ALN-PCS (with doses ranging from 0·015 to 0·400 mg/kg) or placebo. The primary endpoint was safety and tolerability of ALN-PCS. Secondary endpoints were the pharmacokinetic characteristics of ALN-PCS and its pharmacodynamic effects on PCSK9 and LDL cholesterol. Study participants were masked to treatment assignment. Analysis was per protocol and we used ANCOVA to analyse pharmacodynamic endpoint data. This trial is registered with ClinicalTrials.gov, number NCT01437059. FINDINGS: Of 32 participants, 24 were randomly allocated to receive a single dose of ALN-PCS (0·015 mg/kg [n=3], 0·045 mg/kg [n=3], 0·090 mg/kg [n=3], 0·150 mg/kg [n=3], 0·250 mg/kg [n=6], or 0·400 mg/kg [n=6]) and eight to placebo. The proportions of patients affected by treatment-emergent adverse events were similar in the ALN-PCS and placebo groups (19 [79%] vs seven [88%]). ALN-PCS was rapidly distributed, with peak concentration and area under the curve (0 to last measurement) increasing in a roughly dose-proportional way across the dose range tested. In the group given 0·400 mg/kg of ALN-PCS, treatment resulted in a mean 70% reduction in circulating PCSK9 plasma protein (p<0·0001) and a mean 40% reduction in LDL cholesterol from baseline relative to placebo (p<0·0001). INTERPRETATION: Our results suggest that inhibition of PCSK9 synthesis by RNA interference (RNAi) provides a potentially safe mechanism to reduce LDL cholesterol concentration in healthy individuals with raised cholesterol. These results support the further assessment of ALN-PCS in patients with hypercholesterolaemia, including those being treated with statins. This study is the first to show an RNAi drug being used to affect a clinically validated endpoint (ie, LDL cholesterol) in human beings. FUNDING: Alnylam Pharmaceuticals.


Subject(s)
Cholesterol, LDL/blood , Genetic Therapy/methods , Proprotein Convertases/biosynthesis , RNA, Small Interfering/pharmacology , Serine Endopeptidases/biosynthesis , Adult , Cholesterol, LDL/drug effects , Dose-Response Relationship, Drug , Female , Genetic Therapy/adverse effects , Healthy Volunteers , Humans , Male , Middle Aged , Proprotein Convertase 9 , Proprotein Convertases/blood , Proprotein Convertases/genetics , RNA Interference , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/adverse effects , Serine Endopeptidases/blood , Serine Endopeptidases/genetics , Single-Blind Method
7.
N Engl J Med ; 369(9): 819-29, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23984729

ABSTRACT

BACKGROUND: Transthyretin amyloidosis is caused by the deposition of hepatocyte-derived transthyretin amyloid in peripheral nerves and the heart. A therapeutic approach mediated by RNA interference (RNAi) could reduce the production of transthyretin. METHODS: We identified a potent antitransthyretin small interfering RNA, which was encapsulated in two distinct first- and second-generation formulations of lipid nanoparticles, generating ALN-TTR01 and ALN-TTR02, respectively. Each formulation was studied in a single-dose, placebo-controlled phase 1 trial to assess safety and effect on transthyretin levels. We first evaluated ALN-TTR01 (at doses of 0.01 to 1.0 mg per kilogram of body weight) in 32 patients with transthyretin amyloidosis and then evaluated ALN-TTR02 (at doses of 0.01 to 0.5 mg per kilogram) in 17 healthy volunteers. RESULTS: Rapid, dose-dependent, and durable lowering of transthyretin levels was observed in the two trials. At a dose of 1.0 mg per kilogram, ALN-TTR01 suppressed transthyretin, with a mean reduction at day 7 of 38%, as compared with placebo (P=0.01); levels of mutant and nonmutant forms of transthyretin were lowered to a similar extent. For ALN-TTR02, the mean reductions in transthyretin levels at doses of 0.15 to 0.3 mg per kilogram ranged from 82.3 to 86.8%, with reductions of 56.6 to 67.1% at 28 days (P<0.001 for all comparisons). These reductions were shown to be RNAi-mediated. Mild-to-moderate infusion-related reactions occurred in 20.8% and 7.7% of participants receiving ALN-TTR01 and ALN-TTR02, respectively. CONCLUSIONS: ALN-TTR01 and ALN-TTR02 suppressed the production of both mutant and nonmutant forms of transthyretin, establishing proof of concept for RNAi therapy targeting messenger RNA transcribed from a disease-causing gene. (Funded by Alnylam Pharmaceuticals; ClinicalTrials.gov numbers, NCT01148953 and NCT01559077.).


Subject(s)
Amyloid Neuropathies, Familial/therapy , Prealbumin/genetics , RNA, Small Interfering/therapeutic use , Adolescent , Adult , Amyloid Neuropathies, Familial/genetics , Animals , Dose-Response Relationship, Drug , Female , Humans , Liposomes , Macaca fascicularis , Male , Nanocapsules , Prealbumin/metabolism , RNA, Small Interfering/administration & dosage , Young Adult
8.
Mol Ther ; 21(8): 1570-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23799535

ABSTRACT

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.


Subject(s)
Drug Delivery Systems , Gene Transfer Techniques , Lipids/chemistry , Nanoparticles/administration & dosage , RNA, Small Interfering/genetics , Animals , Cell Line , Factor VII/genetics , Factor VII/metabolism , Gene Silencing , Genetic Therapy , Humans , Lipids/pharmacokinetics , Macaca fascicularis , Male , Mice , Nanoparticles/chemistry , Nanoparticles/toxicity , RNA Interference , RNA, Small Interfering/chemistry , Rats
9.
Cancer Discov ; 3(4): 406-17, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358650

ABSTRACT

UNLABELLED: RNA interference (RNAi) is a potent and specific mechanism for regulating gene expression. Harnessing RNAi to silence genes involved in disease holds promise for the development of a new class of therapeutics. Delivery is key to realizing the potential of RNAi, and lipid nanoparticles (LNP) have proved effective in delivery of siRNAs to the liver and to tumors in animals. To examine the activity and safety of LNP-formulated siRNAs in humans, we initiated a trial of ALN-VSP, an LNP formulation of siRNAs targeting VEGF and kinesin spindle protein (KSP), in patients with cancer. Here, we show detection of drug in tumor biopsies, siRNA-mediated mRNA cleavage in the liver, pharmacodynamics suggestive of target downregulation, and antitumor activity, including complete regression of liver metastases in endometrial cancer. In addition, we show that biweekly intravenous administration of ALN-VSP was safe and well tolerated. These data provide proof-of-concept for RNAi therapeutics in humans and form the basis for further development in cancer. SIGNIFICANCE: The fi ndings in this report show safety, pharmacokinetics, RNAi mechanism of action, and clinical activity with a novel fi rst-in-class LNP-formulated RNAi therapeutic in patients with cancer. The ability to harness RNAi to facilitate specifi c multitargeting, as well as increase the number of druggable targets, has important implications for future drug development in oncology.


Subject(s)
Kinesins/genetics , Liver Neoplasms/therapy , Nanoparticles/administration & dosage , RNA Interference , RNA, Small Interfering/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Adult , Aged , Animals , Cell Line, Tumor , Cytokines/blood , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, SCID , Middle Aged , RNA, Messenger/metabolism , Xenograft Model Antitumor Assays
10.
Mol Ther ; 20(8): 1582-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22850721

ABSTRACT

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.


Subject(s)
Lipoproteins/administration & dosage , Lipoproteins/chemistry , RNA, Small Interfering/administration & dosage , Animals , Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/chemistry , Apolipoproteins E/administration & dosage , Apolipoproteins E/chemistry , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/chemistry , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/chemistry , Male , Mice , Mice, Inbred C57BL , RNA Interference/physiology , RNA, Small Interfering/genetics
11.
Silence ; 1(1): 16, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20731861

ABSTRACT

BACKGROUND: While increasing numbers of small interfering RNA (siRNA) therapeutics enter into clinical trials, the quantification of siRNA from clinical samples for pharmacokinetic studies remains a challenge. This challenge is even more acute for the quantification of chemically modified and formulated siRNAs such as those typically required for systemic delivery. RESULTS: Here, we describe a novel method, heating-in-Triton quantitative reverse transcription PCR (HIT qRT-PCR) that improves upon the stem-loop RT-PCR technique for the detection of formulated and chemically modified siRNAs from plasma and tissue. The broad dynamic range of this assay spans five orders of magnitude and can detect as little as 70 pg duplex in 1 g of liver or in 1 ml of plasma. We have used this assay to quantify intravenously administrated siRNA in rodents and have reliably correlated target reduction with tissue drug concentrations. We were able to detect siRNA in rat liver for at least 10 days post injection and determined that for a modified factor VII (FVII) siRNA, on average, approximately 500 siRNA molecules per cell are required to achieve a 50% target reduction. CONCLUSIONS: HIT qRT-PCR is a novel approach that simplifies the in vivo quantification of siRNA and provides a highly sensitive and reproducible tool to measure the silencing efficiency of chemically modified and formulated siRNAs.

12.
Thromb Haemost ; 104(3): 563-70, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20589313

ABSTRACT

Desmopressin aggravates thrombocytopenia in type 2B von Willebrand disease (VWF type 2B) by release of large and hyper-adhesive von Willebrand Factor (VWF) multimers. This pilot study investigated whether the anti-VWF aptamer ARC1779 can prevent desmopressin-induced thrombocytopenia and interferes with the excessive VWF turnover in patients with VWF type 2B. Concentration effect curves of ARC1779 were established for five patients in vitro and two patients with VWF type 2B were treated by infusion of ARC1779, desmopressin, or their combination in a randomised, controlled, double-blind design. ARC1779 concentrations in the range of 1-3 microg/ml blocked free A1 domain binding sites by 90% in vitro. In vivo, desmopressin alone induced a profound (-90%) drop in platelet counts in one of the patients. ARC1779 (4-5 microg/ml) completely inhibited VWF A1 domains and prevented this desmopressin-induced platelet drop. Desmopressin alone increased VWF antigen two- to three-fold, accompanied by concordant changes in VWF Ristocetin cofactor activity (RCo) and coagulation factor VIII activity. ARC1779 substantially enhanced the desmopressin-induced maximal increase in these parameters, and improved multimer patterns. No treatment related adverse events were observed and no bleeding occurred despite marked thrombocytopenia. These data provide first proof of concept in humans and evidence that ARC1779 is a potent inhibitor of VWF. ARC1779 prevented the rapid consumption of VWF multimers together with agglutinated platelets that occurred in response to desmopressin challenge in patients with VWD type 2B.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Hemostatics/therapeutic use , von Willebrand Disease, Type 2/drug therapy , von Willebrand Factor/antagonists & inhibitors , Adult , Aged , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/pharmacokinetics , Austria , Binding Sites , Deamino Arginine Vasopressin/administration & dosage , Deamino Arginine Vasopressin/adverse effects , Double-Blind Method , Factor VIII/metabolism , Female , Hemostatics/administration & dosage , Hemostatics/adverse effects , Hemostatics/pharmacokinetics , Humans , Infusions, Intravenous , Male , Middle Aged , Partial Thromboplastin Time , Pilot Projects , Platelet Count , Platelet Function Tests , Protein Multimerization , Thrombocytopenia/blood , Thrombocytopenia/chemically induced , Time Factors , Treatment Outcome , von Willebrand Disease, Type 2/blood , von Willebrand Factor/metabolism
13.
Silence ; 1(1): 14, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20615220

ABSTRACT

Fire and Mello initiated the current explosion of interest in RNA interference (RNAi) biology with their seminal work in Caenorhabditis elegans. These observations were closely followed by the demonstration of RNAi in Drosophila melanogaster. However, the full potential of these new discoveries only became clear when Tuschl and colleagues showed that 21-22 bp RNA duplexes with 3" overhangs, termed small interfering (si)RNAs, could reliably execute RNAi in a range of mammalian cells. Soon afterwards, it became clear that many different human cell types had endogenous machinery, the RNA-induced silencing complex (RISC), which could be harnessed to silence any gene in the genome. Beyond the availability of a novel way to dissect biology, an important target validation tool was now available. More importantly, two key properties of the RNAi pathway - sequence-mediated specificity and potency - suggested that RNAi might be the most important pharmacological advance since the advent of protein therapeutics. The implications were profound. One could now envisage selecting disease-associated targets at will and expect to suppress proteins that had remained intractable to inhibition by conventional methods, such as small molecules. This review attempts to summarize the current understanding on siRNA lead discovery, the delivery of RNAi therapeutics, typical in vivo pharmacological profiles, preclinical safety evaluation and an overview of the 14 programs that have already entered clinical practice.

14.
Transfusion ; 49(10): 2181-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19538431

ABSTRACT

BACKGROUND: Plasma exchange is the main therapy for thrombotic thrombocytopenic purpura (TTP). No treatments other than plasma exchange have been documented to be effective nor are approved for treatment of TTP. The anti-von Willebrand factor (VWF) aptamer ARC1779 effectively inhibits VWF activity in plasma samples of TTP patients and thus shear-dependent platelet (PLT) function as measured by the PLT function analyzer PFA-100 (Dade Behring). It was hypothesized that ARC1779 would offer a potentially effective treatment option for a critically ill patient, refractory to standard care. CASE REPORT: A 39-year-old male patient with idiopathic TTP, refractory to daily plasma exchange, rituximab, steroids, and splenectomy, was additionally treated with a continuous infusion of the anti-von Willebrand factor (VWF) aptamer ARC1779 for 3 weeks. RESULTS: Plasma concentrations of approximately 10 microg/mL ARC1779 decreased VWF activity by more than 96%. Plasma exchange treatment acutely decreased the plasma concentrations of ARC1779 by a mean of 47% (range, 40%-61%). Thus, additional minibolus infusions of ARC1779 were given after each plasma exchange to rapidly restore steady-state concentrations. ARC1779 resulted in an increase of PLT counts as long as ARC1779 was given. On three occasions the infusion was stopped, each accompanied by a decrease in PLT counts and worsening of microangiopathy. No serious adverse effects were observed during the treatment with ARC1779. CONCLUSION: ARC1779 caused a clear and reproducible increase in PLT counts in an otherwise refractory TTP case. These clinical, pharmacokinetic, and pharmacodynamic data provide a rational basis for clinical trials with ARC1779 in TTP.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Fibrinolytic Agents/therapeutic use , Purpura, Thrombotic Thrombocytopenic/drug therapy , von Willebrand Factor/antagonists & inhibitors , Adult , Humans , Male , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/pathology
15.
Circulation ; 116(23): 2678-86, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18025536

ABSTRACT

BACKGROUND: ARC1779 is a therapeutic aptamer antagonist of the A1 domain of von Willebrand Factor (vWF), the ligand for receptor glycoprotein 1b on platelets. ARC1779 is being developed as a novel antithrombotic agent for use in patients with acute coronary syndromes. METHODS AND RESULTS: This was a randomized, double-blind, placebo-controlled study in 47 healthy volunteers of doses of ARC1779 from 0.05 to 1.0 mg/kg. Pharmacodynamic effects were measured by an ELISA for free vWF A1 binding sites and by a platelet function analyzer. In terms of pharmacokinetics, the concentration-time profile of ARC1779 appeared monophasic. The observed concentration and area under the curve were dose proportional. The mean apparent elimination half-life was approximately 2 hours, and mean residence time was approximately 3 hours. The mean apparent volumes of distribution (at steady state and during terminal phase) were approximately one half the blood volume, suggesting that ARC1779 distribution is in the central compartment. The mean clearance ranged from approximately 10% to approximately 21% of the glomerular filtration rate, suggesting that renal filtration may not be a major mechanism of clearance of ARC1779. Inhibition of vWF A1 binding activity was achieved with an EC(90) value of 2.0 mug/mL (151 nmol/L) and of platelet function with an EC(90) value of 2.6 mug/mL (196 nmol/L). ARC1779 was generally well tolerated, and no bleeding was observed. Adverse events tended to be minor and not dose related. CONCLUSIONS: This is the first-in-human evaluation of a novel aptamer antagonist of vWF. ARC1779 produced dose- and concentration-dependent inhibition of vWF activity and platelet function with duration of effect suitable for the intended clinical use in acute coronary syndromes.


Subject(s)
Acute Coronary Syndrome/drug therapy , Aptamers, Nucleotide/pharmacokinetics , Fibrinolytic Agents/pharmacokinetics , von Willebrand Factor/antagonists & inhibitors , Adolescent , Adult , Aged , Aptamers, Nucleotide/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fibrinolytic Agents/adverse effects , Humans , Male , Middle Aged , Platelet Function Tests , Platelet Glycoprotein GPIb-IX Complex/agonists , Protein Structure, Tertiary , Time Factors
16.
Nat Med ; 9(1): 47-52, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12483208

ABSTRACT

Cytokines can initiate and perpetuate human diseases, and are among the best-validated of therapeutic targets. Cytokines can be blocked by the use of soluble receptors; however, the use of this approach for cytokines such as interleukin (IL)-1, IL-4, IL-6 and IL-13 that use multi-component receptor systems is limited because monomeric soluble receptors generally exhibit low affinity or function as agonists. We describe here a generally applicable method to create very high-affinity blockers called 'cytokine traps' consisting of fusions between the constant region of IgG and the extracellular domains of two distinct cytokine receptor components involved in binding the cytokine. Traps potently block cytokines in vitro and in vivo and represent a substantial advance in creating novel therapeutic candidates for cytokine-driven diseases.


Subject(s)
Antigens, CD/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Immunoglobulin Fc Fragments/metabolism , Membrane Glycoproteins/metabolism , Receptors, Interleukin-6/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Cell Division/physiology , Cell Line , Cytokine Receptor gp130 , Cytokines/immunology , Dimerization , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Macaca fascicularis , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred Strains , Protein Binding , Random Allocation , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...