ABSTRACT
Phosphorothioate-modified antisense oligonucleotides (PS-ASOs) interact with a host of plasma, cell-surface and intracellular proteins which govern their therapeutic properties. Given the importance of PS backbone for interaction with proteins, we systematically replaced anionic PS-linkages in toxic ASOs with charge-neutral alkylphosphonate linkages. Site-specific incorporation of alkyl phosphonates altered the RNaseH1 cleavage patterns but overall rates of cleavage and activity versus the on-target gene in cells and in mice were only minimally affected. However, replacing even one PS-linkage at position 2 or 3 from the 5'-side of the DNA-gap with alkylphosphonates reduced or eliminated toxicity of several hepatotoxic gapmer ASOs. The reduction in toxicity was accompanied by the absence of nucleolar mislocalization of paraspeckle protein P54nrb, ablation of P21 mRNA elevation and caspase activation in cells, and hepatotoxicity in mice. The generality of these observations was further demonstrated for several ASOs versus multiple gene targets. Our results add to the types of structural modifications that can be used in the gap-region to enhance ASO safety and provide insights into understanding the biochemistry of PS ASO protein interactions.
Subject(s)
Cell Membrane/metabolism , Cytoplasm/metabolism , Oligonucleotides, Antisense/chemistry , Organophosphonates/chemistry , Phosphorothioate Oligonucleotides/chemistry , 3T3-L1 Cells , Animals , Caspases/metabolism , Cell Line , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , DNA-Binding Proteins , HeLa Cells , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred BALB C , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , Oligonucleotides, Antisense/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , Protein Binding , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease H/genetics , Ribonuclease H/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolismABSTRACT
Trans-differentiation of quiescent hepatic stellate cells (HSCs) into active myofibroblasts secretes excess amounts of extracellular matrix (ECM) proteins. miR-29b1 has the potential to treat liver fibrosis, because it targets several profibrotic genes. We previously demonstrated that miR-29b1 and the hedgehog (Hh) pathway inhibitor GDC-0449 could, together, inhibit the activation of HSCs and ECM production in common bile-duct-ligated (CBDL) mice. Herein, we determined the effect of chemical modifications of miR-29b1 on its stability, immunogenicity, and Argonaute-2 (Ago2) loading in vitro, after modifying its antisense strand with phosphorothioate (PS-miR-29b1), 2'-O-methyl-phosphorothioate (OMe-miR-29b1), locked nucleic acid (LNA-miR-29b1), and N,N'-diethyl-4-(4-nitronaphthalen-1-ylazo)-phenylamine (ZEN-miR-29b1). Chemical modifications significantly improved stability of miR-29b1 in 50% FBS. Among all the modified miRNAs tested, OMe-PS-miR-29b1 showed the highest stability with low immunogenicity, without the loss of efficacy in vitro. Therefore, OMe-PS-miR-29b1 was complexed with poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylenecarbonate-graft-dodecanol-graft-tetraethylenepentamine (mPEG-b-PCC-g-DC-g-TEPA) cationic micelles, and anti-fibrotic efficacy was evaluated in CBDL mice. There was a significant improvement in liver histology and decrease in the levels of injury markers. Further, mRNA/protein levels of collagen, α-SMA, and TIMP-1 were significantly lower for the OMe-PS-miR-29b1-loaded micelles compared to miR-29b1-loaded micelles. In conclusion, micellar delivery of OMe-PS-miR-29b1 is a promising strategy to treat liver fibrosis.
Subject(s)
Liver Cirrhosis/genetics , MicroRNAs/genetics , Phosphorothioate Oligonucleotides/genetics , Animals , Argonaute Proteins/metabolism , Base Sequence , Biomarkers , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gene Silencing , Humans , Inflammation Mediators/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Male , Mice , Micelles , Molecular Structure , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemistryABSTRACT
In this study, a series of derivatives of the amphipathic proline-rich sweet arrow peptide (SAP) were covalently linked to antisense oligonucleotides designed to inhibit Renilla luciferase gene. Oligonucleotide-peptide conjugates carrying lysine (Lys) and ornithine (Orn) residues were prepared using the stepwise approach by assembling first the peptide sequence followed by the assembly of the DNA molecule. The resulting Lys, Orn-conjugates were transformed to the corresponding arginine and homoarginine oligonucleotide-peptide conjugates by reaction with O-methylisourea. The introduction of the SAP at 3'-termini of a phosphorothioate oligonucleotide did not affect the ability to inhibit gene expression when transfected with lipofectamine. However, these conjugates were not able to enter cells without transfecting agent. Further studies using SAP as a transfection agent showed promising results for the conjugates carrying the Orn-SAP. All conjugates showed high duplex stabilities.
Subject(s)
Oligonucleotides, Antisense/chemistry , Peptides/chemistry , Phosphorothioate Oligonucleotides/chemistry , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Gene Expression , Genetic Vectors , Humans , Luciferases, Firefly/genetics , Luciferases, Renilla/genetics , Oligonucleotides, Antisense/administration & dosage , Peptides/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , Plasmids , Proline/chemistry , Transfection/methodsABSTRACT
BACKGROUND: TPI ASM8 contains two modified antisense oligonucleotides (AON) targeting the beta subunit (ß(c) ) of the IL-3, IL-5, GM-CSF receptors and the chemokine receptor CCR3. A previous study suggested that TPI ASM8 had broader effects than just inhibition of eosinophils in asthmatics. OBJECTIVE: We assessed whether TPI ASM8 caused a dose-dependent attenuation in the inflammatory and physiological changes after inhaled allergen challenge (AIC). METHODS: This single-center, open-label, stepwise-ascending dose study was conducted in fourteen stable, mild allergic asthmatics. Following placebo AIC, subjects underwent AIC after 4 days treatment with 1, 2, and 4 mg BID and finally 8 mg once daily (OD) of TPI ASM8, inhaled via the I-Neb™ nebuliser. Treatments were separated by 2-3-week washout periods. RESULTS: TPI ASM8 was safe and well tolerated at all doses. TPI ASM8 8 mg OD reduced eosinophils in sputum after AIC (by 60.9% at 7 h and 68.4% at 24 h post-AIC, P=0.016 and P=0.007, respectively). Additionally, TPI ASM8 8 mg OD significantly attenuated the early and late airway responses as shown by the reduction in the area under the curve by 45% (P=0.016) and 59%, (P=0.0015), respectively, the increase in eosinophil cationic protein (ECP) by up to 57% (P=0.021), and airway responsiveness to methacholine by more than 1 doubling dose (P=0.012). A dose-response relationship was noted, and efficacy was maintained with once per day administration. CONCLUSIONS: TPI ASM8 attenuated a broad range of inflammatory and physiological changes after AIC, suggesting that CCR3, IL-3, and GM-CSF also are important targets for the management of asthma.
Subject(s)
Allergens/immunology , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/immunology , Phosphorothioate Oligonucleotides/therapeutic use , Adolescent , Adult , Allergens/administration & dosage , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/pharmacokinetics , Asthma/genetics , Cytokine Receptor Common beta Subunit/genetics , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Humans , Male , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/adverse effects , Phosphorothioate Oligonucleotides/pharmacokinetics , RNA, Messenger/genetics , Receptors, CCR3/genetics , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology , Sputum/immunology , Young AdultABSTRACT
Molecular beacons (MBs) have shown great potential for the imaging of RNAs within single living cells; however, the ability to perform accurate measurements of RNA expression can be hampered by false-positives resulting from nonspecific interactions and/or nuclease degradation. These false-positives could potentially be avoided by introducing chemically modified oligonucleotides into the MB design. In this study, fluorescence microscopy experiments were performed to elucidate the subcellular trafficking, false-positive signal generation, and functionality of 2'-O-methyl (2Me) and 2'-O-methyl-phosphorothioate (2MePS) MBs. The 2Me MBs exhibited rapid nuclear sequestration and a gradual increase in fluorescence over time, with nearly 50% of the MBs being opened nonspecifically within 24 h. In contrast, the 2MePS MBs elicited an instantaneous increase in false-positives, corresponding to approximately 5-10% of the MBs being open, but little increase was observed over the next 24 h. Moreover, trafficking to the nucleus was slower. After 24 h, both MBs were localized in the nucleus and lysosomal compartments, but only the 2MePS MBs were still functional. When the MBs were retained in the cytoplasm, via conjugation to NeutrAvidin, a significant reduction in false-positives and improvement in functionality was observed. Overall, these results have significant implications for the design and applications of MBs for intracellular RNA measurement.
Subject(s)
Fluorescent Dyes/chemistry , Oligonucleotides, Antisense/chemistry , Phosphorothioate Oligonucleotides/chemistry , Animals , Avidin/chemistry , Biological Transport , Cells, Cultured , Mice , Microinjections , Microscopy, Fluorescence , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/metabolism , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/metabolism , RNA/analysisABSTRACT
The superfamily of cation/Ca2+ plasma-membrane exchangers contains two branches, the K+-independent Na+-Ca2+ exchangers (NCXs) and the K+-dependent Na+-Ca2+ exchangers (NCKXs), widely expressed in mammals. NCKX2 is the major neuronally expressed isoform among NCKX members. Despite its importance in maintaining Na+, Ca2+, and K+ homeostasis in the CNS, the role of NCKX2 during cerebral ischemia, a condition characterized by an alteration of ionic concentrations, has not yet been investigated. The present study examines NCKX2 role in the development of ischemic brain damage in permanent middle cerebral artery occlusion (pMCAO) and transient middle cerebral artery occlusion. Furthermore, to evaluate the effect of nckx2 ablation on neuronal survival, nckx2-/- primary cortical neurons were subjected to oxygen glucose deprivation plus reoxygenation. NCKX2 mRNA and protein expression was evaluated in the ischemic core and surrounding ipsilesional areas, at different time points after pMCAO in rats. In ischemic core and in periinfarctual area, NCKX2 mRNA and protein expression were downregulated. In addition, NCKX2 knock-down by antisense oligodeoxynucleotide and NCKX2 knock-out by genetic disruption dramatically increased infarct volume. Accordingly, nckx2-/- primary cortical neurons displayed a higher vulnerability and a greater [Ca2+]i increase under hypoxic conditions, compared with nckx2+/+ neurons. In addition, NCKX currents both in the forward and reverse mode of operation were significantly reduced in nckx2-/- neurons compared with nckx2+/+ cells. Overall, these results indicate that NCKX2 is involved in brain ischemia, and it may represent a new potential target to be investigated in the study of the molecular mechanisms involved in cerebral ischemia.
Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Sodium-Calcium Exchanger/metabolism , Analysis of Variance , Animals , Autoradiography , Blood Gas Analysis/methods , Blood Pressure/genetics , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Calcium/metabolism , Cerebral Cortex/pathology , Cerebrovascular Circulation/genetics , Cerebrovascular Circulation/physiology , Disease Models, Animal , In Vitro Techniques , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Patch-Clamp Techniques/methods , Phosphopyruvate Hydratase/metabolism , Phosphorothioate Oligonucleotides/administration & dosage , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/geneticsABSTRACT
BACKGROUND: Antisense-mediated exon skipping is a putative treatment for Duchenne muscular dystrophy (DMD). Using antisense oligonucleotides (AONs), the disrupted DMD reading frame is restored, allowing generation of partially functional dystrophin and conversion of a severe Duchenne into a milder Becker muscular dystrophy phenotype. In vivo studies are mainly performed using 2'-O-methyl phosphorothioate (2OMePS) or morpholino (PMO) AONs. These compounds were never directly compared. METHODS: mdx and humanized (h)DMD mice were injected intramuscularly and intravenously with short versus long 2OMePS and PMO for mouse exon 23 and human exons 44, 45, 46 and 51. RESULTS: Intramuscular injection showed that increasing the length of 2OMePS AONs enhanced skipping efficiencies of human exon 45, but decreased efficiency for mouse exon 23. Although PMO induced more mouse exon 23 skipping, PMO and 2OMePS were more comparable for human exons. After intravenous administration, exon skipping and novel protein was shown in the heart with both chemistries. Furthermore, PMO showed lower intramuscular concentrations with higher exon 23 skipping levels compared to 2OMePS, which may be due to sequestration in the extracellular matrix. Finally, two mismatches rendered 2OMePS but not PMO AONs nearly ineffective. CONCLUSIONS: The results obtained in the present study indicate that increasing AON length improves skipping efficiency in some but not all cases. It is feasible to induce exon skipping and dystrophin restoration in the heart after injection of 2OMePS and unconjugated PMO. Furthermore, differences in efficiency between PMO and 2OMePS appear to be sequence and not chemistry dependent. Finally, the results indicate that PMOs may be less sequence specific than 2OMePS.
Subject(s)
Exons/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Muscular Dystrophy, Duchenne , Oligonucleotides, Antisense , Phosphorothioate Oligonucleotides , Animals , Base Sequence , Humans , Mice , Mice, Inbred mdx , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Myocardium/cytology , Myocardium/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/geneticsABSTRACT
Nuclear factor-kappaB (NF-kappaB) plays a key role in the expression of several genes involved in the immune and inflammatory process. Previously, we demonstrated that NF-kappaB activation can be significantly inhibited by a double stranded oligodeoxynucleotide (ODN). Nevertheless, the therapeutic use of ODN requires a delivery system able to improve poor crossing of cell membranes and rapid in vivo enzymatic degradation. Poly(D,L-lactide-co-glycolide) (PLGA) microspheres can increase ODN stability in biological environment and release the encapsulated drug in long time frames. Here, we used a decoy ODN against NF-kappaB and we investigated its effect, when administered in naked form or when delivered by PLGA microspheres, in a rat model of chronic inflammation. The subcutaneous implant of lambda-carrageenin-soaked sponges caused leukocyte infiltration and formation of granulation tissue which were inhibited up to 15 days by co-injection of microspheres releasing decoy ODN whereas naked decoy ODN showed this effect only up to 5 days. Molecular analysis performed on granulation tissue demonstrated an inhibition of NF-kappaB activation correlated to a decrease of tumor necrosis factor-alpha (TNF-alpha) and inducible nitric oxide synthase (iNOS) expression. Our results suggest that microspheres could be an useful tool to improve pharmacokinetics of decoy ODN and may represent a strategy to inhibit NF-kappaB activation in chronic inflammation.
Subject(s)
Drug Carriers/chemistry , Inflammation/drug therapy , Lactic Acid/chemistry , NF-kappa B/metabolism , Phosphorothioate Oligonucleotides/therapeutic use , Polyglycolic Acid/chemistry , Animals , Blotting, Western , Chronic Disease , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Granulation Tissue/drug effects , Granulation Tissue/metabolism , Inflammation/metabolism , Male , Microspheres , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Phosphorothioate Oligonucleotides/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesisABSTRACT
RATIONALE: The drug product TPI ASM8 contains two modified phosphorothioate antisense oligonucleotides designed to inhibit allergic inflammation by down-regulating human CCR3 and the common beta chain (beta(c)) of IL-3, IL-5, and granulocyte-macrophage colony-stimulating factor receptors. OBJECTIVES: This study examined the effects of inhaled TPI ASM8 on sputum cellular influx, CCR3 and beta(c) mRNA and protein levels, and the airway physiologic response after inhaled allergen. METHODS: Seventeen subjects with mild atopic asthma were randomized in a crossover study to inhale 1,500 microg TPI ASM8 or placebo by nebulizer, once daily for 4 days. On Day 3, subjects underwent allergen inhalation challenge. Sputum samples were collected before and after allergen. CCR3 and beta(c) protein levels were measured by flow cytometry, mRNA was measured using real-time quantitative polymerase chain reaction, and the FEV1 was measured over 7 hours after challenge. MEASUREMENTS AND MAIN RESULTS: Compared with placebo, TPI ASM8 inhibited sputum eosinophil influx by 46% (P = 0.02) and blunted the increase in total cells (63%) after allergen challenge. TPI ASM8 significantly reduced the early asthmatic response (P = 0.04) with a trend for the late asthmatic response (P = 0.08). The allergen-induced (Day 2 to Day 3) levels of beta(c) mRNA and CCR3 mRNA in sputum-derived cells were inhibited by TPI ASM8 (P = 0.039 and P = 0.054, respectively), with no significant effects on the cell surface protein expression of CCR3 and beta(c) (P > 0.05). No serious adverse events were reported. CONCLUSIONS: TPI ASM8 attenuates the allergen-induced increase in target gene mRNA and airway responses in subjects with mild asthma. Clinical trial registered with www.clinicaltrials.gov (NCT 00264966).
Subject(s)
Allergens/adverse effects , Asthma/drug therapy , Oligonucleotides, Antisense/therapeutic use , Phosphorothioate Oligonucleotides/therapeutic use , Pulmonary Eosinophilia/drug therapy , Receptors, CCR3/antagonists & inhibitors , Receptors, Cytokine/metabolism , Administration, Inhalation , Adult , Asthma/genetics , Asthma/metabolism , Cross-Over Studies , Double-Blind Method , Drug Combinations , Female , Flow Cytometry , Follow-Up Studies , Forced Expiratory Volume , Gene Expression , Humans , Male , Middle Aged , Nebulizers and Vaporizers , Oligonucleotides, Antisense/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/metabolism , RNA, Messenger/genetics , Receptors, CCR3/genetics , Receptors, CCR3/metabolism , Receptors, Cytokine/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sputum/cytology , Sputum/metabolism , Treatment OutcomeABSTRACT
Advances in medicinal chemistry have produced new chemical classes of antisense oligonucleotides (ASOs) with enhanced therapeutic properties. Conjugation of the triantennary N-acetylgalactosamine (GalNAc3) moiety to the extensively characterized phosphorothioate (PS)-modified 2'-O-methoxyethyl (2'MOE) ASO exemplifies such an advance. This structure-activity optimized moiety effects receptor-mediated uptake of the ASO prodrug through the asialoglycoprotein receptor 1 to support selective targeting of RNAs expressed by hepatocytes. In this study we report the integrated assessment of data available from randomized placebo-controlled dose-ranging studies of this chemical class of ASOs administered systemically to healthy human volunteers. First, we compare the pharmacokinetic and pharmacodynamic profiles of a subset of the GalNAc3-conjugated PS-modified 2'MOE ASOs to the parent PS-modified 2'MOE ASOs for which plasma analytes are available. We then evaluate the safety profile of the full set of GalNAc3-conjugated PS-modified 2'MOE ASO conjugates by the incidence of signals in standardized laboratory tests and by the mean laboratory test results as a function of dose level over time. With hepatocyte targeted delivery, the ED50 for the GalNAc3-conjugated PS-modified 2'MOE ASO subset ranges from 4 to 10 mg/week, up to 30-fold more potent than the parent PS-modified 2'MOE ASO. No GalNAc3-conjugated PS-modified 2'MOE ASO class effects were identified from the assessment of the integrated laboratory test data across all doses tested with either single or multidose regimens. The increase in potency supports an increase in the safety margin for this new chemical class of ASOs now under broad investigation in the clinic. Although the total exposure is limited in the initial phase 1 trials, ongoing and future investigations in patient populations will support evaluation of the effects of long-term exposure.
Subject(s)
Acetylgalactosamine/administration & dosage , Asialoglycoprotein Receptor/genetics , Oligonucleotides, Antisense/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , Acetylgalactosamine/blood , Acetylgalactosamine/pharmacokinetics , Asialoglycoprotein Receptor/blood , Biomarkers, Pharmacological/blood , Dose-Response Relationship, Drug , Female , Healthy Volunteers , Hepatocytes/drug effects , Humans , Male , Middle Aged , Oligonucleotides, Antisense/blood , Oligonucleotides, Antisense/pharmacokinetics , Phosphorothioate Oligonucleotides/blood , Phosphorothioate Oligonucleotides/pharmacokinetics , RNA/antagonists & inhibitors , RNA/blood , RNA/genetics , Structure-Activity RelationshipABSTRACT
Enzyme kinetics of GTI-2040 (5'-GGC TAA ATC GCT CCA CCA AG-3'), a phosphorothioate ribonucleotide reductase antisense, were investigated for the first time in 3' exonuclease solution and human liver microsomes (HLMs), using the ion-pair high-performance liquid chromatogram method for quantification of the parent drug and two major 3'N-1 and 3'N-2 metabolites. Enzyme kinetics of GTI-2040 in 3'-exonuclease solution were found to be well characterized by the Michaelis-Menten model, using the sum of formation rates of 3'N-1 and 3'N-2 (approximately total metabolism) because of sequential metabolism. In HLMs, a biphasic binding was observed for GTI-2040 with high- and low-affinity constants (K(d)s) of 0.03 and 3.8 microM, respectively. Enzyme kinetics of GTI-2040 in HLMs were found to deviate from Michaelis-Menten kinetics when the total GTI-2040 substrate was used. However, after correction for the unbound fractions, the formation rate of total metabolites could be described by Michaelis-Menten kinetics. Using the free substrate fraction, the K(m) and V(max) of GTI-2040 were determined to be 6.33 +/- 3.2 microM and 16.5 +/- 8.4 nmol/mg/h, respectively. Using these values, in vitro hepatic intrinsic clearance (CL(int)) in HLM was estimated to be 2.61 +/- 0.56 ml/h. The CL(int) was then used to predict GTI-2040's in vivo intrinsic clearance in humans by a microsomal protein scaling factor, which gave a mean value of 182.7 l/h, representing 24.1% of the observed in vivo mean scaled hepatic intrinsic clearance of 758.7 l/h in patients with acute myeloid leukemia. We concluded that the saturable nonspecific binding of GTI-2040 in HLMs complicated the interpretation of its enzyme kinetics, and scaled intrinsic clearance from HLMs only partially predicted the in vivo intrinsic clearance.
Subject(s)
Drug Delivery Systems/methods , Oligodeoxyribonucleotides/metabolism , Phosphorothioate Oligonucleotides/metabolism , Ribonucleotide Reductases/metabolism , Animals , Crotalus , Female , Humans , Kidney/drug effects , Kidney/enzymology , Kidney/metabolism , Kinetics , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/pharmacology , Ribonucleotide Reductases/antagonists & inhibitorsABSTRACT
Treatment of systemic disease with phosphorothioate antisense oligonucleotides (PS ASOs) has been accomplished using local or parenteral routes of administration to date. This report describes, for the first time, the effective oral delivery of a second generation oligonucleotide where significant milligram amounts of intact drug are absorbed in human subjects. In this study, a variety of oral solid dosage formulations were evaluated and it was determined that pulsing the delivery of sodium caprate (C10), a well-known permeation enhancer, in a novel manner may provide optimal ASO plasma bioavailability. Further, these dosage forms, containing C10 and ASO, were well tolerated in both fasted and fed volunteers. Oral absorption of the 2'-O-(2-methoxyethyl) modified antisense oligonucleotide (2'-MOE ASO), ISIS 104838, was demonstrated in healthy volunteers with an average 9.5% plasma bioavailability across four formulations tested. The greatest average performance achieved in this study for a single formulation was 12.0% bioavailability within an individual dose and subject range of 1.96-27.5%. The totality of the data suggests that formulations can be devised that allow oral administration of oligonucleotides that maintain systemic concentrations associated with inhibition of targeted human mRNA.
Subject(s)
Phosphorothioate Oligonucleotides/administration & dosage , Administration, Oral , Adolescent , Adult , Area Under Curve , Biological Availability , Capsules , Chemistry, Pharmaceutical , Cross-Over Studies , Delayed-Action Preparations , Gamma Cameras , Gastric Emptying , Humans , Male , Middle Aged , Oligoribonucleotides/chemistry , Oxides/chemistry , Phosphorothioate Oligonucleotides/adverse effects , Phosphorothioate Oligonucleotides/pharmacokinetics , Samarium/chemistry , SolubilityABSTRACT
Cubic membranes are soft three-dimensional crystals found within cell organelles in a variety of living systems, despite the aphorism of Fedorov: 'crystallization is death'. They consist of multi-bilayer lipid-protein stacks, folded onto anticlastic surfaces that resemble triply periodic minimal surfaces, forming highly swollen crystalline sponges. Although cubic membranes have been observed in numerous cell types and under different pathophysiological conditions, knowledge about the formation and potential function(s) of non-lamellar, cubic structures in biological systems is scarce. We report that mitochondria with this cubic membrane organization isolated from starved amoeba Chaos carolinense interact sufficiently with short segments of phosphorothioate oligonucleotides (PS-ODNs) to give significant ODNs uptake. ODNs condensed within the convoluted channels of cubic membrane by an unknown passive targeting mechanism. Moreover, the interaction between ODNs and cubic membrane is sufficient to retard electrophoretic mobility of the ODN component in the gel matrix. These ODN-cubic membrane complexes are readily internalized within the cytoplasm of cultured mammalian cells. Transmission electron microscopic analysis confirms ODNs uptake by cubic membranes and internalization of ODN-cubic membrane complexes into the culture cells. Cubic membranes thus may offer a new, potentially benign medium for gene transfection.
Subject(s)
Amoeba/chemistry , DNA/chemistry , Mitochondrial Membranes/chemistry , Phosphorothioate Oligonucleotides/chemistry , Animals , Cell Line, Tumor , DNA/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Phosphorothioate Oligonucleotides/administration & dosage , TransfectionABSTRACT
Antisense oligonucleotides (AONs) are versatile molecules that can be used to modulate gene expression by binding to RNA. The therapeutic potential of AONs appears particularly high in the central nervous system, due to excellent distribution and uptake in brain cells, as well as good tolerability in clinical trials thus far. Nonetheless, immune stimulation in response to AON treatment in the brain remains a concern. For this reason we performed RNA sequencing analysis of brain tissue from mice treated intracerebroventricularly with phosphorothioate, 2'-O-methyl modified AONs. A significant upregulation of immune system associated genes was observed in brains of AON treated mice, with the striatum showing largest transcriptional changes. Strongest upregulation was seen for the antiviral enzyme 2'-5'-oligoadenylate synthase-like protein 2 (Oasl2) and Bone marrow stromal antigen 2 (Bst2). Histological analysis confirmed activation of microglia and astrocytes in striatum. The upregulation of immune system associated genes was detectable for at least 2 months after the last AON administration, consistent with a continuous immune response to the AON.
Subject(s)
Brain/drug effects , Immunity, Innate/drug effects , Infusions, Intraventricular , Oligonucleotides, Antisense/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Astrocytes/drug effects , Brain/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , RNA/genetics , RNA/metabolism , Sequence Analysis, RNAABSTRACT
Efficiency of polyethylenimine (PEI) for nucleic acid delivery is affected by the size of the carrier and length of the nucleic acids. For instance, PEIs with molecular weights between 10-30kDa provide optimal DNA delivery activity whereas PEIs with molecular weights below 1.8kDa are ineffective. The activity of PEI is also severely diminished by substitution of DNA for shorter nucleic acids such as mRNA or siRNA. Here, through chemical modification of the primary amines to aromatic domains we achieved nucleic acid delivery by the 1.8kDa polyethylenimine (PEI) particles. This modification did not affect the PEI buffering abilities but enhanced its pH-sensitive aggregation, enabling stabilization of the polyplex outside the cell while still allowing nucleic acid release following cellular entry. The aromatic PEIs were then evaluated for their gene, mRNA, siRNA and 2'O-methyl phosphorothioate oligonucleotide in vitro transfection abilities. The salicylamide-grafted PEI showed to be a reliable carrier for delivering nucleic acids with cytoplasmic activity such as the mRNA and siRNA or nuclear diffusible oligonucleotide. It was then further equipped with polyethyleneglycol (PEG) and the delivery efficiency of the copolymer was tested in vivo for regeneration of dystrophin in the muscle of mdx mouse through a 2'O-methyl phosphorothioate-mediated splicing modulation. Intramuscular administration of polyplexes resulted in dystrophin-positive fibers in a mouse model of Duchenne muscular dystrophy without apparent toxicity. These findings indicate that precise modifications of low molecular weight PEI improve its bio-responsiveness and yield delivery vehicles for nucleic acids of various types in vitro and in vivo.
Subject(s)
DNA/administration & dosage , Gene Transfer Techniques , Phosphorothioate Oligonucleotides/administration & dosage , Plasmids/administration & dosage , Polyethyleneimine/chemistry , RNA, Messenger/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , DNA/genetics , Exons , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice, Inbred mdx , Phosphorothioate Oligonucleotides/genetics , Plasmids/genetics , RNA, Messenger/genetics , RNA, Small Interfering/geneticsABSTRACT
Phosphorothioate (PS) modified antisense oligonucleotides (ASOs) have progressed rapidly in the clinic for treating a variety of disease indications. We previously demonstrated that the activity of PS ASOs in the liver can be enhanced by co-infusion of an excipient oligonucleotide (EON). It was posited that the EON saturates a nonproductive uptake pathway(s) thereby permitting accumulation of the PS ASO in a productive tissue compartment. In this report, we measured PS ASO activity following administration by bolus, infusion or co-fusion with EON within hepatocytes and nonparenchymal cells (NPCs), of the liver. This revealed that while ASOs accumulate preferentially in NPCs, they are intrinsically more active in hepatocytes. Furthermore, we show that the EON enhances ASO potency when infused up to 72 h before or after administration of the active ASO suggesting that the EON can saturate and displace the ASO from nonproductive to productive compartments. Physical presence of the EON in tissues was required for optimal potentiation suggesting that there is a dynamic distribution of the ASO and EON between the compartments. Lastly, using a candidate approach, we confirmed Stabilin-2 as a molecular pathway for ASO uptake in sinusoidal endothelial cells and the ASGR as a pathway for ASO uptake into hepatocytes in the liver.
Subject(s)
Excipients/pharmacokinetics , Liver/metabolism , Oligonucleotides, Antisense/pharmacokinetics , Phosphorothioate Oligonucleotides/pharmacokinetics , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , Coculture Techniques , Endothelial Cells/metabolism , Excipients/administration & dosage , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/cytology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligonucleotides, Antisense/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , Tissue DistributionABSTRACT
Single-stranded (ss) 2'-fluoro (2'-F)-modified oligonucleotides (ONs) with a full phosphorothioate (PS) backbone have been reported to be cytotoxic and cause DNA double-strand breaks (DSBs) when transfected into HeLa cells. However, the molecular determinants of these effects have not been fully explored. In this study, we investigated the impact of ON structure, chemistry, delivery method, and cell type on in vitro cytotoxicity and DSBs. We found that ss PS-ONs were more cytotoxic than double-stranded (ds) PS-ONs, irrespective of the 2'-ribose chemistry, inclusive of the 2'-F modification. Cytotoxicity of ss ONs was most affected by the total PS content, with an additional contribution of 2'-F substitutions in HeLa, but not HepG2, cells. The relatively mild cytotoxicity of ds ONs was most impacted by long contiguous PS stretches combined with 2'-F substitutions. None of the tested ds 2'-F-modified PS-ONs caused DSBs, while the previously reported DSBs caused by ss 2'-F-modified PS-ONs were PS dependent. HeLa cells were more sensitive to ON-mediated toxicity when transfected with Lipofectamine 2000 versus Lipofectamine RNAiMax. Importantly, asialoglycoprotein receptor-mediated uptake of N-acetylgalactosamine-conjugated ss or ds PS-ONs, even those with long PS stretches and high 2'-F content, was neither cytotoxic nor caused DSBs at transfection-equivalent exposures. These results suggest that in vitro cytotoxicity and DSBs associated with ONs are delivery method dependent and primarily determined by single-stranded nature and PS content of ONs.
Subject(s)
DNA Breaks, Double-Stranded , Oligoribonucleotides, Antisense/toxicity , Phosphorothioate Oligonucleotides/toxicity , RNA, Small Interfering/toxicity , Asialoglycoprotein Receptor/chemistry , Asialoglycoprotein Receptor/metabolism , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Survival , Drug Delivery Systems , HeLa Cells , Hep G2 Cells , Humans , Lipids/chemistry , Nanoconjugates/administration & dosage , Nuclear Proteins/metabolism , Oligoribonucleotides, Antisense/chemistry , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemistry , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , RNA-Binding Proteins/metabolism , TransfectionABSTRACT
PURPOSE: The aim of the study is to determine whether delivery into tumor cells in vivo of a 99mTc-labeled antisense phosphorothioate DNA targeting the mdr1 mRNA improves after electrostatic complexation with the transmembrane transfector (TF) carriers Neophectin or jetPEI as was observed by us in vitro. METHODS: The biodistribution of the labeled antisense DNA before and after complexation with either TF was determined in nude mice bearing KB-G2 (Pgp++) tumors. RESULTS: Complexation with either TF resulted in significantly higher background radioactivity levels in almost all normal tissues and modest improvement in tumor accumulation at best. The tumor accumulation was lower compared to naked at six hours (0.34 and 0.23 vs. 0.40% ID/g) and modestly higher at 24-28 hours (0.15 and 0.15 vs. 0.12% ID/g) for Neophectin and jetPEI, respectively. That blood was less than 0.18% ID/g for both TFs even at six hours suggests that tumor accumulations may have suffered from rapid blood clearance. CONCLUSION: The results of this investigation show that because of the unfavorable pharmacokinetics of radiolabeled phosphorothioate DNAs when electrostatically complexed to jetPEI or Neophectin, neither TF appears to be useful in vivo despite favorable results in vitro. Future studies will devote greater consideration to the relative rates of tumor accumulation and blood clearance.
Subject(s)
DNA, Antisense/administration & dosage , Drug Delivery Systems , Genetic Therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Organotechnetium Compounds/metabolism , Animals , Humans , KB Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorothioate Oligonucleotides/administration & dosageABSTRACT
The effects of repeated complement activation in cynomolgus monkeys after chronic antisense oligonucleotide (ASO) treatment were evaluated by using ISIS 104838, a representative 2'-O-methoxyethyl (2'-MOE) modified ASO. The treatment was up to 9 months with a total weekly dose of 30 mg/kg, given either as daily [4.3 mg/kg/day, subcutaneous (s.c.) injection] or once weekly [30 mg/kg, either as s.c. injection or 30-min intravenous (i.v.) infusion]. Acute elevations of complement split products (Bb and C3a) and a transient decrease in C3 occurred after the first dose and were drug plasma concentration dependent. However, with repeated complement activation after chronic ASO treatment, there were progressive increases in basal (predose) levels of Bb and C3a, and a sustained C3 reduction in all treated groups. There was also a progressive increase in C3d-bound circulating immune complex (CIC) that was considered secondary to the C3 depletion. Evidence of vascular inflammation was observed, mostly in the liver, kidney, and heart, and correlated with severe C3 depletion and increases in plasma IgG and IgM. Vascular inflammation was accompanied by increased C3 and IgM immunereactivity in the affected vasculatures and endothelial activation markers in serum. In summary, repeated complement activations in monkeys lead to a sustained decrease in circulating C3 over time. The concomitantly increased inflammatory signals and decreased CIC clearance due to impairment of complement function may lead to vascular inflammation after chronic ASO treatment in monkeys. However, based on the known sensitivity of monkeys to ASO-induced complement activation, these findings have limited relevance to humans.
Subject(s)
Complement Activation/drug effects , Oligonucleotides, Antisense/administration & dosage , Phosphorothioate Oligonucleotides/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Animals , Dose-Response Relationship, Drug , Heart/drug effects , Humans , Kidney/drug effects , Liver/drug effects , Macaca fascicularis , Oligonucleotides, Antisense/chemistry , Phosphorothioate Oligonucleotides/chemistry , Tumor Necrosis Factor-alpha/antagonists & inhibitorsABSTRACT
BACKGROUND: The application of newer signaling pathway-targeted agents has become an important addition to chemotherapy in the treatment of advanced non-small cell lung cancer (NSCLC). In this study, we evaluated the efficacy and toxicities of PKC inhibitors combined with chemotherapy versus chemotherapy alone for patients with advanced NSCLC systematically. PATIENTS AND MATERIALS: Literature retrieval, trials selection and assessment, data collection, and statistic analysis were performed according to the Cochrane Handbook 5.1.0. The outcome measures were tumor response rate, disease control rate, progression-free survival (PFS), overall survival (OS), and adverse effects. RESULTS: Five randomized controlled trials, comprising totally 1,005 patients, were included in this study. Meta-analysis showed significantly decreased response rate (RR 0.79; 95 % CI 0.64-0.99) and disease control rate (RR 0.90; 95 % CI 0.82-0.99) in PKC inhibitors-chemotherapy groups versus chemotherapy groups. There was no significant difference between the two treatment groups regarding progression-free survival (PFS, HR 1.05; 95 % CI 0.91-1.22) and overall survival (OS, HR 1.00; 95 % CI 0.86-1.16). The risk of grade 3/4 neutropenia, leucopenia, and thrombosis/embolism increased significantly in PKC inhibitors combination groups as compared with chemotherapy alone groups. CONCLUSION: The use of PKC inhibitors in addition to chemotherapy was not a valid alternative for patients with advanced NSCLC.