Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 48(10): 5235-5253, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32356888

ABSTRACT

Antisense oligonucleotides (ASOs) interact with target RNAs via hybridization to modulate gene expression through different mechanisms. ASO therapeutics are chemically modified and include phosphorothioate (PS) backbone modifications and different ribose and base modifications to improve pharmacological properties. Modified PS ASOs display better binding affinity to the target RNAs and increased binding to proteins. Moreover, PS ASO protein interactions can affect many aspects of their performance, including distribution and tissue delivery, cellular uptake, intracellular trafficking, potency and toxicity. In this review, we summarize recent progress in understanding PS ASO protein interactions, highlighting the proteins with which PS ASOs interact, the influence of PS ASO protein interactions on ASO performance, and the structure activity relationships of PS ASO modification and protein interactions. A detailed understanding of these interactions can aid in the design of safer and more potent ASO drugs, as illustrated by recent findings that altering ASO chemical modifications dramatically improves therapeutic index.


Subject(s)
Phosphorothioate Oligonucleotides/chemistry , Proteins/chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Intracellular Space/chemistry , Intracellular Space/metabolism , Ligands , Phosphorothioate Oligonucleotides/metabolism , Phosphorothioate Oligonucleotides/pharmacology , Phosphorothioate Oligonucleotides/toxicity , Protein Binding , Protein Domains , Proteins/metabolism , Proteins/toxicity , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/metabolism
2.
J Thromb Haemost ; 15(5): 983-997, 2017 05.
Article in English | MEDLINE | ID: mdl-28296036

ABSTRACT

Essentials CpG oligodeoxynucleotide (ODN) immuotherapeutics cause undesired platelet activating effects. It is crucial to understand the mechanisms of these effects to identify protective strategies. CpG ODN-induced platelet activation depends on C-type lectin-like receptor 2 (CLEC-2) and P2Y12. Targeting CLEC-2 or P2Y12 fully prevents CpG ODN-induced platelet activation and thrombosis. SUMMARY: Background Synthetic phosphorothioate-modified CpG oligodeoxynucleotides (ODNs) show potent immunostimulatory properties that are widely exploited in clinical trials of anticancer treatment. Unexpectedly, a recent study indicated that CpG ODNs activate human platelets via the immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptor glycoprotein VI. Objective To further analyze the mechanisms of CpG ODN-induced platelet activation and identify potential inhibitory strategies. Methods In vitro analyses were performed on human and mouse platelets, and on cell lines expressing platelet ITAM receptors. CpG ODN platelet-activating effects were evaluated in a mouse model of thrombosis. Results We demonstrated platelet uptake of CpG ODNs, resulting in platelet activation and aggregation. C-type lectin-like receptor 2 (CLEC-2) expressed in DT40 cells bound CpG ODNs. CpG ODN uptake did not occur in CLEC-2-deficient mouse platelets. Inhibition of human CLEC-2 with a blocking antibody inhibited CpG ODN-induced platelet aggregation. CpG ODNs caused CLEC-2 dimerization, and provoked its internalization. They induced dense granule release before the onset of aggregation. Accordingly, pretreating platelets with apyrase, or inhibiting P2Y12 with cangrelor or clopidogrel, prevented CpG ODN platelet-activating effect. In vivo, intravenously injected CpG ODN interacted with platelets adhered to mouse injured endothelium, and promoted thrombus growth, which was inhibited by CLEC-2 deficiency or by clopidogrel. Conclusions CLEC-2 and P2Y12 are required for CpG ODN-induced platelet activation and thrombosis, and might be targeted to prevent adverse events in patients at risk.


Subject(s)
Antibodies/pharmacology , Blood Platelets/drug effects , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Phosphorothioate Oligonucleotides/toxicity , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/toxicity , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Line , Disease Models, Animal , Humans , Immunoreceptor Tyrosine-Based Activation Motif , Lectins, C-Type/deficiency , Lectins, C-Type/immunology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Phosphorothioate Oligonucleotides/immunology , Phosphorothioate Oligonucleotides/metabolism , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Receptors, Purinergic P2Y12/metabolism , Signal Transduction/drug effects , Thrombosis/blood , Thrombosis/immunology , Thrombosis/prevention & control , Time Factors
3.
Nucleic Acid Ther ; 27(1): 11-22, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27923110

ABSTRACT

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 , Transfection
4.
Curr Drug Targets ; 15(7): 663-73, 2014.
Article in English | MEDLINE | ID: mdl-24655142

ABSTRACT

BACKGROUND: Many experimental and clinical studies have focused on the antisense strategy. In this context phosphorothioate oligonucleotides are compounds addressed to hybridize to a targeted mRNA inducing a variety of effects including inhibition of the expression of proteins involved in different pathological processes and preventing translation. METHODS: In this review, we provide an update on clinical efficacy and toxicological profile of phosphorothioate oligonucleotides used in experimental and clinical studies, also focusing on the use of the antisense strategy in the context of Duchenne muscular dystrophy which is a key pathology to study different aspects of this therapy. Pubmed/Medline was searched using the keyword "Phosphorotioate" combined with "Antisense", "Oligonucleotide" and "Duchenne muscular dystrophy". CONCLUSIONS: Phosphorothioate oligonucleotide transient activation of the complement cascade represents the most evident toxicological response, as showed by in vivo studies. It is also known that many of these compounds induce a prolongation of activated partial thromboplastin time, a reaction which is often highly transient and proportional to the oligonucleotide plasma concentrations, making that effect clinically insignificant for the current treatment regimens. In summary, current evidence shows limited untoward effects and reversibility of the damage induced, at least for some of those compounds, with promising effectiveness for treatment of various pathologies.


Subject(s)
Phosphorothioate Oligonucleotides/toxicity , Phosphorothioate Oligonucleotides/therapeutic use , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemistry , Treatment Outcome
5.
Nucleic Acid Ther ; 23(5): 302-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23952551

ABSTRACT

Antisense oligonucleotides that recruit RNase H and thereby cleave complementary messenger RNAs are being developed as therapeutics. Dose-dependent hepatic changes associated with hepatocyte necrosis and increases in serum alanine-aminotransferase levels have been observed after treatment with certain oligonucleotides. Although general mechanisms for drug-induced hepatic injury are known, the characteristics of oligonucleotides that determine their hepatotoxic potential are not well understood. Here, we present a comprehensive analysis of the hepatotoxic potential of locked nucleic acid-modified oligonucleotides in mice. We developed a random forests classifier, in which oligonucleotides are regarded as being composed of dinucleotide units, which distinguished between 206 oligonucleotides with high and low hepatotoxic potential with 80% accuracy as estimated by out-of-bag validation. In a validation set, 17 out of 23 oligonucleotides were correctly predicted (74% accuracy). In isolation, some dinucleotide units increase, and others decrease, the hepatotoxic potential of the oligonucleotides within which they are found. However, a complex interplay between all parts of an oligonucleotide can influence the hepatotoxic potential. Using the classifier, we demonstrate how an oligonucleotide with otherwise high hepatotoxic potential can be efficiently redesigned to abate hepatotoxic potential. These insights establish analysis of sequence and modification patterns as a powerful tool in the preclinical discovery process for oligonucleotide-based medicines.


Subject(s)
Alanine Transaminase/blood , Drug Design , Liver/drug effects , Oligonucleotides, Antisense/toxicity , Oligonucleotides/toxicity , Phosphorothioate Oligonucleotides/toxicity , Algorithms , Animals , Body Weight , Female , Liver/pathology , Mice , Mice, Inbred C57BL , Nucleic Acid Conformation , Oligonucleotides/administration & dosage , Oligonucleotides/chemical synthesis , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/chemical synthesis , Organ Size , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemical synthesis , Predictive Value of Tests , Quantitative Structure-Activity Relationship
6.
Nucleic Acid Ther ; 23(3): 213-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23692080

ABSTRACT

ISIS 481464 is a constrained ethyl (cEt) modified phosphorothioate antisense oligonucleotide (ASO) targeting signal transducer and activator of transcription 3 (STAT3) studied in mice and monkey to support oncology clinical trials. Six-week toxicology studies were performed in mice and cynomolgus monkey (up to 70 and 30 mg/kg/week respectively). Reduction in STAT3 protein up to 90% of control was observed in monkey. Cynomolgus monkey was considered the most relevant species to human with respect to pharmacokinetic properties, but mice are useful in their relative sensitivity to the potential proinflammatory and hepatic effects of oligonucleotides. In monkeys, there was no impact on organ function at doses up to 30 mg/kg/week for 6 weeks. Minimal to slight proximal tubular epithelial cell degeneration and regeneration within the kidney was observed, which had no impact on renal function and showed reversibility at the end of the treatment-free period. Additionally, mild and transient activated partial thromboplastin time elevations and mild increases in complement Bb were observed at the higher doses by intravenous dosing only. In mice, the alterations at 70 mg/kg/week included spleen weight increase up to 1.4-fold relative to control, increases in alanine aminotransferase and aspartate aminotransferase up to 1.8-fold over control, interleukin-10 increases up to 3.7-fold, and monocyte chemoattractant protein-1 increase up to 1.9-fold over control. No significant clinical pathology or histopathology changes were seen in mice at 20 mg/kg/week or less. The toxicity profile of ISIS 481464 is consistent with effects observed with phosphorothioate ASOs containing 2'-O-methoxyethylribose modifications instead of cEt.


Subject(s)
Kidney/drug effects , Liver/drug effects , Oligonucleotides, Antisense/toxicity , Phosphorothioate Oligonucleotides/toxicity , Spleen/drug effects , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Drug Administration Routes , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Humans , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Macaca fascicularis , Male , Mice , Oligonucleotides, Antisense/chemical synthesis , Oligonucleotides, Antisense/pharmacokinetics , Organ Size/drug effects , Partial Thromboplastin Time , Phosphorothioate Oligonucleotides/chemical synthesis , Phosphorothioate Oligonucleotides/pharmacokinetics , STAT3 Transcription Factor/antagonists & inhibitors , Spleen/metabolism , Spleen/pathology
7.
Toxicol Sci ; 130(2): 319-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872057

ABSTRACT

The European Medicines Agency has expressed concern regarding (1) the potential for antisense oligonucleotide (ASO) therapeutics to induce sequence-specific mutation at genomic DNA and (2) the capability of ASO degradation products (nucleotide analogues) to incorporate into newly synthesized genomic DNA via DNA polymerase and cause mutation if base pairing occurs with reduced fidelity. Treating human lymphoblastoid cells with a biologically active antisense molecule induced sequence-specific mutation within genomic DNA over fourfold, in a system where RAD51 protein expression was induced. This finding has implications for ASO therapeutics with individuals with an induced DNA damage response, such as cancer patients. Furthermore, a phosphorothioate nucleotide analogue potently induced mutation at genomic DNA two orders of magnitude above control. This study shows that a biologically active ASO molecule can induce heritable sequence alterations, and if degraded, its respective analogue may incorporate into genomic DNA with mutagenic consequences.


Subject(s)
Mutagenesis , Mutation , Oligonucleotides, Antisense/toxicity , Phosphorothioate Oligonucleotides/toxicity , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS Proteins , Base Sequence , Biotransformation , Cell Line , DNA Damage , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Molecular Sequence Data , Mutagenicity Tests , Oligonucleotides, Antisense/metabolism , Phosphorothioate Oligonucleotides/metabolism , Procollagen N-Endopeptidase/genetics , Procollagen N-Endopeptidase/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Up-Regulation
8.
Cancer Chemother Pharmacol ; 68(1): 193-205, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20886212

ABSTRACT

PURPOSE: GTI-2040, a 20-mer phosphorothioate oligonucleotide, was designed to hybridize to the mRNA sequence of human ribonucleotide reductase R2. GTI-2040 has been shown to inhibit human cancer cell proliferation by downregulation of R2 expression in vitro and to significantly inhibit tumor growth in xenograft models of human cancer in mice. As part of the safety evaluation for human clinical trials, the toxicity and toxicokinetics of GTI-2040 were determined in Sprague-Dawley rats and rhesus monkeys. METHODS: GTI-2040 was administered to rats at 2, 10, and 50 mg/kg/day by bolus intravenous injection every second day for 21 days with a 21-day recovery. In monkeys, an acute study was performed with single, escalating doses of GTI-2040 ranging from 10 to 80 mg/kg given as a 24-h continuous intravenous infusion. As well, a 21-day, continuous intravenous infusion study with GTI-2040 was conducted in monkeys at 2, 10, and 50 mg/kg/day, with a 3-week recovery. Blood sampling was done to measure GTI-2040 plasma concentrations, metabolites, and pharmacokinetic parameters, and tissues were collected to assess the distribution of GTI-2040 and/or metabolites. RESULTS: The toxicities of GTI-2040 in both rats and monkeys were typical for the phosphorothioate oligonucleotide class of compounds. In monkeys, there was a dose-related increase in GTI-2040 plasma levels with concomitant increase in complement activation and prolongation of activated partial thromboplastin time. In both rats and monkeys, the tissues having the highest concentrations of GTI-2040 (kidney, liver, spleen) had the largest dose-related toxic effects. Adverse effects were diminished or absent in the recovery animals. CONCLUSIONS: GTI-2040 was well tolerated when infused over 24 h at doses up to 80 mg/kg in monkeys. In rats and monkeys, GTI-2040 was reasonably well tolerated and showed reversible toxicities when administered at doses up to 50 mg/kg/day for 21 days. The no observed adverse effect dose level for GTI-2040 in both animal species was 2 mg/kg/day. There were no apparent sequence-specific effects related to the interaction of GTI-2040 with the R2 component of the mRNA expressing ribonucleotide reductase.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Oligodeoxyribonucleotides/pharmacokinetics , Oligodeoxyribonucleotides/toxicity , Phosphorothioate Oligonucleotides/pharmacokinetics , Phosphorothioate Oligonucleotides/toxicity , Ribonucleotide Reductases/genetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Base Sequence , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Macaca mulatta , Male , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/blood , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/blood , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Ribonucleotide Reductases/metabolism
9.
Pulm Pharmacol Ther ; 21(6): 845-54, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18761414

ABSTRACT

TPI ASM8 and TPI 1100 are two products containing modified phosphorothioate antisense oligonucleotides (AONs), which are undergoing development for the treatment of asthma and chronic obstructive pulmonary disease (COPD), respectively. TPI ASM8 is comprised of two AONs, one targeting the human chemokine receptor 3 (CCR3) and the other targeting the common beta-chain of the IL-3/IL-5/GM-CSF receptors. TPI 1100 is also a dual-AON compound targeting the phosphodiesterase (PDE) 4 and 7 isotypes. For both products, the AONs are present in a 1:1 ratio by weight. Both products will be administered by inhalation to patients, and TPI ASM8 is currently undergoing Phase 2 clinical trials. As part of the safety assessment of both products, the toxicity and disposition (i.e., pharmacokinetics of the AON components in plasma and tissues) were investigated in 14-day inhalation studies in monkeys at doses ranging from 0.05 to 2.5mg/kg/day. Results indicated that both products were safe and well tolerated at all dose levels. Reversible treatment-related alterations were only observed at the high dose levels tested and were limited to changes in the respiratory tract which were characterized primarily by the presence of alveolar macrophages in the absence of a generalized inflammatory response. Plasma pharmacokinetic profiles showed very low plasma concentrations, and no plasma accumulation was observed after repeated doses. While significant amounts of the AONs of both TPI ASM8 and TPI 1100 were measured in trachea and lung, only limited amounts of the AONs could be measured in kidney and liver, which, in combination with the low plasma level data, is indicative of very low systemic exposure. Taken together, these results demonstrate that these two new AON-based products are safe and that delivery via the inhaled route achieves localized deposition in the pulmonary tract with very limited systemic exposure and reduced toxicity compared to other routes of AON administration.


Subject(s)
Oligonucleotides, Antisense/toxicity , Phosphorothioate Oligonucleotides/toxicity , Administration, Inhalation , Animals , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Female , Lung/drug effects , Lymph Nodes/drug effects , Macaca fascicularis , Male , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacokinetics , Phosphorothioate Oligonucleotides/administration & dosage , Phosphorothioate Oligonucleotides/chemistry , Phosphorothioate Oligonucleotides/pharmacokinetics , Respiratory System/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...