Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Molecules ; 24(18)2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31509944

ABSTRACT

Nucleic Acid Therapeutics (NATs), including siRNAs and AntiSense Oligonucleotides (ASOs), have great potential to drug the undruggable genome. Targeting siRNAs and ASOs to specific cell types of interest has driven dramatic improvement in efficacy and reduction in toxicity. Indeed, conjugation of tris-GalNAc to siRNAs and ASOs has shown clinical efficacy in targeting diseases driven by liver hepatocytes. However, targeting non-hepatic diseases with oligonucleotide therapeutics has remained problematic for several reasons, including targeting specific cell types and endosomal escape. Monoclonal antibody (mAb) targeting of siRNAs and ASOs has the potential to deliver these drugs to a variety of specific cell and tissue types. However, most conjugation strategies rely on random chemical conjugation through lysine or cysteine residues resulting in conjugate heterogeneity and a distribution of Drug:Antibody Ratios (DAR). To produce homogeneous DAR-2 conjugates with two siRNAs per mAb, we developed a novel two-step conjugation procedure involving microbial transglutaminase (MTGase) tagging of the antibody C-terminus with an azide-functionalized linker peptide that can be subsequently conjugated to dibenzylcyclooctyne (DBCO) bearing oligonucleotides through azide-alkyne cycloaddition. Antibody-siRNA (and ASO) conjugates (ARCs) produced using this strategy are soluble, chemically defined targeted oligonucleotide therapeutics that have the potential to greatly increase the number of targetable cell types.


Subject(s)
Antibodies/pharmacology , Immunoconjugates/chemistry , Oligonucleotides, Antisense/immunology , RNA, Small Interfering/immunology , Antibodies/chemistry , Antibodies/immunology , Azides/chemistry , Cell Lineage/drug effects , Cycloaddition Reaction , Cyclooctanes/chemistry , Drug Delivery Systems , Endosomes/drug effects , Hepatocytes/drug effects , Hepatocytes/immunology , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Liver/drug effects , Liver/immunology , Oligonucleotides, Antisense/antagonists & inhibitors , Oligonucleotides, Antisense/chemistry , Peptides/chemistry , Peptides/pharmacology , RNA, Small Interfering/antagonists & inhibitors , RNA, Small Interfering/chemistry , Transglutaminases/chemistry , Transglutaminases/immunology , Transglutaminases/pharmacology
2.
Nucleic Acid Ther ; 28(3): 109-118, 2018 06.
Article in English | MEDLINE | ID: mdl-29792572

ABSTRACT

Short-interfering RNA (siRNA)-induced RNAi responses have great potential to treat a wide variety of human diseases from cancer to pandemic viral outbreaks to Parkinson's Disease. However, before siRNAs can become drugs, they must overcome a billion years of evolutionary defenses designed to keep invading RNAs on the outside cells from getting to the inside of cells. Not surprisingly, significant effort has been placed in developing a wide array of delivery technologies. Foremost of these has been the development of N-acetylgalactosamine (GalNAc) siRNA conjugates for delivery to liver. Tris-GalNAc binds to the Asialoglycoprotein receptor that is highly expressed on hepatocytes resulting in rapid endocytosis. While the exact mechanism of escape across the endosomal lipid bilayer membrane remains unknown, sufficient amounts of siRNAs enter the cytoplasm to induce robust, target selective RNAi responses in vivo. Multiple GalNAc-siRNA conjugate clinical trials, including two phase III trials, are currently underway by three biotech companies to treat a wide variety of diseases. GalNAc-siRNA conjugates are a simple solution to the siRNA delivery problem for liver hepatocytes and have shown the RNAi (and antisense oligonucleotide) field the path forward for targeting other tissue types.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Endosomes/metabolism , Gene Transfer Techniques , Glycoconjugates/metabolism , RNA, Small Interfering/genetics , Virus Diseases/therapy , Acetylgalactosamine/metabolism , Asialoglycoprotein Receptor/genetics , Clinical Trials as Topic , Endocytosis , Endosomes/chemistry , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Liver/metabolism , Liver/pathology , Liver/virology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , RNA, Small Interfering/metabolism , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Diseases/pathology
3.
Nucleic Acids Res ; 43(7): e45, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25586224

ABSTRACT

Gene knockout strategies, RNAi and rescue experiments are all employed to study mammalian gene function. However, the disadvantages of these approaches include: loss of function adaptation, reduced viability and gene overexpression that rarely matches endogenous levels. Here, we developed an endogenous gene knockdown/rescue strategy that combines RNAi selectivity with a highly efficient CRISPR directed recombinant Adeno-Associated Virus (rAAV) mediated gene targeting approach to introduce allele-specific mutations plus an allele-selective siRNA Sensitive (siSN) site that allows for studying gene mutations while maintaining endogenous expression and regulation of the gene of interest. CRISPR/Cas9 plus rAAV targeted gene-replacement and introduction of allele-specific RNAi sensitivity mutations in the CDK2 and CDK1 genes resulted in a >85% site-specific recombination of Neo-resistant clones versus ∼8% for rAAV alone. RNAi knockdown of wild type (WT) Cdk2 with siWT in heterozygotic knockin cells resulted in the mutant Cdk2 phenotype cell cycle arrest, whereas allele specific knockdown of mutant CDK2 with siSN resulted in a wild type phenotype. Together, these observations demonstrate the ability of CRISPR plus rAAV to efficiently recombine a genomic locus and tag it with a selective siRNA sequence that allows for allele-selective phenotypic assays of the gene of interest while it remains expressed and regulated under endogenous control mechanisms.


Subject(s)
Alleles , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Dependovirus/genetics , RNA Interference , Base Sequence , CDC2 Protein Kinase , Cell Line , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinases/genetics , DNA Primers , Gene Knockdown Techniques , Humans , Mutation , Polymerase Chain Reaction , Recombination, Genetic
4.
Nat Biotechnol ; 32(12): 1256-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25402614

ABSTRACT

RNA interference (RNAi) has great potential to treat human disease. However, in vivo delivery of short interfering RNAs (siRNAs), which are negatively charged double-stranded RNA macromolecules, remains a major hurdle. Current siRNA delivery has begun to move away from large lipid and synthetic nanoparticles to more defined molecular conjugates. Here we address this issue by synthesis of short interfering ribonucleic neutrals (siRNNs) whose phosphate backbone contains neutral phosphotriester groups, allowing for delivery into cells. Once inside cells, siRNNs are converted by cytoplasmic thioesterases into native, charged phosphodiester-backbone siRNAs, which induce robust RNAi responses. siRNNs have favorable drug-like properties, including high synthetic yields, serum stability and absence of innate immune responses. Unlike siRNAs, siRNNs avidly bind serum albumin to positively influence pharmacokinetic properties. Systemic delivery of siRNNs conjugated to a hepatocyte-specific targeting domain induced extended dose-dependent in vivo RNAi responses in mice. We believe that siRNNs represent a technology that will open new avenues for development of RNAi therapeutics.


Subject(s)
Drug Delivery Systems , Prodrugs/therapeutic use , RNA, Small Interfering/therapeutic use , Animals , Humans , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Prodrugs/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Serum Albumin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...