1
|
Li Z, Tong F, Xiao L, Larson NR, Zhou X, Zhang Y, Immel-Brown JP, Bou-Assaf GM. Establishing stereochemical comparability in phosphorothioate oligonucleotides with nuclease P1 digestion coupled with LCMS analysis. Analyst 2023; 148:5361-5365. [PMID: 37755232 DOI: 10.1039/d3an01392h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Stereochemical comparability is critical for ensuring manufacturing consistency in therapeutic phosphorothioate oligonucleotides. Currently, analytical methods for this assessment are limited. We hereby report on a novel protocol capable of detecting a stereochemistry change in a single phosphorothioate linkage by employing nuclease P1 digestion of the oligonucleotide with subsequent LCMS analysis of the resulting fragments. The method proves valuable for establishing stereochemical comparability and for ensuring manufacturing consistency of oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Zifan Li
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - Fei Tong
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - Li Xiao
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - Nicholas R Larson
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - Xuan Zhou
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - Yueheng Zhang
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - Jonas P Immel-Brown
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| | - George M Bou-Assaf
- Pharmaceutical Operations & Technology, Biogen, 225 Binney Street, Cambridge, MA 02142, USA.
| |
Collapse
|
2
|
Ning S, Liu C, Lou W, Yang JC, Lombard AP, D'Abronzo LS, Batra N, Yu AM, Leslie AR, Sharifi M, Evans CP, Gao AC. Bioengineered BERA-Wnt5a siRNA Targeting Wnt5a/FZD2 Signaling Suppresses Advanced Prostate Cancer Tumor Growth and Enhances Enzalutamide Treatment. Mol Cancer Ther 2022; 21:1594-1607. [PMID: 35930737 PMCID: PMC9547958 DOI: 10.1158/1535-7163.mct-22-0216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/23/2022] [Accepted: 07/28/2022] [Indexed: 01/21/2023]
Abstract
The next-generation antiandrogen drugs such as enzalutamide and abiraterone extend survival times and improve quality of life in patients with advanced prostate cancer. However, resistance to both drugs occurs frequently through mechanisms that are incompletely understood. Wnt signaling, particularly through Wnt5a, plays vital roles in promoting prostate cancer progression and induction of resistance to enzalutamide and abiraterone. Development of novel strategies targeting Wnt5a to overcome resistance is an urgent need. In this study, we demonstrated that Wnt5a/FZD2-mediated noncanonical Wnt pathway is overexpressed in enzalutamide-resistant prostate cancer. In patient databases, both the levels of Wnt5a and FZD2 expression are upregulated upon the development of enzalutamide resistance and correlate with higher Gleason score, biochemical recurrence, and metastatic status, and with shortened disease-free survival duration. Blocking Wnt5a/FZD2 signal transduction not only diminished the activation of noncanonical Wnt signaling pathway, but also suppressed the constitutively activated androgen receptor (AR) and AR variants. Furthermore, we developed a novel bioengineered BERA-Wnt5a siRNA construct and demonstrated that inhibition of Wnt5a expression by the BERA-Wnt5a siRNA significantly suppressed tumor growth and enhanced enzalutamide treatment in vivo. These results indicate that Wnt5a/FZD2 signal pathway plays a critical role in promoting enzalutamide resistance, and targeting this pathway by BERA-Wnt5a siRNA can be developed as a potential therapy to treat advanced prostate cancer.
Collapse
Affiliation(s)
- Shu Ning
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Chengfei Liu
- Department of Urologic Surgery, University of California Davis, Davis, California
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California
| | - Wei Lou
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Joy C Yang
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Alan P Lombard
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Leandro S D'Abronzo
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Neelu Batra
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California
| | - Ai-Ming Yu
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California
| | - Amy R Leslie
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Masuda Sharifi
- Department of Urologic Surgery, University of California Davis, Davis, California
| | - Christopher P Evans
- Department of Urologic Surgery, University of California Davis, Davis, California
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California
| | - Allen C Gao
- Department of Urologic Surgery, University of California Davis, Davis, California
- UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California
- VA Northern California Health Care System, Sacramento, California
| |
Collapse
|
3
|
Halloy F, Iyer P, Ćwiek P, Ghidini A, Barman-Aksözen J, Wildner-Verhey van Wijk N, Theocharides APA, Minder E, Schneider-Yin X, Schümperli D, Hall J. Delivery of oligonucleotides to bone marrow to modulate ferrochelatase splicing in a mouse model of erythropoietic protoporphyria. Nucleic Acids Res 2020; 48:4658-4671. [PMID: 32313951 PMCID: PMC7229840 DOI: 10.1093/nar/gkaa229] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/25/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022] Open
Abstract
Erythropoietic protoporphyria (EPP) is a rare genetic disease in which patients experience acute phototoxic reactions after sunlight exposure. It is caused by a deficiency in ferrochelatase (FECH) in the heme biosynthesis pathway. Most patients exhibit a loss-of-function mutation in trans to an allele bearing a SNP that favors aberrant splicing of transcripts. One viable strategy for EPP is to deploy splice-switching oligonucleotides (SSOs) to increase FECH synthesis, whereby an increase of a few percent would provide therapeutic benefit. However, successful application of SSOs in bone marrow cells is not described. Here, we show that SSOs comprising methoxyethyl-chemistry increase FECH levels in cells. We conjugated one SSO to three prototypical targeting groups and administered them to a mouse model of EPP in order to study their biodistribution, their metabolic stability and their FECH splice-switching ability. The SSOs exhibited distinct distribution profiles, with increased accumulation in liver, kidney, bone marrow and lung. However, they also underwent substantial metabolism, mainly at their linker groups. An SSO bearing a cholesteryl group increased levels of correctly spliced FECH transcript by 80% in the bone marrow. The results provide a promising approach to treat EPP and other disorders originating from splicing dysregulation in the bone marrow.
Collapse
Affiliation(s)
- François Halloy
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Pavithra S Iyer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Paulina Ćwiek
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Alice Ghidini
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | | | | | - Alexandre P A Theocharides
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | | | - Daniel Schümperli
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| | - Jonathan Hall
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland
| |
Collapse
|
4
|
Niu Y, Wan A, Lin Z, Lu X, Wan G. N 6-Methyladenosine modification: a novel pharmacological target for anti-cancer drug development. Acta Pharm Sin B 2018; 8:833-843. [PMID: 30505654 PMCID: PMC6251950 DOI: 10.1016/j.apsb.2018.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/23/2018] [Accepted: 05/27/2018] [Indexed: 12/11/2022] Open
Abstract
N6-Methyladenosine (m6A) modification is the most pervasive modification of human mRNA molecules. It is reversible via regulation of m6A modification methyltransferase, demethylase and proteins that preferentially recognize m6A modification as “writers”, “erasers” and “readers”, respectively. Altered expression levels of the m6A modification key regulators substantially affect their function, leading to significant phenotype changes in the cell and organism. Recent studies have proved that the m6A modification plays significant roles in regulation of metabolism, stem cell self-renewal, and metastasis in a variety of human cancers. In this review, we describe the potential roles of m6A modification in human cancers and summarize their underlying molecular mechanisms. Moreover, we will highlight potential therapeutic approaches by targeting the key m6A modification regulators for cancer drug development.
Collapse
|
5
|
Rangasamy L, Chelvam V, Kanduluru AK, Srinivasarao M, Bandara NA, You F, Orellana EA, Kasinski AL, Low PS. New Mechanism for Release of Endosomal Contents: Osmotic Lysis via Nigericin-Mediated K +/H + Exchange. Bioconjug Chem 2018; 29:1047-1059. [PMID: 29446616 DOI: 10.1021/acs.bioconjchem.7b00714] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although peptides, antibodies/antibody fragments, siRNAs, antisense DNAs, enzymes, and aptamers are all under development as possible therapeutic agents, the breadth of their applications has been severely compromised by their inability to reach intracellular targets. Thus, while macromolecules can often enter cells by receptor-mediated endocytosis, their missions frequently fail due to an inability to escape their entrapping endosomes. In this paper, we describe a general method for promoting release of any biologic material from any entrapping endosome. The strategy relies on the fact that all nascent endosomes contain extracellular (Na+-enriched) medium, but are surrounded by intracellular (K+-enriched) fluid in the cytoplasm. Osmotic swelling and rupture of endosomes will therefore be facilitated if the flow of K+ down its concentration gradient from the cytosol into the endosome can be facilitated without allowing downhill flow of Na+ from the endosome into the cytosol. While any K+ selective ionophore can promote the K+ specific influx, the ideal K+ ionophore will also exchange influxed K+ for an osmotically inactive proton (H+) in order to prevent buildup of an electrical potential that would rapidly halt K+ influx. The only ionophore that catalyzes this exchange of K+ for H+ efficiently is nigericin. We demonstrate here that ligand-targeted delivery of nigericin into endosomes that contain an otherwise impermeable fluorescent dye can augment release of the dye into the cell cytosol via swelling/bursting of the entrapping endosomes. We further show that nigericin-facilitated escape of a folate-targeted luciferase siRNA conjugate from its entrapping endosomes promotes rapid suppression of the intended luciferase reporter gene. Taken together, we propose that ionophore-catalyzed entry of K+ into endosomal compartments can promote the release of otherwise impermeable contents from their encapsulating endosomes.
Collapse
Affiliation(s)
- Loganathan Rangasamy
- Purdue Institute for Drug Discovery , 720 Clinic Drive , West Lafayette Indiana 47907 , United States
| | - Venkatesh Chelvam
- Purdue Institute for Drug Discovery , 720 Clinic Drive , West Lafayette Indiana 47907 , United States.,Discipline of Chemistry, Centre for Biosciences and Biomedical Engineering , Indian Institute of Technology , Indore , Madhya Pradesh , 453552 , India
| | - Ananda Kumar Kanduluru
- Purdue Institute for Drug Discovery , 720 Clinic Drive , West Lafayette Indiana 47907 , United States
| | - Madduri Srinivasarao
- Purdue Institute for Drug Discovery , 720 Clinic Drive , West Lafayette Indiana 47907 , United States
| | - N Achini Bandara
- Purdue Institute for Drug Discovery , 720 Clinic Drive , West Lafayette Indiana 47907 , United States
| | - Fei You
- Endocyte, Inc. , 3000 Kent Avenue, Suite A1-100 , West Lafayette , Indiana 47906 , United States
| | - Esteban A Orellana
- Department of Biological Sciences, Bindley Bioscience Center , Purdue University , West Lafayette , Indiana 47907 , United States
| | - Andrea L Kasinski
- Department of Biological Sciences, Bindley Bioscience Center , Purdue University , West Lafayette , Indiana 47907 , United States
| | - Philip S Low
- Purdue Institute for Drug Discovery , 720 Clinic Drive , West Lafayette Indiana 47907 , United States
| |
Collapse
|
6
|
Setten RL, Lightfoot HL, Habib NA, Rossi JJ. Development of MTL-CEBPA: Small Activating RNA Drug for Hepatocellular Carcinoma. Curr Pharm Biotechnol 2018; 19:611-621. [PMID: 29886828 PMCID: PMC6204661 DOI: 10.2174/1389201019666180611093428] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Oligonucleotide drug development has revolutionised the drug discovery field. Within this field, 'small' or 'short' activating RNAs (saRNA) are a more recently discovered category of short double-stranded RNA with clinical potential. saRNAs promote transcription from target loci, a phenomenon widely observed in mammals known as RNA activation (RNAa). OBJECTIVE The ability to target a particular gene is dependent on the sequence of the saRNA. Hence, the potential clinical application of saRNAs is to increase target gene expression in a sequence-specific manner. saRNA-based therapeutics present opportunities for expanding the "druggable genome" with particular areas of interest including transcription factor activation and cases of haploinsufficiency. RESULTS AND CONCLUSION In this mini-review, we describe the pre-clinical development of the first saRNA drug to enter the clinic. This saRNA, referred to as MTL-CEBPA, induces increased expression of the transcription factor CCAAT/enhancer-binding protein alpha (CEBPα), a tumour suppressor and critical regulator of hepatocyte function. MTL-CEBPA is presently in Phase I clinical trials for hepatocellular carcinoma (HCC). The clinical development of MTL-CEBPA will demonstrate "proof of concept" that saRNAs can provide the basis for drugs which enhance target gene expression and consequently improve treatment outcome in patients.
Collapse
Affiliation(s)
| | | | | | - John J. Rossi
- Address correspondence to this author at the Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA; Tel: 626-218-7390; Fax: 626-301-8371; E-mail:
| |
Collapse
|
7
|
Schürch S. Characterization of nucleic acids by tandem mass spectrometry - The second decade (2004-2013): From DNA to RNA and modified sequences. MASS SPECTROMETRY REVIEWS 2016; 35:483-523. [PMID: 25288464 DOI: 10.1002/mas.21442] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 05/04/2014] [Accepted: 05/04/2014] [Indexed: 06/03/2023]
Abstract
Nucleic acids play key roles in the storage and processing of genetic information, as well as in the regulation of cellular processes. Consequently, they represent attractive targets for drugs against gene-related diseases. On the other hand, synthetic oligonucleotide analogues have found application as chemotherapeutic agents targeting cellular DNA and RNA. The development of effective nucleic acid-based chemotherapeutic strategies requires adequate analytical techniques capable of providing detailed information about the nucleotide sequences, the presence of structural modifications, the formation of higher-order structures, as well as the interaction of nucleic acids with other cellular components and chemotherapeutic agents. Due to the impressive technical and methodological developments of the past years, tandem mass spectrometry has evolved to one of the most powerful tools supporting research related to nucleic acids. This review covers the literature of the past decade devoted to the tandem mass spectrometric investigation of nucleic acids, with the main focus on the fundamental mechanistic aspects governing the gas-phase dissociation of DNA, RNA, modified oligonucleotide analogues, and their adducts with metal ions. Additionally, recent findings on the elucidation of nucleic acid higher-order structures by tandem mass spectrometry are reviewed. © 2014 Wiley Periodicals, Inc., Mass Spec Rev 35:483-523, 2016.
Collapse
Affiliation(s)
- Stefan Schürch
- Department of Chemistry and Biochemistry, University of Bern, CH-3012, Bern, Switzerland
| |
Collapse
|
8
|
Polyamine-oligonucleotide conjugates: a promising direction for nucleic acid tools and therapeutics. Future Med Chem 2015; 7:1733-49. [PMID: 26424049 DOI: 10.4155/fmc.15.90] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chemical modification and/or the conjugation of small functional molecules to oligonucleotides have significantly improved their biological and biophysical properties, addressing issues such as poor cell penetration, stability to nucleases and low affinity for their targets. Here, the authors review the literature reporting on the biophysical, biochemical and biological properties of one particular class of modification - polyamine-oligonucleotide conjugates. Naturally derived and synthetic polyamines have been grafted onto a variety of oligonucleotide formats, including antisense oligonucleotides and siRNAs. In many cases this has had beneficial effects on their properties such as target hybridization, nuclease resistance, cellular uptake and activity. Polyamine-oligonucleotide conjugation, therefore, represents a promising direction for the further development of oligonucleotide-based therapeutics and tools.
Collapse
|
9
|
Niemietz C, Chandhok G, Schmidt H. Therapeutic Oligonucleotides Targeting Liver Disease: TTR Amyloidosis. Molecules 2015; 20:17944-75. [PMID: 26437390 PMCID: PMC6332041 DOI: 10.3390/molecules201017944] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/13/2022] Open
Abstract
The liver has become an increasingly interesting target for oligonucleotide therapy. Mutations of the gene encoding transthyretin (TTR), expressed in vast amounts by the liver, result in a complex degenerative disease, termed familial amyloid polyneuropathy (FAP). Misfolded variants of TTR are linked to the establishment of extracellular protein deposition in various tissues, including the heart and the peripheral nervous system. Recent progress in the chemistry and formulation of antisense (ASO) and small interfering RNA (siRNA) designed for a knockdown of TTR mRNA in the liver has allowed to address the issue of gene-specific molecular therapy in a clinical setting of FAP. The two therapeutic oligonucleotides bind to RNA in a sequence specific manner but exploit different mechanisms. Here we describe major developments that have led to the advent of therapeutic oligonucleotides for treatment of TTR-related disease.
Collapse
MESH Headings
- Amyloid Neuropathies, Familial/genetics
- Amyloid Neuropathies, Familial/therapy
- Animals
- Clinical Studies as Topic
- Drug Evaluation, Preclinical
- Gene Silencing
- Genetic Therapy
- Humans
- Liver Diseases/genetics
- Liver Diseases/therapy
- Mutation
- Oligonucleotides/administration & dosage
- Oligonucleotides/chemistry
- Oligonucleotides/genetics
- Oligonucleotides/therapeutic use
- Oligonucleotides, Antisense/administration & dosage
- Oligonucleotides, Antisense/chemistry
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/therapeutic use
- Prealbumin/genetics
- RNA Interference
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Christoph Niemietz
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, D-48149 Münster, Germany.
| | - Gursimran Chandhok
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, D-48149 Münster, Germany.
| | - Hartmut Schmidt
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, D-48149 Münster, Germany.
| |
Collapse
|
10
|
Jeker LT, Marone R. Targeting microRNAs for immunomodulation. Curr Opin Pharmacol 2015; 23:25-31. [PMID: 26021286 DOI: 10.1016/j.coph.2015.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/29/2022]
Abstract
microRNAs (miRNA) are small regulatory RNAs exerting pleiotropic functions in virtually any immune cell-type. Dozens of miRNAs with a known function in the immune system constitute interesting drug targets for immunomodulation. Chemical modifications of nucleic acid-based miRNA mimics and inhibitors largely solved instability issues but delivery to immune cells remains a major challenge. However, recent success targeting the acidic tumor microenvironment is very promising for inflammatory diseases. Moreover, small molecules are being explored as an interesting alternative. Although RNA is often considered 'undruggable' by small molecules recent progress modulating miRNA function through small molecules is encouraging. Computational approaches even allow predictions about specific small molecule/RNA interactions. Finally, recent clinical success demonstrates that drugs targeting RNAs work in humans.
Collapse
Affiliation(s)
- Lukas T Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Romina Marone
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| |
Collapse
|
11
|
Jahns H, Roos M, Imig J, Baumann F, Wang Y, Gilmour R, Hall J. Stereochemical bias introduced during RNA synthesis modulates the activity of phosphorothioate siRNAs. Nat Commun 2015; 6:6317. [PMID: 25744034 PMCID: PMC4366519 DOI: 10.1038/ncomms7317] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 01/19/2015] [Indexed: 12/18/2022] Open
Abstract
An established means of improving the pharmacokinetics properties of oligoribonucleotides (ORNs) is to exchange their phosphodiester linkages for phosphorothioates (PSs). However, this strategy has not been pursued for small interfering RNAs (siRNAs), possibly because of sporadic reports that PS siRNAs show reduced inhibitory activity. The PS group is chiral at phosphorous (Rp/Sp centres), and conventional solid-phase synthesis of PS ORNs produces a population of diastereoisomers. Here we show that the choice of the activating agent for the synthesis of a PS ORN influences the Rp/Sp ratio of PS linkages throughout the strand. Furthermore, PS siRNAs composed of ORNs with a higher fraction of Rp centres show greater resistance to nucleases in serum and are more effective inhibitors in cells than their Sp counterparts. The finding that a stereochemically biased population of ORN diastereoisomers can be synthesized and exploited pharmacologically is important because uniform PS modification of siRNAs may provide a useful compromise of their pharmacokinetics and pharmacodynamics properties in RNAi therapeutics. Therapeutic oligonucleotides can be made more stable by substituting their achiral phosphodiester groups for chiral phosphorothioate linkages. Here, the authors present a synthesis of phosphorothioated RNAs, where the activator controls strand stereochemistry, and also the activity of assembled siRNAs.
Collapse
Affiliation(s)
- Hartmut Jahns
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg-4, CH-8093 Zürich, Switzerland
| | - Martina Roos
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg-4, CH-8093 Zürich, Switzerland
| | - Jochen Imig
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg-4, CH-8093 Zürich, Switzerland
| | - Fabienne Baumann
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg-4, CH-8093 Zürich, Switzerland
| | - Yuluan Wang
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg-4, CH-8093 Zürich, Switzerland
| | - Ryan Gilmour
- Institute for Organic Chemistry, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Jonathan Hall
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg-4, CH-8093 Zürich, Switzerland
| |
Collapse
|
12
|
Wang X, Zhang J, Li Y, Chen G, Wang X. Enzymatic synthesis of modified oligonucleotides by PEAR using Phusion and KOD DNA polymerases. Nucleic Acid Ther 2014; 25:27-34. [PMID: 25517220 DOI: 10.1089/nat.2014.0513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antisense synthetic oligonucleotides have been developed as potential gene-targeted therapeutics. We previously reported polymerase-endonuclease amplification reaction (PEAR) for amplification of natural and 5'-O-(1-thiotriphosphate) (S)-modified oligonucleotides. Here, we extended the PEAR technique for enzymatic preparation of 2'-deoxy-2'-fluoro-(2'-F) and 2'-F/S double-modified oligonucleotides. The result showed that KOD and Phusion DNA polymerase could synthesize oligonucleotides with one or two modified nucleotides, and KOD DNA polymerase is more suitable than Phusion DNA polymerase for PEAR amplification of 2'-F and 2'-F/S double modified oligonucleotides. The composition of PEAR products were analyzed by electrospray ionization liquid chromatography mass spectrometry (ESI/LC/MS) detection and showed that the sequence of the PEAR products are maintained at an extremely high accuracy (>99.9%), and after digestion the area percent of full-length modified oligonucleotides reaches 89.24%. PEAR is suitable for synthesis of modified oligonucleotides efficiently and with high purity.
Collapse
Affiliation(s)
- Xuxiang Wang
- Department of Biotechnology, College of Marine Life Sciences, Ocean University of China , Qingdao, Shandong Province, China
| | | | | | | | | |
Collapse
|
13
|
Rigo F, Seth PP, Bennett CF. Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 825:303-52. [PMID: 25201110 DOI: 10.1007/978-1-4939-1221-6_9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Before a messenger RNA (mRNA) is translated into a protein in the cytoplasm, its pre-mRNA precursor is extensively processed through capping, splicing and polyadenylation in the nucleus. Defects in the processing of pre-mRNAs due to mutations in RNA sequences often cause disease. Traditional small molecules or protein-based therapeutics are not well suited for correcting processing defects by targeting RNA. However, antisense oligonucleotides (ASOs) designed to bind RNA by Watson-Crick base pairing can target most RNA transcripts and have emerged as the ideal therapeutic agents for diseases that are caused by pre-mRNA processing defects. Here we review the diverse ASO-based mechanisms that can be exploited to modulate the expression of RNA. We also discuss how advancements in medicinal chemistry and a deeper understanding of the pharmacokinetic and toxicological properties of ASOs have enabled their use as therapeutic agents. We end by describing how ASOs have been used successfully to treat various pre-mRNA processing diseases in animal models.
Collapse
Affiliation(s)
- Frank Rigo
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, USA,
| | | | | |
Collapse
|
14
|
Eckstein F. Phosphorothioates, Essential Components of Therapeutic Oligonucleotides. Nucleic Acid Ther 2014; 24:374-87. [DOI: 10.1089/nat.2014.0506] [Citation(s) in RCA: 335] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Fritz Eckstein
- Max-Planck-Institut für Experimentelle Medizin, Göttingen, Germany
| |
Collapse
|
15
|
Trembley JH, Unger GM, Korman VL, Abedin MJ, Nacusi LP, Vogel RI, Slaton JW, Kren BT, Ahmed K. Tenfibgen ligand nanoencapsulation delivers bi-functional anti-CK2 RNAi oligomer to key sites for prostate cancer targeting using human xenograft tumors in mice. PLoS One 2014; 9:e109970. [PMID: 25333839 PMCID: PMC4198192 DOI: 10.1371/journal.pone.0109970] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/02/2014] [Indexed: 01/11/2023] Open
Abstract
Protected and specific delivery of nucleic acids to malignant cells remains a highly desirable approach for cancer therapy. Here we present data on the physical and chemical characteristics, mechanism of action, and pilot therapeutic efficacy of a tenfibgen (TBG)-shell nanocapsule technology for tumor-directed delivery of single stranded DNA/RNA chimeric oligomers targeting CK2αα' to xenograft tumors in mice. The sub-50 nm size TBG nanocapsule (s50-TBG) is a slightly negatively charged, uniform particle of 15 - 20 nm size which confers protection to the nucleic acid cargo. The DNA/RNA chimeric oligomer (RNAi-CK2) functions to decrease CK2αα' expression levels via both siRNA and antisense mechanisms. Systemic delivery of s50-TBG-RNAi-CK2 specifically targets malignant cells, including tumor cells in bone, and at low doses reduces size and CK2-related signals in orthotopic primary and metastatic xenograft prostate cancer tumors. In conclusion, the s50-TBG nanoencapsulation technology together with the chimeric oligomer targeting CK2αα' offer significant promise for systemic treatment of prostate malignancy.
Collapse
Affiliation(s)
- Janeen H. Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | | | - Vicci L. Korman
- GeneSegues Inc., Chaska, Minnesota, United States of America
| | - Md. Joynal Abedin
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Lucas P. Nacusi
- GeneSegues Inc., Chaska, Minnesota, United States of America
| | - Rachel I. Vogel
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joel W. Slaton
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Betsy T. Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Khalil Ahmed
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
16
|
Leisvuori A, Lönnberg H, Ora M. 4-Acetylthio-2,2-dimethyl-3-oxobutyl Group as an Esterase- and Thermo-Labile Protecting Group for Oligomeric Phosphodiesters. European J Org Chem 2014. [DOI: 10.1002/ejoc.201402412] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
17
|
Hosten TA, Zhao K, Han HQ, Liu G, He XH. Alicaforsen: An Emerging Therapeutic Agent for Ulcerative Colitis and Refractory Pouchitis. Gastroenterology Res 2014; 7:51-55. [PMID: 27785270 PMCID: PMC5051075 DOI: 10.14740/gr599w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2014] [Indexed: 12/29/2022] Open
Abstract
Pouchitis is a relatively common complication that develops following ileal pouch-anal anastomosis in patients with complicated ulcerative colitis (UC). Both pouchitis and UC share similarities in their development, as well as in the mechanisms involving mediators of the inflammatory process. In the recent years, the discovery and investigation of biological therapies have led to advancement in the management of these disorders, and the continuation of research on this novel area holds strong implications for a future reduction in the use of invasive surgical procedures. Alicaforsen represents one of these emerging therapeutic agents, and has demonstrated promising results in both preclinical and clinical settings. This article reviews the therapeutic effects of alicaforsen for the management of UC and refractory pouchitis, with special emphasis on the mechanism of action of this therapeutic agent and the clinical studies asserting its effectiveness.
Collapse
Affiliation(s)
- Terron Anthony Hosten
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ke Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong Qiu Han
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Hui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
18
|
Tolcher AW, Rodrigueza WV, Rasco DW, Patnaik A, Papadopoulos KP, Amaya A, Moore TD, Gaylor SK, Bisgaier CL, Sooch MP, Woolliscroft MJ, Messmann RA. A phase 1 study of the BCL2-targeted deoxyribonucleic acid inhibitor (DNAi) PNT2258 in patients with advanced solid tumors. Cancer Chemother Pharmacol 2014; 73:363-71. [PMID: 24297683 PMCID: PMC3909249 DOI: 10.1007/s00280-013-2361-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 11/13/2013] [Indexed: 11/04/2022]
Abstract
PURPOSE Maximum tolerated dose, safety, pharmacokinetics, and pharmacodynamics were assessed in this phase 1 study of PNT2258, a BCL-2-targeted liposomal formulation of a 24-base DNA oligonucleotide called PNT100. METHODS Patients with malignant solid tumors were assigned sequentially to one of ten dose-escalation cohorts of PNT2258 at 1, 2, 4, 8, 16, 32, 64, 85, 113, and 150 mg/m(2) administered intravenously on days 1 through 5 of each 21-day cycle. Pharmacokinetics were determined on days 1 and 5 of the first cycle. Lymphocyte and platelets concentrations were measured for evidence of BCL2-targeted effect. CT scans were used to identify radiologic evidence of anti-tumor effect. RESULTS Twenty-two subjects received PNT2258, and the maximum tolerated dose for PNT2258 was not reached. Doses at or above 32 mg/m(2) resulted in exposure to PNT2258 above the exposure level required for anti-tumor activity in preclinical xenograft testing of 22,377 ng h/ml (PK analysis 2012). Fatigue was the most commonly reported adverse event. Dose-limiting toxicity, manifesting as a transient increase in aspartate aminotransferase, occurred at 150 mg/m(2), the highest dose tested. Four subjects, two each with diagnosis of non-small-cell lung cancer and sarcoma, treated at doses of 64 mg/m(2) or higher, remained on study for 5-8 cycles. CONCLUSIONS PNT2258 was safe and well tolerated at the doses tested up to 150 mg/m(2). Exposure to PNT2258 resulted in clinically manageable decreases in lymphocyte and platelet concentrations.
Collapse
Affiliation(s)
- Anthony W. Tolcher
- South Texas Accelerated Research Therapeutics (START), LLC, San Antonio, TX USA
| | - Wendi V. Rodrigueza
- ProNAi Therapeutics, Inc., 46701 Commerce Center Dr., Plymouth, MI 48170 USA
| | - Drew W. Rasco
- South Texas Accelerated Research Therapeutics (START), LLC, San Antonio, TX USA
| | - Amita Patnaik
- South Texas Accelerated Research Therapeutics (START), LLC, San Antonio, TX USA
| | | | - Alex Amaya
- South Texas Accelerated Research Therapeutics (START), LLC, San Antonio, TX USA
| | | | - Shari K. Gaylor
- ProNAi Therapeutics, Inc., 46701 Commerce Center Dr., Plymouth, MI 48170 USA
| | - Charles L. Bisgaier
- ProNAi Therapeutics, Inc., 46701 Commerce Center Dr., Plymouth, MI 48170 USA
| | - Mina P. Sooch
- ProNAi Therapeutics, Inc., 46701 Commerce Center Dr., Plymouth, MI 48170 USA
| | | | - Richard A. Messmann
- ProNAi Therapeutics, Inc., 46701 Commerce Center Dr., Plymouth, MI 48170 USA
| |
Collapse
|
19
|
Sharma VK, Rungta P, Prasad AK. Nucleic acid therapeutics: basic concepts and recent developments. RSC Adv 2014. [DOI: 10.1039/c3ra47841f] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
20
|
Järver P, O'Donovan L, Gait MJ. A chemical view of oligonucleotides for exon skipping and related drug applications. Nucleic Acid Ther 2013; 24:37-47. [PMID: 24171481 DOI: 10.1089/nat.2013.0454] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Peter Järver
- Medical Research Council , Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | |
Collapse
|
21
|
Glinsky GV. RNA-guided diagnostics and therapeutics for next-generation individualized nanomedicine. J Clin Invest 2013; 123:2350-2. [PMID: 23728168 DOI: 10.1172/jci69268] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The absence of reliable quantitative laboratory tests for measurements of microRNAs and other classes of small noncoding RNAs in archived, formalin- fixed, paraffin-embedded human samples with sufficient specificity and sensitivity has significantly limited the development of clinically relevant noncoding RNA–based diagnostic and therapeutic applications. A report by Renwick et al. in this issue of the JCI presents a significant technical and methodological advance toward the development of reliable clinical laboratory- compatible multicolor RNA FISH methodology for molecular diagnostic applications and the near-term prospect of introduction of microRNA- based biomarkers into clinical practice. Further, this work is likely to advance the development of RNA-based therapeutics and next-generation individualized nanomedicine.
Collapse
Affiliation(s)
- Gennadi V Glinsky
- Sanford-Burnham Medical Research Institute, La Jolla, California, USA.
| |
Collapse
|