351
|
Crooke ST, Baker BF, Crooke RM, Liang XH. Antisense technology: an overview and prospectus. Nat Rev Drug Discov 2021; 20:427-453. [PMID: 33762737 DOI: 10.1038/s41573-021-00162-z] [Citation(s) in RCA: 383] [Impact Index Per Article: 95.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
Antisense technology is now beginning to deliver on its promise to treat diseases by targeting RNA. Nine single-stranded antisense oligonucleotide (ASO) drugs representing four chemical classes, two mechanisms of action and four routes of administration have been approved for commercial use, including the first RNA-targeted drug to be a major commercial success, nusinersen. Although all the approved drugs are for use in patients with rare diseases, many of the ASOs in late- and middle-stage clinical development are intended to treat patients with very common diseases. ASOs in development are showing substantial improvements in potency and performance based on advances in medicinal chemistry, understanding of molecular mechanisms and targeted delivery. Moreover, the ASOs in development include additional mechanisms of action and routes of administration such as aerosol and oral formulations. Here, we describe the key technological advances that have enabled this progress and discuss recent clinical trials that illustrate the impact of these advances on the performance of ASOs in a wide range of therapeutic applications. We also consider strategic issues such as target selection and provide perspectives on the future of the field.
Collapse
|
352
|
Zhao B, Zhou B, Shi K, Zhang R, Dong C, Xie D, Tang L, Tian Y, Qian Z, Yang L. Sustained and targeted delivery of siRNA/DP7-C nanoparticles from injectable thermosensitive hydrogel for hepatocellular carcinoma therapy. Cancer Sci 2021; 112:2481-2492. [PMID: 33792132 PMCID: PMC8177784 DOI: 10.1111/cas.14903] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers in humans. The inhibition of peptidyl-prolyl cis/trans isomerase (Pin1) gene expression may have great potential in the treatment of HCC. N-Acetylgalactosamine (GalNAc) was used to target the liver. Cholesterol-modified antimicrobial peptide DP7 (DP7-C) acts as a carrier, the GalNAc-siRNA/DP7-C complex increases the uptake of GalNAc-siRNA and the escape of endosomes in hepatocytes. In addition, DP7-C nanoparticles and hydrogel-assisted GalNAc-Pin1 siRNA delivery can effectively enhance the stability and prolong the silencing effects of Pin1 siRNA. In an orthotopic liver cancer model, the GalNAc-Pin1 siRNA/DP7-C/hydrogel complex can potentially regulate Pin1 expression in hepatocellular carcinoma cells and effectively inhibit tumor progression. Our study proves that Pin1 siRNA is an efficient method for the treatment of HCC and provides a sustainable and effective drug delivery system for the suppression of liver cancer.
Collapse
Affiliation(s)
- Binyan Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Rui Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Chunyan Dong
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Daoyuan Xie
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Lin Tang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduChina
| |
Collapse
|
353
|
Kulkarni JA, Witzigmann D, Thomson SB, Chen S, Leavitt BR, Cullis PR, van der Meel R. The current landscape of nucleic acid therapeutics. NATURE NANOTECHNOLOGY 2021; 16:630-643. [PMID: 34059811 DOI: 10.1038/s41565-021-00898-0] [Citation(s) in RCA: 749] [Impact Index Per Article: 187.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 03/11/2021] [Indexed: 05/20/2023]
Abstract
The increasing number of approved nucleic acid therapeutics demonstrates the potential to treat diseases by targeting their genetic blueprints in vivo. Conventional treatments generally induce therapeutic effects that are transient because they target proteins rather than underlying causes. In contrast, nucleic acid therapeutics can achieve long-lasting or even curative effects via gene inhibition, addition, replacement or editing. Their clinical translation, however, depends on delivery technologies that improve stability, facilitate internalization and increase target affinity. Here, we review four platform technologies that have enabled the clinical translation of nucleic acid therapeutics: antisense oligonucleotides, ligand-modified small interfering RNA conjugates, lipid nanoparticles and adeno-associated virus vectors. For each platform, we discuss the current state-of-the-art clinical approaches, explain the rationale behind its development, highlight technological aspects that facilitated clinical translation and provide an example of a clinically relevant genetic drug. In addition, we discuss how these technologies enable the development of cutting-edge genetic drugs, such as tissue-specific nucleic acid bioconjugates, messenger RNA and gene-editing therapeutics.
Collapse
Affiliation(s)
- Jayesh A Kulkarni
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
- NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
- NanoVation Therapeutics, Vancouver, British Columbia, Canada
| | - Dominik Witzigmann
- NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
- NanoVation Therapeutics, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah B Thomson
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sam Chen
- Integrated Nanotherapeutics, Vancouver, British Columbia, Canada
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pieter R Cullis
- NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
354
|
Ren Y, Liu X, Ge H, Guo Y, Zhang Q, Xie M, Wang P, Zhu X, Zhang C. A Combinatorial Approach Based on Nucleic Acid Assembly and Electrostatic Compression for siRNA Delivery. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1168-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
355
|
The Non-Coding RNA Landscape in IgA Nephropathy-Where Are We in 2021? J Clin Med 2021; 10:jcm10112369. [PMID: 34071162 PMCID: PMC8198207 DOI: 10.3390/jcm10112369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
IgA nephropathy (IgAN) is the most commonly diagnosed primary glomerulonephritis worldwide. It is a slow progressing disease with approximately 30% of cases reaching end-stage kidney disease within 20 years of diagnosis. It is currently only diagnosed by an invasive biopsy and treatment options are limited. However, the current surge in interest in RNA interference is opening up new horizons for the use of this new technology in the field of IgAN management. A greater understanding of the fundamentals of RNA interference offers exciting possibilities both for biomarker discovery and, more importantly, for novel therapeutic approaches to target key pathogenic pathways in IgAN. This review aims to summarise the RNA interference literature in the context of microRNAs and their association with the multifaceted aspects of IgA nephropathy.
Collapse
|
356
|
Evaluation of Floxuridine Oligonucleotide Conjugates Carrying Potential Enhancers of Cellular Uptake. Int J Mol Sci 2021; 22:ijms22115678. [PMID: 34073599 PMCID: PMC8199350 DOI: 10.3390/ijms22115678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Conjugation of small molecules such as lipids or receptor ligands to anti-cancer drugs has been used to improve their pharmacological properties. In this work, we studied the biological effects of several small-molecule enhancers into a short oligonucleotide made of five floxuridine units. Specifically, we studied adding cholesterol, palmitic acid, polyethyleneglycol (PEG 1000), folic acid and triantennary N-acetylgalactosamine (GalNAc) as potential enhancers of cellular uptake. As expected, all these molecules increased the internalization efficiency with different degrees depending on the cell line. The conjugates showed antiproliferative activity due to their metabolic activation by nuclease degradation generating floxuridine monophosphate. The cytotoxicity and apoptosis assays showed an increase in the anti-cancer activity of the conjugates related to the floxuridine oligomer, but this effect did not correlate with the internalization results. Palmitic and folic acid conjugates provide the highest antiproliferative activity without having the highest internalization results. On the contrary, cholesterol oligomers that were the best-internalized oligomers had poor antiproliferative activity, even worse than the unmodified floxuridine oligomer. Especially relevant is the effect induced by palmitic and folic acid derivatives generating the most active drugs. These results are of special interest for delivering other therapeutic oligonucleotides.
Collapse
|
357
|
Forbes TA, Brown BD, Lai C. Therapeutic RNA interference: A novel approach to the treatment of primary hyperoxaluria. Br J Clin Pharmacol 2021; 88:2525-2538. [PMID: 34022071 PMCID: PMC9291495 DOI: 10.1111/bcp.14925] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/19/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
RNA interference (RNAi) is a natural biological pathway that inhibits gene expression by targeted degradation or translational inhibition of cytoplasmic mRNA by the RNA induced silencing complex. RNAi has long been exploited in laboratory research to study the biological consequences of the reduced expression of a gene of interest. More recently RNAi has been demonstrated as a therapeutic avenue for rare metabolic diseases. This review presents an overview of the cellular RNAi machinery as well as therapeutic RNAi design and delivery. As a clinical example we present primary hyperoxaluria, an ultrarare inherited disease of increased hepatic oxalate production which leads to recurrent calcium oxalate kidney stones. In the most common form of the disease (Type 1), end‐stage kidney disease occurs in childhood or young adulthood, often necessitating combined kidney and liver transplantation. In this context we discuss nedosiran (Dicerna Pharmaceuticals, Inc.) and lumasiran (Alnylam Pharmaceuticals), which are both novel RNAi therapies for primary hyperoxaluria that selectively reduce hepatic expression of lactate dehydrogenase and glycolate oxidase respectively, reducing hepatic oxalate production and urinary oxalate levels. Finally, we consider future optimizations advances in RNAi therapies.
Collapse
Affiliation(s)
- Thomas A Forbes
- Royal Children's Hospital, Parkville, Victoria, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
358
|
Overcoming the challenges of tissue delivery for oligonucleotide therapeutics. Trends Pharmacol Sci 2021; 42:588-604. [PMID: 34020790 DOI: 10.1016/j.tips.2021.04.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022]
Abstract
Synthetic therapeutic oligonucleotides (STO) represent the third bonafide platform for drug discovery in the pharmaceutical industry after small molecule and protein therapeutics. So far, thirteen STOs have been approved by regulatory agencies and over one hundred of them are in different stages of clinical trials. STOs hybridize to their target RNA or DNA in cells via Watson-Crick base pairing to exert their pharmacological effects. This unique class of therapeutic agents has the potential to target genes and gene products that are considered undruggable by other therapeutic platforms. However, STOs must overcome several extracellular and intracellular obstacles to interact with their biological RNA targets inside cells. These obstacles include degradation by extracellular nucleases, scavenging by the reticuloendothelial system, filtration by the kidney, traversing the capillary endothelium to access the tissue interstitium, cell-surface receptor-mediated endocytic uptake, and escape from endolysosomal compartments to access the nuclear and/or cytoplasmic compartments where their targets reside. In this review, we present the recent advances in this field with a specific focus on antisense oligonucleotides (ASOs) and siRNA therapeutics.
Collapse
|
359
|
Smart Nucleic Acids as Future Therapeutics. Trends Biotechnol 2021; 39:1289-1307. [PMID: 33980422 DOI: 10.1016/j.tibtech.2021.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 11/23/2022]
Abstract
Nucleic acid therapeutics (NATs) hold promise in treating undruggable diseases and are recognized as the third major category of therapeutics in addition to small molecules and antibodies. Despite the milestones that NATs have made in clinical translation over the past decade, one important challenge pertains to increasing the specificity of this class of drugs. Activating NATs exclusively in disease-causing cells is highly desirable because it will safely broaden the application of NATs to a wider range of clinical indications. Smart NATs are triggered through a photo-uncaging reaction or a specific molecular input such as a transcript, protein, or small molecule, thus complementing the current strategy of targeting cells and tissues with receptor-specific ligands to enhance specificity. This review summarizes the programmable modalities that have been incorporated into NATs to build in responsive behaviors. We discuss the various inputs, transduction mechanisms, and output response functions that have been demonstrated to date.
Collapse
|
360
|
Inclisiran: A Novel Agent for Lowering Apolipoprotein B-Containing Lipoproteins. J Cardiovasc Pharmacol 2021; 78:e157-e174. [PMID: 33990512 DOI: 10.1097/fjc.0000000000001053] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022]
Abstract
ABSTRACT Hypercholesterolemia is a leading cause of cardiovascular morbidity and mortality. Accordingly, efforts to lower apolipoprotein B-containing lipoproteins in plasma are the centerpiece of strategies for cardiovascular prevention and treatment in primary and secondary management. Despite the importance of this endeavor, many patients do not achieve appropriate low density lipoprotein cholesterol (LDL-C) and non-high density lipoprotein cholesterol (non-HDL-C) goals, even among those who have experienced atherosclerotic cardiovascular disease (ASCVD). The development of new LDL-C-lowering medications with alternative mechanisms of action will facilitate improved goal achievement in high risk patients. Inclisiran is a novel small interfering ribonucleic acid (siRNA)-based drug that is experimental in the US and approved for clinical use in the EU. It lowers LDL-C and other apolipoprotein B-containing lipoproteins by reducing production of Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9), a protein that normally contributes to LDL-receptor (LDLR) degradation, thereby increasing LDLR density and recycling in hepatocytes. Although the lipid-lowering efficacy of inclisiran is comparable to results achieved with PCSK9-blocking monoclonal antibodies (PCSK9i) (alirocumab and evolocumab), there are several important differences between the two drug classes. First, inclisiran reduces levels of PCSK9 both intracellularly and extracellularly by blocking translation of and degrading PCSK9 messenger RNA. Second, the long biological half-life of inclisiran produces sustained LDL-C-lowering with twice yearly dosing. Third, although PCSK9i drugs are proven to reduce ASCVD events, clinical outcomes trials with inclisiran are still in progress. In this manuscript, we review the clinical development of inclisiran, its mechanism of action, lipid-lowering efficacy, safety and tolerability, and potential clinical role of this promising new agent.
Collapse
|
361
|
Schlich M, Palomba R, Costabile G, Mizrahy S, Pannuzzo M, Peer D, Decuzzi P. Cytosolic delivery of nucleic acids: The case of ionizable lipid nanoparticles. Bioeng Transl Med 2021; 6:e10213. [PMID: 33786376 PMCID: PMC7995196 DOI: 10.1002/btm2.10213] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Ionizable lipid nanoparticles (LNPs) are the most clinically advanced nano-delivery system for therapeutic nucleic acids. The great effort put in the development of ionizable lipids with increased in vivo potency brought LNPs from the laboratory benches to the FDA approval of patisiran in 2018 and the ongoing clinical trials for mRNA-based vaccines against SARS-CoV-2. Despite these success stories, several challenges remain in RNA delivery, including what is known as "endosomal escape." Reaching the cytosol is mandatory for unleashing the therapeutic activity of RNA molecules, as their accumulation in other intracellular compartments would simply result in efficacy loss. In LNPs, the ability of ionizable lipids to form destabilizing non-bilayer structures at acidic pH is recognized as the key for endosomal escape and RNA cytosolic delivery. This is motivating a surge in studies aiming at designing novel ionizable lipids with improved biodegradation and safety profiles. In this work, we describe the journey of RNA-loaded LNPs across multiple intracellular barriers, from the extracellular space to the cytosol. In silico molecular dynamics modeling, in vitro high-resolution microscopy analyses, and in vivo imaging data are systematically reviewed to distill out the regulating mechanisms underlying the endosomal escape of RNA. Finally, a comparison with strategies employed by enveloped viruses to deliver their genetic material into cells is also presented. The combination of a multidisciplinary analytical toolkit for endosomal escape quantification and a nature-inspired design could foster the development of future LNPs with improved cytosolic delivery of nucleic acids.
Collapse
Affiliation(s)
- Michele Schlich
- Fondazione Istituto Italiano di TecnologiaLaboratory of Nanotechnology for Precision MedicineGenoaItaly
- Department of Life and Environmental SciencesUniversity of CagliariCagliariItaly
| | - Roberto Palomba
- Fondazione Istituto Italiano di TecnologiaLaboratory of Nanotechnology for Precision MedicineGenoaItaly
| | - Gabriella Costabile
- Fondazione Istituto Italiano di TecnologiaLaboratory of Nanotechnology for Precision MedicineGenoaItaly
| | - Shoshy Mizrahy
- Fondazione Istituto Italiano di TecnologiaLaboratory of Nanotechnology for Precision MedicineGenoaItaly
- Laboratory of Precision NanoMedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel AvivIsrael
- Cancer Biology Research CenterTel Aviv UniversityTel AvivIsrael
| | - Martina Pannuzzo
- Fondazione Istituto Italiano di TecnologiaLaboratory of Nanotechnology for Precision MedicineGenoaItaly
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel AvivIsrael
- Cancer Biology Research CenterTel Aviv UniversityTel AvivIsrael
| | - Paolo Decuzzi
- Fondazione Istituto Italiano di TecnologiaLaboratory of Nanotechnology for Precision MedicineGenoaItaly
| |
Collapse
|
362
|
Distribution and biotransformation of therapeutic antisense oligonucleotides and conjugates. Drug Discov Today 2021; 26:2244-2258. [PMID: 33862193 DOI: 10.1016/j.drudis.2021.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/09/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023]
Abstract
Drug properties of antisense oligonucleotides (ASOs) differ significantly from those of traditional small-molecule therapeutics. In this review, we focus on ASO disposition, mainly as characterized by distribution and biotransformation, of nonconjugated and conjugated ASOs. We introduce ASO chemistry to allow the following in-depth discussion on bioanalytical methods and determination of distribution and elimination kinetics at low concentrations over extended periods of time. The resulting quantitative data on the parent oligonucleotide, and the identification and quantification of formed metabolites define the disposition. Proper quantitative understanding of disposition is pivotal for nonclinical to clinical predictions, supports communication with health agencies, and increases the probability of delivering optimal ASO therapy to patients.
Collapse
|
363
|
Xu M, Zhang K, Song J. Targeted Therapy in Cardiovascular Disease: A Precision Therapy Era. Front Pharmacol 2021; 12:623674. [PMID: 33935716 PMCID: PMC8085499 DOI: 10.3389/fphar.2021.623674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Targeted therapy refers to exploiting the specific therapeutic drugs against the pathogenic molecules (a protein or a gene) or cells. The drug specifically binds to disease-causing molecules or cells without affecting normal tissue, thus enabling personalized and precision treatment. Initially, therapeutic drugs included antibodies and small molecules, (e.g. nucleic acid drugs). With the advancement of the biology technology and immunotherapy, the gene editing and cell editing techniques are utilized for the disease treatment. Currently, targeted therapies applied to treat cardiovascular diseases (CVDs) mainly include protein drugs, gene editing technologies, nucleic acid drugs and cell therapy. Although targeted therapy has demonstrated excellent efficacy in pre-clinical and clinical trials, several limitations need to be recognized and overcome in clinical application, (e.g. off-target events, gene mutations, etc.). This review introduces the mechanisms of different targeted therapies, and mainly describes the targeted therapy applied in the CVDs. Furthermore, we made comparative analysis to clarify the advantages and disadvantages of different targeted therapies. This overview is expected to provide a new concept to the treatment of the CVDs.
Collapse
Affiliation(s)
- Mengda Xu
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailun Zhang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Kailun Zhang, ; Jiangping Song,
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China,*Correspondence: Kailun Zhang, ; Jiangping Song,
| |
Collapse
|
364
|
Letter to the Editor: Brain renin-angiotensin system and liver-directed siRNA targeted to angiotensinogen. Clin Sci (Lond) 2021; 135:907-910. [PMID: 33835151 DOI: 10.1042/cs20210163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/20/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022]
|
365
|
Vinarov Z, Abrahamsson B, Artursson P, Batchelor H, Berben P, Bernkop-Schnürch A, Butler J, Ceulemans J, Davies N, Dupont D, Flaten GE, Fotaki N, Griffin BT, Jannin V, Keemink J, Kesisoglou F, Koziolek M, Kuentz M, Mackie A, Meléndez-Martínez AJ, McAllister M, Müllertz A, O'Driscoll CM, Parrott N, Paszkowska J, Pavek P, Porter CJH, Reppas C, Stillhart C, Sugano K, Toader E, Valentová K, Vertzoni M, De Wildt SN, Wilson CG, Augustijns P. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Adv Drug Deliv Rev 2021; 171:289-331. [PMID: 33610694 DOI: 10.1016/j.addr.2021.02.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Although oral drug delivery is the preferred administration route and has been used for centuries, modern drug discovery and development pipelines challenge conventional formulation approaches and highlight the insufficient mechanistic understanding of processes critical to oral drug absorption. This review presents the opinion of UNGAP scientists on four key themes across the oral absorption landscape: (1) specific patient populations, (2) regional differences in the gastrointestinal tract, (3) advanced formulations and (4) food-drug interactions. The differences of oral absorption in pediatric and geriatric populations, the specific issues in colonic absorption, the formulation approaches for poorly water-soluble (small molecules) and poorly permeable (peptides, RNA etc.) drugs, as well as the vast realm of food effects, are some of the topics discussed in detail. The identified controversies and gaps in the current understanding of gastrointestinal absorption-related processes are used to create a roadmap for the future of oral drug absorption research.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Philippe Berben
- Pharmaceutical Development, UCB Pharma SA, Braine- l'Alleud, Belgium
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - James Butler
- GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gøril Eide Flaten
- Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | | | | | | | | - Martin Kuentz
- Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | - Alan Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | | | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Petr Pavek
- Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | | | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Kiyohiko Sugano
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Elena Toader
- Faculty of Medicine, University of Medicine and Pharmacy of Iasi, Romania
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saskia N De Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
366
|
LYTACs that engage the asialoglycoprotein receptor for targeted protein degradation. Nat Chem Biol 2021; 17:937-946. [PMID: 33767387 PMCID: PMC8387313 DOI: 10.1038/s41589-021-00770-1] [Citation(s) in RCA: 285] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Selective protein degradation platforms have afforded new development opportunities for therapeutics and tools for biological inquiry. The first lysosome targeting chimeras (LYTACs) targeted extracellular and membrane proteins for degradation by bridging a target protein to the cation-independent mannose-6-phosphate receptor (CI-M6PR). Here, we developed LYTACs that engage the asialoglycoprotein receptor (ASGPR), a liver-specific lysosomal targeting receptor, to degrade extracellular proteins in a cell type-specific manner. We conjugated binders to a tri-GalNAc motif that engages ASGPR to drive downregulation of proteins. Degradation of EGFR by GalNAc-LYTAC attenuated EGFR signaling compared to inhibition with an antibody. Furthermore, we demonstrated that a LYTAC comprising a 3.4 kDa peptide binder linked to a tri-GalNAc ligand degrades integrins and reduces cancer cell proliferation. Degradation with a single tri-GalNAc ligand prompted site-specific conjugation on antibody scaffolds, which improved the pharmacokinetic profile of GalNAc-LYTACs in vivo. GalNAc-LYTACs thus represent an avenue for cell-type restricted protein degradation.
Collapse
|
367
|
Swerdlow DI, Rider DA, Yavari A, Lindholm MW, Campion GV, Nissen SE. Treatment and prevention of lipoprotein(a)-mediated cardiovascular disease: the emerging potential of RNA interference therapeutics. Cardiovasc Res 2021; 118:1218-1231. [PMID: 33769464 PMCID: PMC8953457 DOI: 10.1093/cvr/cvab100] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/19/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid- and lipoprotein-modifying therapies have expanded substantially in the last 25 years, resulting in reduction in the incidence of major adverse cardiovascular events. However, no specific lipoprotein(a) [Lp(a)]-targeting therapy has yet been shown to reduce cardiovascular disease risk. Many epidemiological and genetic studies have demonstrated that Lp(a) is an important genetically determined causal risk factor for coronary heart disease, aortic valve disease, stroke, heart failure, and peripheral vascular disease. Accordingly, the need for specific Lp(a)-lowering therapy has become a major public health priority. Approximately 20% of the global population (1.4 billion people) have elevated levels of Lp(a) associated with higher cardiovascular risk, though the threshold for determining ‘high risk’ is debated. Traditional lifestyle approaches to cardiovascular risk reduction are ineffective at lowering Lp(a). To address a lifelong risk factor unmodifiable by non-pharmacological means, Lp(a)-lowering therapy needs to be safe, highly effective, and tolerable for a patient population who will likely require several decades of treatment. N-acetylgalactosamine-conjugated gene silencing therapeutics, such as small interfering RNA (siRNA) and antisense oligonucleotide targeting LPA, are ideally suited for this application, offering a highly tissue- and target transcript-specific approach with the potential for safe and durable Lp(a) lowering with as few as three or four doses per year. In this review, we evaluate the causal role of Lp(a) across the cardiovascular disease spectrum, examine the role of established lipid-modifying therapies in lowering Lp(a), and focus on the anticipated role for siRNA therapeutics in treating and preventing Lp(a)-related disease.
Collapse
Affiliation(s)
| | | | - Arash Yavari
- Experimental Therapeutics, Radcliffe, Department of Medicine, University of Oxford, UK
| | | | | | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
368
|
Shchegravina ES, Sachkova AA, Usova SD, Nyuchev AV, Gracheva YA, Fedorov AY. Carbohydrate Systems in Targeted Drug Delivery: Expectation and Reality. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1068162021010222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
369
|
Monty MA, Islam MA, Nan X, Tan J, Tuhin IJ, Tang X, Miao M, Wu D, Yu L. Emerging role of RNA interference in immune cells engineering and its therapeutic synergism in immunotherapy. Br J Pharmacol 2021; 178:1741-1755. [PMID: 33608889 DOI: 10.1111/bph.15414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
RNAi effectors (e.g. siRNA, shRNA and miRNA) can trigger the silencing of specific genes causing alteration of genomic functions becoming a new therapeutic area for the treatment of infectious diseases, neurodegenerative disorders and cancer. In cancer treatment, RNAi effectors showed potential immunomodulatory actions by down-regulating immuno-suppressive proteins, such as PD-1 and CTLA-4, which restrict immune cell function and present challenges in cancer immunotherapy. Therefore, compared with extracellular targeting by antibodies, RNAi-mediated cell-intrinsic disruption of inhibitory pathways in immune cells could promote an increased anti-tumour immune response. Along with non-viral vectors, DNA-based RNAi strategies might be a more promising method for immunomodulation to silence multiple inhibitory pathways in T cells than immune checkpoint blockade antibodies. Thus, in this review, we discuss diverse RNAi implementation strategies, with recent viral and non-viral mediated RNAi synergism to immunotherapy that augments the anti-tumour immunity. Finally, we provide the current progress of RNAi in clinical pipeline.
Collapse
Affiliation(s)
- Masuma Akter Monty
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Md Ariful Islam
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Nan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Jingwen Tan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Israth Jahan Tuhin
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xiaowen Tang
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miao Miao
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| |
Collapse
|
370
|
Subhan MA, Attia SA, Torchilin VP. Advances in siRNA delivery strategies for the treatment of MDR cancer. Life Sci 2021; 274:119337. [PMID: 33713664 DOI: 10.1016/j.lfs.2021.119337] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022]
Abstract
RNA interference (RNAi) represents a promising therapeutic method that uses siRNA for cancer treatment. Although the RNAi technique has been increasingly used for clinical trials, systemic siRNA delivery into targeted cells is still challenging. The barriers impeding siRNA therapeutics delivery and impacting the treatment outcome must overcome with negligible systemic toxicity for a desirable and successful delivery of siRNA to MDR cancer cells. Nano delivery strategies have been investigated for nanocarrier functionalization, cancer immunotherapy and cancer targeting. Lipid nanoparticles (LNPs), dynamic polyconjugates (DPC™), GalNAc-siRNA conjugates, exosome and RBC systems have shown potential for efficient delivery of siRNA to cancer cells. Delivery of siRNA to tumor cells, immune cells to regulate T cell functions for immunotherapy are promising approaches.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh.
| | - Sara Aly Attia
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; Department of Oncology, Radiotherapy and Plastic Surgery I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
371
|
de la Fuente IF, Sawant SS, Tolentino MQ, Corrigan PM, Rouge JL. Viral Mimicry as a Design Template for Nucleic Acid Nanocarriers. Front Chem 2021; 9:613209. [PMID: 33777893 PMCID: PMC7987652 DOI: 10.3389/fchem.2021.613209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Therapeutic nucleic acids hold immense potential in combating undruggable, gene-based diseases owing to their high programmability and relative ease of synthesis. While the delivery of this class of therapeutics has successfully entered the clinical setting, extrahepatic targeting, endosomal escape efficiency, and subcellular localization. On the other hand, viruses serve as natural carriers of nucleic acids and have acquired a plethora of structures and mechanisms that confer remarkable transfection efficiency. Thus, understanding the structure and mechanism of viruses can guide the design of synthetic nucleic acid vectors. This review revisits relevant structural and mechanistic features of viruses as design considerations for efficient nucleic acid delivery systems. This article explores how viral ligand display and a metastable structure are central to the molecular mechanisms of attachment, entry, and viral genome release. For comparison, accounted for are details on the design and intracellular fate of existing nucleic acid carriers and nanostructures that share similar and essential features to viruses. The review, thus, highlights unifying themes of viruses and nucleic acid delivery systems such as genome protection, target specificity, and controlled release. Sophisticated viral mechanisms that are yet to be exploited in oligonucleotide delivery are also identified as they could further the development of next-generation nonviral nucleic acid vectors.
Collapse
Affiliation(s)
| | | | | | | | - Jessica L. Rouge
- Department of Chemistry, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
372
|
Liu R, Zuo R, Hudalla GA. Harnessing molecular recognition for localized drug delivery. Adv Drug Deliv Rev 2021; 170:238-260. [PMID: 33484737 PMCID: PMC8274479 DOI: 10.1016/j.addr.2021.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022]
Abstract
A grand challenge in drug delivery is providing the right dose, at the right anatomic location, for the right duration of time to maximize therapeutic efficacy while minimizing off-target toxicity and other deleterious side-effects. Two general modalities are receiving broad attention for localized drug delivery. In the first, referred to as "targeted accumulation", drugs or drug carriers are engineered to have targeting moieties that promote their accumulation at a specific tissue site from circulation. In the second, referred to as "local anchoring", drugs or drug carriers are inserted directly into the tissue site of interest where they persist for a specified duration of time. This review surveys recent advances in harnessing molecular recognition between proteins, peptides, nucleic acids, lipids, and carbohydrates to mediate targeted accumulation and local anchoring of drugs and drug carriers.
Collapse
Affiliation(s)
- Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
373
|
Murphy D, de Jong OG, Evers MJW, Nurazizah M, Schiffelers RM, Vader P. Natural or Synthetic RNA Delivery: A Stoichiometric Comparison of Extracellular Vesicles and Synthetic Nanoparticles. NANO LETTERS 2021; 21:1888-1895. [PMID: 33570966 PMCID: PMC8023702 DOI: 10.1021/acs.nanolett.1c00094] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
RNA therapeutics have high potential that is yet to be fully realized, largely due to challenges involved in the appropriate delivery to target cells. Extracellular vesicles (EVs) are lipid bound nanoparticles released by cells of all types and possess numerous features that may help overcome this hurdle and have emerged as a promising RNA delivery vehicle candidate. Despite extensive research into the engineering of EVs for RNA delivery, it remains unclear how the intrinsic RNA delivery efficiency of EVs compares to currently used synthetic RNA delivery vehicles. Using a novel CRISPR/Cas9-based RNA transfer reporter system, we compared the delivery efficiency of EVs to clinically approved state-of-the-art DLin-MC3-DMA lipid nanoparticles and several in vitro transfection reagents. We found that EVs delivered RNA several orders of magnitude more efficiently than these synthetic systems. This finding supports the continued research into EVs as potential RNA delivery vehicles.
Collapse
Affiliation(s)
- Daniel
E. Murphy
- CDL
Research, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | - Olivier G. de Jong
- CDL
Research, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | - Martijn J. W. Evers
- CDL
Research, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | | | | | - Pieter Vader
- CDL
Research, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
- Department
of Experimental Cardiology, University Medical
Center Utrecht, Utrecht 3584CX, The Netherlands
| |
Collapse
|
374
|
Cook A, Decuzzi P. Harnessing Endogenous Stimuli for Responsive Materials in Theranostics. ACS NANO 2021; 15:2068-2098. [PMID: 33555171 PMCID: PMC7905878 DOI: 10.1021/acsnano.0c09115] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/02/2021] [Indexed: 05/04/2023]
Abstract
Materials that respond to endogenous stimuli are being leveraged to enhance spatiotemporal control in a range of biomedical applications from drug delivery to diagnostic tools. The design of materials that undergo morphological or chemical changes in response to specific biological cues or pathologies will be an important area of research for improving efficacies of existing therapies and imaging agents, while also being promising for developing personalized theranostic systems. Internal stimuli-responsive systems can be engineered across length scales from nanometers to macroscopic and can respond to endogenous signals such as enzymes, pH, glucose, ATP, hypoxia, redox signals, and nucleic acids by incorporating synthetic bio-inspired moieties or natural building blocks. This Review will summarize response mechanisms and fabrication strategies used in internal stimuli-responsive materials with a focus on drug delivery and imaging for a broad range of pathologies, including cancer, diabetes, vascular disorders, inflammation, and microbial infections. We will also discuss observed challenges, future research directions, and clinical translation aspects of these responsive materials.
Collapse
Affiliation(s)
- Alexander
B. Cook
- Laboratory of Nanotechnology
for Precision Medicine, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology
for Precision Medicine, Istituto Italiano
di Tecnologia, Via Morego
30, 16163 Genova, Italy
| |
Collapse
|
375
|
Gangopadhyay S, Nikam RR, Gore KR. Folate Receptor-Mediated siRNA Delivery: Recent Developments and Future Directions for RNAi Therapeutics. Nucleic Acid Ther 2021; 31:245-270. [PMID: 33595381 DOI: 10.1089/nat.2020.0882] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RNA interference (RNAi), a gene regulatory process mediated by small interfering RNAs (siRNAs), has made remarkable progress as a potential therapeutic agent against various diseases. However, RNAi is associated with fundamental challenges such as poor systemic delivery and susceptibility to the nucleases. Targeting ligand-bound delivery vehicles has improved the accumulation of drug at the target site, which has resulted in high transfection efficiency and enhanced gene silencing. Recently, folate receptor (FR)-mediated targeted delivery of siRNAs has garnered attention due to their enhanced cellular uptake and high transfection efficiency toward tumor cells. Folic acid (FA), due to its small size, low immunogenicity, high in vivo stability, and high binding affinity toward FRs, has attracted much attention for targeted siRNA delivery. FRs are overexpressed in a large number of tumors, including ovarian, breast, kidney, and lung cancer cells. In this review, we discuss recent advances in FA-mediated siRNA delivery to treat cancers and inflammatory diseases. This review summarizes various FA-conjugated nanoparticle systems reported so far in the literature, including liposome, silica, metal, graphene, dendrimers, chitosan, organic copolymers, and RNA nanoparticles. This review will help in the design and development of potential delivery vehicles for siRNA drug targeting to tumor cells using an FR-mediated approach.
Collapse
Affiliation(s)
- Sumit Gangopadhyay
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rahul R Nikam
- Department of Chemistry, University of Mumbai, Mumbai, India
| | - Kiran R Gore
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
376
|
Liu B, Wang B, Zhang X, Lock R, Nash T, Vunjak-Novakovic G. Cell type-specific microRNA therapies for myocardial infarction. Sci Transl Med 2021; 13:eabd0914. [PMID: 33568517 PMCID: PMC8848299 DOI: 10.1126/scitranslmed.abd0914] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 01/19/2021] [Indexed: 12/13/2022]
Abstract
Current interventions fail to recover injured myocardium after infarction and prompt the need for development of cardioprotective strategies. Of increasing interest is the therapeutic use of microRNAs to control gene expression through specific targeting of mRNAs. In this Review, we discuss current microRNA-based therapeutic strategies, describing the outcomes and limitations of key microRNAs with a focus on target cell types and molecular pathways. Last, we offer a perspective on the outlook of microRNA therapies for myocardial infarction, highlighting the outstanding challenges and emerging strategies.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bryan Wang
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor Nash
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Columbia University, New York, NY 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| |
Collapse
|
377
|
Physiologically based pharmacokinetic (PBPK) modeling of RNAi therapeutics: Opportunities and challenges. Biochem Pharmacol 2021; 189:114468. [PMID: 33577889 DOI: 10.1016/j.bcp.2021.114468] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is a powerful tool with many demonstrated applications in various phases of drug development and regulatory review. RNA interference (RNAi)-based therapeutics are a class of drugs that have unique pharmacokinetic properties and mechanisms of action. With an increasing number of RNAi therapeutics in the pipeline and reaching the market, there is a considerable amount of active research in this area requiring a multidisciplinary approach. The application of PBPK models for RNAi therapeutics is in its infancy and its utility to facilitate the development of this new class of drugs is yet to be fully evaluated. From this perspective, we briefly discuss some of the current computational modeling approaches used in support of efficient development and approval of RNAi therapeutics. Considerations for PBPK model development are highlighted both in a relative context between small molecules and large molecules such as monoclonal antibodies and as it applies to RNAi therapeutics. In addition, the prospects for drawing upon other recognized avenues of PBPK modeling and some of the foreseeable challenges in PBPK model development for these chemical modalities are briefly discussed. Finally, an exploration of the potential application of PBPK model development for RNAi therapeutics is provided. We hope these preliminary thoughts will help initiate a dialogue between scientists in the relevant sectors to examine the value of PBPK modeling for RNAi therapeutics. Such evaluations could help standardize the practice in the future and support appropriate guidance development for strengthening the RNAi therapeutics development program.
Collapse
|
378
|
Wang H, Zhang S, Lv J, Cheng Y. Design of polymers for siRNA delivery: Recent progress and challenges. VIEW 2021. [DOI: 10.1002/viw.20200026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou China
| | - Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou China
| | - Jia Lv
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology School of Molecular Science and Engineering South China University of Technology Guangzhou China
- Shanghai Key Laboratory of Regulatory Biology School of Life Sciences East China Normal University Shanghai China
| |
Collapse
|
379
|
Katzmann JL, Packard CJ, Chapman MJ, Katzmann I, Laufs U. Targeting RNA With Antisense Oligonucleotides and Small Interfering RNA: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 76:563-579. [PMID: 32731935 DOI: 10.1016/j.jacc.2020.05.070] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022]
Abstract
There is an unmet clinical need to reduce residual cardiovascular risk attributable to apolipoprotein B-containing lipoproteins, particularly low-density lipoprotein and remnant particles. Pharmacological targeting of messenger RNA represents an emerging, innovative approach. Two major classes of agents have been developed-antisense oligonucleotides and small interfering RNA. Early problems with their use have been overcome by conjugation with N-acetylgalactosamine, an adduct that targets their delivery to the primary site of action in the liver. Using these agents to inhibit the translation of key regulatory proteins such as PCSK9, apolipoprotein CIII, apolipoprotein(a), and angiopoietin-like 3 has been shown to be effective in attenuating dyslipidemic states. Cardiovascular outcome trials with N-acetylgalactosamine-conjugated RNA-targeting drugs are ongoing. The advantages of these agents include long dosing intervals of up to 6 months and the potential to regulate the abundance of any disease-related protein. Long-term safety has yet to be demonstrated in large-scale clinical trials.
Collapse
Affiliation(s)
- Julius L Katzmann
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - M John Chapman
- Endocrinology-Metabolism Division, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France; National Institute for Health and Medical Research (INSERM), Paris, France
| | - Isabell Katzmann
- Department of Internal Medicine, Zeisigwaldkliniken Bethanien Chemnitz, Chemnitz, Germany
| | - Ulrich Laufs
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
380
|
Aho A, Äärelä A, Korhonen H, Virta P. Expanding the Scope of the Cleavable N-(methoxy)oxazolidine Linker for the Synthesis of Oligonucleotide Conjugates. Molecules 2021; 26:490. [PMID: 33477693 PMCID: PMC7838870 DOI: 10.3390/molecules26020490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Oligonucleotides modified by a 2'-deoxy-2'-(N-methoxyamino) ribonucleotide react readily with aldehydes in slightly acidic conditions to yield the corresponding N-(methoxy)oxazolidine-linked oligonucleotide-conjugates. The reaction is reversible and dynamic in slightly acidic conditions, while the products are virtually stable above pH 7, where the reaction is in a ''switched off-state''. Small molecular examinations have demonstrated that aldehyde constituents affect the cleavage rate of the N-(methoxy)oxazolidine-linkage. This can be utilized to adjust the stability of this pH-responsive cleavable linker for drug delivery applications. In the present study, Fmoc-β-Ala-H was immobilized to a serine-modified ChemMatrix resin and used for the automated assembly of two peptidealdehydes and one aldehyde-modified peptide nucleic acid (PNA). In addition, a triantennary N-acetyl-d-galactosamine-cluster with a β-Ala-H unit has been synthesized. These aldehydes were conjugated via N-(methoxy)oxazolidine-linkage to therapeutically relevant oligonucleotide phosphorothioates and one DNA-aptamer in 19-47% isolated yields. The cleavage rates of the conjugates were studied in slightly acidic conditions. In addition to the diverse set of conjugates synthesized, these experiments and a comparison to published data demonstrate that the simple conversion of Gly-H to β-Ala-H residue resulted in a faster cleavage of the N-(methoxy)oxazolidine-linker at pH 5, being comparable (T0.5 ca 7 h) to hydrazone-based structures.
Collapse
Affiliation(s)
| | | | | | - Pasi Virta
- Department of Chemistry, University of Turku, 20014 Turku, Finland; (A.A.); (A.Ä.); (H.K.)
| |
Collapse
|
381
|
Impact of Galactoxyloglucan Coated Iron Oxide Nanoparticles on Reactive Oxygen Species Generation and Magnetic Resonance Imaging for Tumor Management. J CLUST SCI 2021. [DOI: 10.1007/s10876-020-01971-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
382
|
Fakih HH, Katolik A, Malek-Adamian E, Fakhoury JJ, Kaviani S, Damha MJ, Sleiman HF. Design and enhanced gene silencing activity of spherical 2'-fluoroarabinose nucleic acids (FANA-SNAs). Chem Sci 2021; 12:2993-3003. [PMID: 34164068 PMCID: PMC8179377 DOI: 10.1039/d0sc06645a] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Drug delivery vectors for nucleic acid therapeutics (NATs) face significant barriers for translation into the clinic. Spherical nucleic acids (SNAs) – nanoparticles with an exterior shell made up of DNA strands and a hydrophobic interior – have recently shown great potential as vehicles to improve the biodistribution and efficacy of NATs. To date, SNA design has not taken advantage of the powerful chemical modifications available to NATs. Here, we modify SNAs with 2′-deoxy-2′-fluoro-d-arabinonucleic acid (FANA-SNA), and show increased stability, enhanced gene silencing potency and unaided uptake (gymnosis) as compared to free FANA. By varying the spacer region between the nucleic acid strand and the attached hydrophobic polymer, we show that a cleavable DNA based spacer is essential for maximum activity. This design feature will be important when implementing functionalized nucleic acids into nanostructures for gene silencing. The modularity of the FANA-SNA was demonstrated by silencing two different targets. Transfection-free delivery was superior for the modified SNA compared to the free FANA oligonucleotide. Optimizing FANA modified spherical nucleic acids (FANA-SNAs) for highly efficient delivery of nucleic acid therapeutics.![]()
Collapse
Affiliation(s)
- Hassan H Fakih
- Department of Chemistry, McGill University Montreal Quebec H3A 0B8 Canada
| | - Adam Katolik
- Department of Chemistry, McGill University Montreal Quebec H3A 0B8 Canada
| | | | - Johans J Fakhoury
- Department of Chemistry, McGill University Montreal Quebec H3A 0B8 Canada
| | - Sepideh Kaviani
- Department of Chemistry, McGill University Montreal Quebec H3A 0B8 Canada
| | - Masad J Damha
- Department of Chemistry, McGill University Montreal Quebec H3A 0B8 Canada
| | - Hanadi F Sleiman
- Department of Chemistry, McGill University Montreal Quebec H3A 0B8 Canada
| |
Collapse
|
383
|
Zavoiura O, Brunner B, Casteels P, Zimmermann L, Ozog M, Boutton C, Helms MW, Wagenaar T, Adam V, Peterka J, Metz-Weidmann C, Deschaght P, Scheidler S, Jahn-Hofmann K. Nanobody-siRNA Conjugates for Targeted Delivery of siRNA to Cancer Cells. Mol Pharm 2021; 18:1048-1060. [PMID: 33444501 DOI: 10.1021/acs.molpharmaceut.0c01001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Targeted extrahepatic delivery of siRNA remains a challenging task in the field of nucleic acid therapeutics. An ideal delivery tool must internalize siRNA exclusively into the cells of interest without affecting the silencing activity of siRNA. Here, we report the use of anti-EGFR Nanobodies (trademark of Ablynx N.V.) as tools for targeted siRNA delivery. A straightforward procedure for site-specific conjugation of siRNA to an engineered C-terminal cysteine residue on the Nanobody (trademark of Ablynx N.V.) is described. We show that siRNA-conjugated Nanobodies (Nb-siRNA) retain their binding to EGFR and enter EGFR-positive cells via receptor-mediated endocytosis. The activity of Nb-siRNAs was assessed by measuring the knockdown of a housekeeping gene (AHSA1) in EGFR-positive and EGFR-negative cells. We demonstrate that Nb-siRNAs are active in vitro and induce mRNA cleavage in the targeted cell line. In addition, we discuss the silencing activity of siRNA conjugated to fused Nbs with various combinations of EGFR-binding building blocks. Finally, we compare the performance of Nb-siRNA joined by four different linkers and discuss the advantages and limitations of using cleavable and noncleavable linkers in the context of Nanobody-mediated siRNA delivery.
Collapse
Affiliation(s)
- Oleksandr Zavoiura
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Bodo Brunner
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Peter Casteels
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Luciana Zimmermann
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Matthias Ozog
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Carlo Boutton
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Mike W Helms
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Timothy Wagenaar
- Sanofi, Research, 640 Memorial Drive, Cambridge, 02139 Massachusetts, United States
| | - Volker Adam
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Josefine Peterka
- Sanofi, TIDES platform, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | | | - Pieter Deschaght
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Sabine Scheidler
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Kerstin Jahn-Hofmann
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
384
|
Honcharenko D, Druceikaite K, Honcharenko M, Bollmark M, Tedebark U, Strömberg R. New Alkyne and Amine Linkers for Versatile Multiple Conjugation of Oligonucleotides. ACS OMEGA 2021; 6:579-593. [PMID: 33458510 PMCID: PMC7807750 DOI: 10.1021/acsomega.0c05075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/08/2020] [Indexed: 05/08/2023]
Abstract
Oligonucleotide (ON) conjugates are increasingly important tools for various molecular diagnostics, nanotechnological applications, and for the development of nucleic acid-based therapies. Multiple labeling of ONs can further equip ON-conjugates and provide improved or additional tailored properties. Typically, the preparation of ON multiconjugates involves additional synthetic steps and/or manipulations in post-ON assembly. This report describes the simplified methodology allowing for multiple labeling of ONs on a solid support and is compatible with phosphodiester as well as phosphorothioate (PS) ONs. The current approach utilizes two novel alkyne- and amino-functionalized linker phosphoramidites that can be readily synthesized from a common aminodiol intermediate in three steps. The combination of new linkers provides orthogonal functionalities, which allow for multiple attachments of similar or varied moieties. The linkers are incorporated into ONs during automated solid-phase ON synthesis, and the conjugation with functional entities is achieved by either amide bond formation or by copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC). The versatility of the approach is demonstrated by the synthesis of 5'-site ON multiconjugates with small molecules, peptides, and fatty acids as well as in the preparation of an internal peptide-ON conjugate.
Collapse
Affiliation(s)
- Dmytro Honcharenko
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 14183 Huddinge, Sweden
| | - Kristina Druceikaite
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 14183 Huddinge, Sweden
- RISE
Chemical Process and Pharmaceutical Development, Forskargatan 20J, 15136 Södertälje, Sweden
| | | | - Martin Bollmark
- RISE
Chemical Process and Pharmaceutical Development, Forskargatan 20J, 15136 Södertälje, Sweden
| | - Ulf Tedebark
- RISE
Chemical Process and Pharmaceutical Development, Forskargatan 20J, 15136 Södertälje, Sweden
| | - Roger Strömberg
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 14183 Huddinge, Sweden
| |
Collapse
|
385
|
Varley AJ, Desaulniers JP. Chemical strategies for strand selection in short-interfering RNAs. RSC Adv 2021; 11:2415-2426. [PMID: 35424193 PMCID: PMC8693850 DOI: 10.1039/d0ra07747j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Therapeutic small interfering RNAs (siRNAs) are double stranded RNAs capable of potent and specific gene silencing through activation of the RNA interference (RNAi) pathway. The potential of siRNA drugs has recently been highlighted by the approval of multiple siRNA therapeutics. These successes relied heavily on chemically modified nucleic acids and their impact on stability, delivery, potency, and off-target effects. Despite remarkable progress, clinical trials still face failure due to off-target effects such as off-target gene dysregulation. Each siRNA strand can downregulate numerous gene targets while also contributing towards saturation of the RNAi machinery, leading to the upregulation of miRNA-repressed genes. Eliminating sense strand uptake effectively reduces off-target gene silencing and helps limit the disruption to endogenous regulatory mechanisms. Therefore, our understanding of strand selection has a direct impact on the success of future siRNA therapeutics. In this review, the approaches used to improve strand uptake are discussed and effective methods are summarized.
Collapse
Affiliation(s)
- Andrew J Varley
- Faculty of Science, University of Ontario Institute of Technology Oshawa Ontario L1G 0C5 Canada +1 905 721 3304 +1 905 721 8668 (ext. 3621)
| | - Jean-Paul Desaulniers
- Faculty of Science, University of Ontario Institute of Technology Oshawa Ontario L1G 0C5 Canada +1 905 721 3304 +1 905 721 8668 (ext. 3621)
| |
Collapse
|
386
|
Bockman MR, Dalal RJ, Kumar R, Reineke TM. Facile synthesis of GalNAc monomers and block polycations for hepatocyte gene delivery. Polym Chem 2021. [DOI: 10.1039/d1py00250c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Here, we present a facile synthetic route for a monomer displaying N-acetyl-d-galactosamine and subsequent copolymerization in a block format with cationic subunits readily accessing liver-targeted polymeric pDNA delivery vehicles with low toxicity.
Collapse
Affiliation(s)
| | - Rishad J. Dalal
- Department of Chemistry
- University of Minnesota
- Minneapolis
- USA
| | - Ramya Kumar
- Department of Chemistry
- University of Minnesota
- Minneapolis
- USA
| | | |
Collapse
|
387
|
Souza PVSD, Badia BDML, Farias IB, Gonçalves EA, Pinto WBVDR, Oliveira ASB. Acute hepatic porphyrias for the neurologist: current concepts and perspectives. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:68-80. [PMID: 33656101 DOI: 10.1590/0004-282x20200096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Acute hepatic porphyrias represent an expanding group of complex inherited metabolic disorders due to inborn errors of metabolism involving heme biosynthesis. OBJECTIVE We aimed to review the main clinical and therapeutic aspects associated with acute hepatic porphyrias. METHODS The authors provided a wide non-systematic review of current concepts and recently acquired knowledge about acute hepatic porphyrias. RESULTS Acute neurovisceral attacks are the most common and life-threatening presentation of this group and are often considered the main clinical manifestation by clinicians during differential diagnosis and the start of proper diagnostic work-up for acute porphyrias. However, atypical presentations with central nervous system involvement, neuropsychiatric disturbances, and some subtypes with photosensitivity usually make the definite diagnosis difficult and late. Early therapeutic interventions are essential during emergency treatment and intercritical periods to avoid recurrent severe presentations. The availability of new disease-modifying therapeutic proposals based on small interfering RNA (siRNA)-based therapies, complementary to the classic intravenous glucose infusion and hemin-based treatments, emphasizes the importance of early diagnosis and genetic counseling of patients. CONCLUSIONS This review article highlights the main biochemical, pathophysiological, clinical, and therapeutic aspects of acute hepatic porphyrias in clinical practice.
Collapse
Affiliation(s)
- Paulo Victor Sgobbi de Souza
- Universidade Federal de São Paulo, Department of Neurology and Neurosurgery, Division of Neuromuscular Diseases, São Paulo SP, Brazil
| | - Bruno de Mattos Lombardi Badia
- Universidade Federal de São Paulo, Department of Neurology and Neurosurgery, Division of Neuromuscular Diseases, São Paulo SP, Brazil
| | - Igor Braga Farias
- Universidade Federal de São Paulo, Department of Neurology and Neurosurgery, Division of Neuromuscular Diseases, São Paulo SP, Brazil
| | - Eduardo Augusto Gonçalves
- Universidade Federal de São Paulo, Department of Neurology and Neurosurgery, Division of Neuromuscular Diseases, São Paulo SP, Brazil
| | | | - Acary Souza Bulle Oliveira
- Universidade Federal de São Paulo, Department of Neurology and Neurosurgery, Division of Neuromuscular Diseases, São Paulo SP, Brazil
| |
Collapse
|
388
|
Yang Q, Humphreys SC, Lade JM, Li AP. Prolonged cultured human hepatocytes as an in vitro experimental system for the evaluation of potency and duration of activity of RNA therapeutics: Demonstration of prolonged duration of gene silencing effects of a GalNAc-conjugated human hypoxanthine phosphoribosyl transferase (HPRT1) siRNA. Biochem Pharmacol 2020; 189:114374. [PMID: 33358826 DOI: 10.1016/j.bcp.2020.114374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022]
Abstract
We report here the evaluation of a novel in vitro experimental model, prolonged cultured human hepatocytes (PCHC), as an experimental system to evaluate the potency and duration of effects of oligonucleotide therapeutics. A novel observation was made on the redifferentiation of PCHC upon prolonged culturing based on mRNA profiling of characteristic hepatic differentiation marker genes albumin, transferrin, and transthyretin. Consistent with the known de-differentiation of cultured human hepatocytes, decreases in marker gene expression were observed upon culturing of the hepatocytes for 2 days. A novel observation of re-differentiation was observed on day 7 as demonstrated by an increase in expression of the marker genes to levels similar to that observed on the first day of culture. The expression of the differentiation marker genes was highest on day 7, followed by a gradual decrease but remained higher than that on day 2 for up to the longest culture duration evaluated of 41 days. The redifferentiation phenomenon suggests that PCHC may be useful for the evaluation of the duration of effects of oligonucleotide therapeutics on gene expression in human hepatocytes. A proof of concept study was thereby conducted with PCHC with a GalNAc-conjugated siRNA targeting human hypoxanthine phosphoribosyl transferase1 (HPRT1). HPRT1 mRNA expression in siRNA-treated cultures decreased to 21% of that in untreated hepatocytes on day 1, <10% from days 2 to 12, <20% from days 16 to 33, and eventually recovered to 64% by day 41. Our results suggest that PCHC represent a clinically-relevant cost- and time-efficient experimental tool to aid in the evaluation of GalNAc-siRNA silencing activity, providing information on both efficacy and duration of efficacy. PCHC may be applicable in the drug development setting as a species- and cell type-relevant experimental tool to aid the development of oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Qian Yang
- In Vitro ADMET Laboratories Inc., Columbia, MD (QY, APL) and Amgen Inc., San Francisco, CA (SCH, JML), United States
| | - Sara C Humphreys
- In Vitro ADMET Laboratories Inc., Columbia, MD (QY, APL) and Amgen Inc., San Francisco, CA (SCH, JML), United States
| | - Julie M Lade
- In Vitro ADMET Laboratories Inc., Columbia, MD (QY, APL) and Amgen Inc., San Francisco, CA (SCH, JML), United States
| | - Albert P Li
- In Vitro ADMET Laboratories Inc., Columbia, MD (QY, APL) and Amgen Inc., San Francisco, CA (SCH, JML), United States.
| |
Collapse
|
389
|
Liver-targeted polymeric prodrugs of 8-aminoquinolines for malaria radical cure. J Control Release 2020; 331:213-227. [PMID: 33378692 DOI: 10.1016/j.jconrel.2020.12.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/18/2022]
Abstract
Primaquine and tafenoquine are the two 8-aminoquinoline (8-AQ) antimalarial drugs approved for malarial radical cure - the elimination of liver stage hypnozoites after infection with Plasmodium vivax. A single oral dose of tafenoquine leads to high efficacy against intra-hepatocyte hypnozoites after efficient first pass liver uptake and metabolism. Unfortunately, both drugs cause hemolytic anemia in G6PD-deficient humans. This toxicity prevents their mass administration without G6PD testing given the approximately 400 million G6PD deficient people across malarial endemic regions of the world. We hypothesized that liver-targeted delivery of 8-AQ prodrugs could maximize liver exposure and minimize erythrocyte exposure to increase their therapeutic window. Primaquine and tafenoquine were first synthesized as prodrug vinyl monomers with self-immolative hydrolytic linkers or cathepsin-cleavable valine-citrulline peptide linkers. RAFT polymerization was exploited to copolymerize these prodrug monomers with hepatocyte-targeting GalNAc monomers. Pharmacokinetic studies of released drugs after intravenous administration showed that the liver-to-plasma AUC ratios could be significantly improved, compared to parent drug administered orally. Single doses of the liver-targeted, enzyme-cleavable tafenoquine polymer were found to be as efficacious as an equivalent dose of the oral parent drug in the P. berghei causal prophylaxis model. They also elicited significantly milder hemotoxicity in the humanized NOD/SCID mouse model engrafted with red blood cells from G6PD deficient donors. The clinical application is envisioned as a single subcutaneous administration, and the lead tafenoquine polymer also showed excellent bioavailability and liver-to-blood ratios exceeding the IV administered polymer. The liver-targeted tafenoquine polymers warrant further development as a single-dose therapeutic via the subcutaneous route with the potential for broader patient administration without a requirement for G6PD diagnosis.
Collapse
|
390
|
Brown CR, Gupta S, Qin J, Racie T, He G, Lentini S, Malone R, Yu M, Matsuda S, Shulga-Morskaya S, Nair AV, Theile CS, Schmidt K, Shahraz A, Goel V, Parmar RG, Zlatev I, Schlegel MK, Nair JK, Jayaraman M, Manoharan M, Brown D, Maier MA, Jadhav V. Investigating the pharmacodynamic durability of GalNAc-siRNA conjugates. Nucleic Acids Res 2020; 48:11827-11844. [PMID: 32808038 PMCID: PMC7708070 DOI: 10.1093/nar/gkaa670] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
One hallmark of trivalent N-acetylgalactosamine (GalNAc)-conjugated siRNAs is the remarkable durability of silencing that can persist for months in preclinical species and humans. Here, we investigated the underlying biology supporting this extended duration of pharmacological activity. We found that siRNA accumulation and stability in acidic intracellular compartments is critical for long-term activity. We show that functional siRNA can be liberated from these compartments and loaded into newly generated Argonaute 2 protein complexes weeks after dosing, enabling continuous RNAi activity over time. Identical siRNAs delivered in lipid nanoparticles or as GalNAc conjugates were dose-adjusted to achieve similar knockdown, but only GalNAc–siRNAs supported an extended duration of activity, illustrating the importance of receptor-mediated siRNA trafficking in the process. Taken together, we provide several lines of evidence that acidic intracellular compartments serve as a long-term depot for GalNAc–siRNA conjugates and are the major contributor to the extended duration of activity observed in vivo.
Collapse
Affiliation(s)
| | - Swati Gupta
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - June Qin
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Timothy Racie
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Guo He
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Scott Lentini
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Ryan Malone
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Mikyung Yu
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | | | | | - Anil V Nair
- MGH Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Karyn Schmidt
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Azar Shahraz
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - Varun Goel
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | | | - Ivan Zlatev
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | | | | | | | | | - Dennis Brown
- MGH Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Vasant Jadhav
- Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| |
Collapse
|
391
|
Mishra A, Castañeda TR, Bader E, Elshorst B, Cummings S, Scherer P, Bangari DS, Loewe C, Schreuder H, Pöverlein C, Helms M, Jones S, Zech G, Licher T, Wagner M, Schudok M, de Hoop M, Plowright AT, Atzrodt J, Kannt A, Laitinen I, Derdau V. Triantennary GalNAc Molecular Imaging Probes for Monitoring Hepatocyte Function in a Rat Model of Nonalcoholic Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002997. [PMID: 33344141 PMCID: PMC7739951 DOI: 10.1002/advs.202002997] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/08/2020] [Indexed: 05/12/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease that can lead to irreversible liver cirrhosis and cancer. Early diagnosis of NASH is vital to detect disease before it becomes life-threatening, yet noninvasively differentiating NASH from simple steatosis is challenging. Herein, bifunctional probes have been developed that target the hepatocyte-specific asialoglycoprotein receptor (ASGPR), the expression of which decreases during NASH progression. The results show that the probes allow longitudinal, noninvasive monitoring of ASGPR levels by positron emission tomography in the newly developed rat model of NASH. The probes open new possibilities for research into early diagnosis of NASH and development of drugs to slow or reverse its progression.
Collapse
Affiliation(s)
| | - Tamara R. Castañeda
- R&D DiabetesSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
| | - Erik Bader
- Industriepark Höchst65926FrankfurtGermany
| | | | - Sheila Cummings
- Global Discovery PathologyTranslational In Vivo ModelsSanofi GenzymeThe Mountain RoadFraminghamMA01701USA
| | - Petra Scherer
- Global BioimagingTranslational In Vivo ModelsSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
| | - Dinesh S. Bangari
- Global Discovery PathologyTranslational In Vivo ModelsSanofi GenzymeThe Mountain RoadFraminghamMA01701USA
| | | | | | | | - Mike Helms
- Global BioimagingTranslational In Vivo ModelsSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
| | - Seth Jones
- Industriepark Höchst65926FrankfurtGermany
| | | | | | | | - Manfred Schudok
- R&D Drug Metabolism and PharmacokineticsSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
| | | | - Alleyn T. Plowright
- Industriepark Höchst65926FrankfurtGermany
- Wren Therapeutics Ltd.Department of ChemistryUniversity of CambridgeLensfield RdCambridgeCB2 1EWUK
| | - Jens Atzrodt
- R&D Transversal OperationsGerman R&D HubSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
| | - Aimo Kannt
- R&D DiabetesSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
- Experimental PharmacologyMedical Faculty MannheimUniversity of Heidelberg68167MannheimGermany
- Fraunhofer IMETranslational Medicine and Pharmacology60596FrankfurtGermany
| | - Iina Laitinen
- Global BioimagingTranslational In Vivo ModelsSanofi‐Aventis Deutschland GmbHIndustriepark Höchst65926FrankfurtGermany
- Present address:
Antaros Medical, Bioventure HubMölndal431 83Sweden
| | | |
Collapse
|
392
|
Salim L, Desaulniers JP. To Conjugate or to Package? A Look at Targeted siRNA Delivery Through Folate Receptors. Nucleic Acid Ther 2020; 31:21-38. [PMID: 33121373 DOI: 10.1089/nat.2020.0893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RNA interference (RNAi) applications have evolved from experimental tools to study gene function to the development of a novel class of gene-silencing therapeutics. Despite decades of research, it was not until August 2018 that the US FDA approved the first-ever RNAi drug, marking a new era for RNAi therapeutics. Although there are many limitations associated with the inherent structure of RNA, delivery to target cells and tissues remains the most challenging. RNAs are unable to diffuse across cellular membranes due to their large size and polyanionic backbone and, therefore, require a delivery vector. RNAi molecules can be conjugated to a targeting ligand or packaged into a delivery vehicle. Alnylam has used both strategies in their FDA-approved formulations to achieve efficient delivery to the liver. To harness the full potential of RNAi therapeutics, however, we must be able to target additional cells and tissues. One promising target is the folate receptor α, which is overexpressed in a variety of tumors despite having limited expression and distribution in normal tissues. Folate can be conjugated directly to the RNAi molecule or used to functionalize delivery vehicles. In this review, we compare both delivery strategies and discuss the current state of research in the area of folate-mediated delivery of RNAi molecules.
Collapse
Affiliation(s)
- Lidya Salim
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, Canada
| | | |
Collapse
|
393
|
Kumar P, Caruthers MH. DNA Analogues Modified at the Nonlinking Positions of Phosphorus. Acc Chem Res 2020; 53:2152-2166. [PMID: 32885957 DOI: 10.1021/acs.accounts.0c00078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemically modified oligonucleotides are being developed as a new class of medicines for curing conditions that previously remained untreatable. Three primary classes of therapeutic oligonucleotides are single-stranded antisense oligonucleotides (ASOs), double stranded small interfering RNAs (siRNAs), and oligonucleotides that induce exon skipping. Recently, ASOs, siRNAs, and exon skipping oligonucleotides have been approved for patients with unmet medical needs, and many other candidates are being tested in late stage clinical trials. In coming years, therapeutic oligonucleotides may match the promise of small molecules and antibodies. Interestingly, in the 1980s when we developed chemical methods for synthesizing oligonucleotides, no one would have imagined that these highly charged macromolecules could become future medicines. Indeed, the anionic nature and poor metabolic stability of the natural phosphodiester backbone provided a major challenge for the use of oligonucleotides as therapeutic drugs. Thus, chemical modifications of oligonucleotides were essential in order to improve their pharmacokinetic properties. Keeping this view in mind, my laboratory has developed a series of novel oligonucleotides where one or both nonbridging oxygens in the phosphodiester backbone are replaced with an atom or molecule that introduces molecular properties that enhance biological activity. We followed two complementary approaches. One was the use of phosphoramidites that could act directly as synthons for the solid phase synthesis of oligonucleotide analogues. This approach sometimes was not feasible due to instability of various synthons toward the reagents used during synthesis of oligonucleotides. Therefore, using a complementary approach, we developed phosphoramidite synthons that can be incorporated into oligonucleotides with minimum changes in the solid phase DNA synthesis protocols but contain a handle for generating appropriate analogues postsynthetically.This Account summarizes our efforts toward preparing these types of analogues over the past three decades and discusses synthesis and properties of backbone modified oligonucleotides that originated from the Caruthers' laboratory. For example, by replacing one of the internucleotide oxygens with an acetate group, we obtained so-called phosphonoacetate oligonucleotides that were stable to nucleases and, when delivered as esters, entered into cells unaided. Alternatively oligonucleotides bearing borane phosphonate linkages were found to be RNase H active and compatible with the endogenous RNA induced silencing complex (RISC). Oligonucleotides containing an alkyne group directly linked to phosphorus in the backbone were prepared as well and used to attach molecules such as amino acids and peptides.
Collapse
Affiliation(s)
- Pawan Kumar
- Broad Institute of Harvard and MIT, Stanley Center for Psychiatric Research, 450 Main Street, Cambridge, Massachusetts 02142, United States
| | - Marvin H. Caruthers
- Department of Biochemistry, University of Colorado, Boulder, Colorado 80303, United States
| |
Collapse
|
394
|
Wang Q, Liu X, Zhao J, Zhu R. Circular RNAs: novel diagnostic and therapeutic targets for ischemic stroke. Expert Rev Mol Diagn 2020; 20:1039-1049. [PMID: 32954841 DOI: 10.1080/14737159.2020.1826313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Qianwen Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xu Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jingjing Zhao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
395
|
Dammes N, Peer D. Paving the Road for RNA Therapeutics. Trends Pharmacol Sci 2020; 41:755-775. [PMID: 32893005 PMCID: PMC7470715 DOI: 10.1016/j.tips.2020.08.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022]
Abstract
Therapeutic RNA molecules possess high potential for treating medical conditions if they can successfully reach the target cell upon administration. However, unmodified RNA molecules are rapidly degraded and cleared from the circulation. In addition, their large size and negative charge complicates their passing through the cell membrane. The difficulty of RNA therapy, therefore, lies in the efficient intracellular delivery of intact RNA molecules to the tissue of interest without inducing adverse effects. Here, we outline the recent developments in therapeutic RNA delivery and discuss the wide potential in manipulating the function of cells with RNAs. The focus is not only on the variety of delivery strategies but also on the versatile nature of RNA and its wide applicability. This wide applicability is especially interesting when considering the modular nature of nucleic acids. An optimal delivery vehicle, therefore, can facilitate numerous clinical applications of RNA.
Collapse
Affiliation(s)
- Niels Dammes
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel,School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel,Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel,Center for Nanoscience and Nanotechnology, and Tel Aviv University, Tel Aviv 69978, Israel,Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, and Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
396
|
Juliano RL. Addressing cancer signal transduction pathways with antisense and siRNA oligonucleotides. NAR Cancer 2020; 2:zcaa025. [PMID: 33015625 PMCID: PMC7520847 DOI: 10.1093/narcan/zcaa025] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/24/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Signal transduction pathways play key roles in the initiation, progression and dissemination of cancer. Thus, signaling molecules are attractive targets for cancer therapeutics and enormous efforts have gone into the development of small molecule inhibitors of these pathways. However, regrettably, there has been only moderate progress to date, primarily in connection with the RAS signaling pathway. Oligonucleotide-based drugs potentially offer several advantages for addressing signaling pathways, including their exquisite selectivity and their ability to exploit both enzymatic and nonenzymatic targets. Nonetheless, there are problems inherent in the oligonucleotide approach, not the least being the challenge of effectively delivering these complex molecules to intracellular sites within tumors. This survey article will provide a selective review of recent studies where oligonucleotides were used to address cancer signaling and will discuss both positive aspects and limitations of those studies. This will be set in the context of an overview of various cancer signaling pathways and small molecule approaches to regulate those pathways. The survey will also evaluate the challenges and opportunities implicit in the oligonucleotide-based approach to cancer signaling and will point out several possibilities for future research.
Collapse
|
397
|
Chen S, Thorne RF, Zhang XD, Wu M, Liu L. Non-coding RNAs, guardians of the p53 galaxy. Semin Cancer Biol 2020; 75:72-83. [PMID: 32927018 DOI: 10.1016/j.semcancer.2020.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022]
Abstract
The TP53 gene is arguably the most important tumor suppressor gene known, contributing multifaceted roles to the process of tumor development. Its protein product p53, is a crucial sequence-specific transcription factor which regulates the expression of a large network of protein-coding genes, as well as thousands of noncoding RNAs (ncRNAs), notably microRNAs and long ncRNAs (lncRNAs). Through a variety of direct and indirect mechanisms, ncRNAs in turn modulate p53 levels and activity. Here the numbers of studies are steadily building which link the contributions of dysregulated ncRNAs to tumorigenesis via their participation throughout the p53 regulatory network. In this review, we will examine how the principal forms of ncRNAs, namely microRNAs, lncRNAs and circular RNAs (circRNAs) function as either effectors or regulators amongst the diversity of p53's cellular responses. We first discuss the more recently discovered connections between miRNAs and p53 signaling before focusing on the remarkable diversity of crosstalk evident between lncRNAs and p53, and subsequently, developing reports linking circRNAs to p53. Highlighted throughout the review are the mechanistic impacts of dysregulated ncRNAs on p53 functions as well as the possible prognostic implications of these interactions. We also describe the emerging connections between ncRNAs and the often-perplexing functions of mutant p53. Finally, in the context of p53 therapeutic approaches, we describe some of the challenges in ncRNA research and their potential for translation.
Collapse
Affiliation(s)
- Song Chen
- Translational Research Institute, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China; Molecular Pathology Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, Jiangsu, 223300, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China; Molecular Pathology Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Environmental and Life Sciences, the University of Newcastle, NSW, 2258, Australia
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China; Molecular Pathology Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Biomedical Sciences and Pharmacy, the University of Newcastle, NSW, 2308, Australia
| | - Mian Wu
- Translational Research Institute, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, Zhengzhou, 450003, China; Molecular Pathology Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230027, China.
| | - Lianxin Liu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
398
|
Weingärtner A, Bethge L, Weiss L, Sternberger M, Lindholm MW. Less Is More: Novel Hepatocyte-Targeted siRNA Conjugates for Treatment of Liver-Related Disorders. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:242-250. [PMID: 32590173 PMCID: PMC7321783 DOI: 10.1016/j.omtn.2020.05.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/30/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
N-acetyl-galactosamine (GalNAc) conjugation enhances liver specificity for therapeutic oligonucleotides. Here we report on a novel design with improved activity and stability compared with a triantennary design. We applied a versatile monovalent serinol-GalNAc conjugation strategy. First, 1-4 serial serinol-linked GalNAc units were conjugated to terminal positions of small interfering RNA (siRNA) molecules. In primary hepatocytes, 5' antisense GalNAc conjugates were inactive, whereas 3' antisense and 3' or 5' sense conjugates displayed low activity for single GalNAc units, while 2-4 serial GalNAc conjugates were all equally potent. In mice, 5' sense conjugates with 2-4 serial GalNAc units were all as potent as a triantennary GalNAc control (1 mg/kg). Second, increased spacing between two serial 5' sense-conjugated GalNAc units did not affect in vitro activity. Finally, two single GalNAc units were positioned at opposite ends of the sense strand. A single dose (0.3 mg/kg) of this novel conjugate in mice showed a 3-fold reduction of serum target protein level at day 7 and 4-fold lower serum level at day 27, relative to an equimolar dose of a triantennary GalNAc conjugate of the same siRNA. Improved tritosome stability (by liquid chromatography-mass spectrometry [LC-MS] analysis) can at least partially explain the increased activity and duration of action for the novel GalNAc conjugate.
Collapse
Affiliation(s)
- Adrien Weingärtner
- Silence Therapeutics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| | - Lucas Bethge
- Silence Therapeutics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Lisa Weiss
- Silence Therapeutics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Maria Sternberger
- Silence Therapeutics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | | |
Collapse
|
399
|
Our emerging understanding of the roles of long non-coding RNAs in normal liver function, disease, and malignancy. JHEP Rep 2020; 3:100177. [PMID: 33294829 PMCID: PMC7689550 DOI: 10.1016/j.jhepr.2020.100177] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/06/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are important biological mediators that regulate numerous cellular processes. New experimental evidence suggests that lncRNAs play essential roles in liver development, normal liver physiology, fibrosis, and malignancy, including hepatocellular carcinoma and cholangiocarcinoma. In this review, we summarise our current understanding of the function of lncRNAs in the liver in both health and disease, as well as discuss approaches that could be used to target these non-coding transcripts for therapeutic purposes.
Collapse
Key Words
- ABCA1, ATP-binding cassette transporter A1
- ACTA2/ɑ-SMA, α-smooth muscle actin
- APO, apolipoprotein
- ASO, antisense oligonucleotides
- BDL, bile duct ligation
- CCA, cholangiocarcinoma
- CCl4, carbon tetrachloride
- COL1A1, collagen type I α 1
- CYP, cytochrome P450
- Cholangiocarcinoma
- DANCR, differentiation antagonising non-protein coding RNA
- DE, definitive endoderm
- DEANR1, definitive endoderm-associated lncRNA1
- DIGIT, divergent to goosecoid, induced by TGF-β family signalling
- DILC, downregulated in liver cancer stem cells
- EST, expression sequence tag
- EpCAM, epithelial cell adhesion molecule
- FBP1, fructose-bisphosphatase 1
- FENDRR, foetal-lethal non-coding developmental regulatory RNA
- FXR, farnesoid X receptor
- GAS5, growth arrest-specific transcript 5
- H3K18ac, histone 3 lysine 18 acetylation
- H3K36me3, histone 3 lysine 36 trimethylation
- H3K4me3, histone 3 lysine 4 trimethylation
- HCC, hepatocellular carcinoma
- HEIH, high expression In HCC
- HNRNPA1, heterogenous nuclear protein ribonucleoprotein A1
- HOTAIR, HOX transcript antisense RNA
- HOTTIP, HOXA transcript at the distal tip
- HSC, hepatic stellate cells
- HULC, highly upregulated in liver cancer
- Hepatocellular carcinoma
- HuR, human antigen R
- LCSC, liver cancer stem cell
- LSD1, lysine-specific demethylase 1
- LXR, liver X receptors
- LeXis, liver-expressed LXR-induced sequence
- Liver cancer
- Liver fibrosis
- Liver metabolism
- Liver-specific lncRNAs
- LncLSTR, lncRNA liver-specific triglyceride regulator
- MALAT1, metastasis-associated lung adenocarcinoma transcript 1
- MEG3, maternally expressed gene 3
- NAT, natural antisense transcript
- NEAT1, nuclear enriched abundant transcript 1
- ORF, open reading frame
- PKM2, pyruvate kinase muscle isozyme M2
- PPAR-α, peroxisome proliferator-activated receptor-α
- PRC, polycomb repressive complex
- RACE, rapid amplification of cDNA ends
- RNA Pol, RNA polymerase
- S6K1, S6 kinase 1
- SHP, small heterodimer partner
- SREBPs, steroid response binding proteins
- SREs, sterol response elements
- TGF-β, transforming growth factor-β
- TTR, transthyretin
- XIST, X-inactive specific transcript
- ZEB1, zinc finger E-box-binding homeobox 1
- ceRNA, competing endogenous RNA
- eRNA, enhancer RNAs
- lincRNA, long intervening non-coding RNA
- lncRNA
- lncRNA, long non-coding RNA
- mTOR, mammalian target of rapamycin
- siRNA, small interfering RNA
Collapse
|
400
|
Duffy K, Arangundy-Franklin S, Holliger P. Modified nucleic acids: replication, evolution, and next-generation therapeutics. BMC Biol 2020; 18:112. [PMID: 32878624 PMCID: PMC7469316 DOI: 10.1186/s12915-020-00803-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Modified nucleic acids, also called xeno nucleic acids (XNAs), offer a variety of advantages for biotechnological applications and address some of the limitations of first-generation nucleic acid therapeutics. Indeed, several therapeutics based on modified nucleic acids have recently been approved and many more are under clinical evaluation. XNAs can provide increased biostability and furthermore are now increasingly amenable to in vitro evolution, accelerating lead discovery. Here, we review the most recent discoveries in this dynamic field with a focus on progress in the enzymatic replication and functional exploration of XNAs.
Collapse
Affiliation(s)
- Karen Duffy
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | | | - Philipp Holliger
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|