1
|
Filtz A, Parihar S, Greenberg GS, Park CM, Scotti A, Lorenzatti D, Badimon JJ, Soffer DE, Toth PP, Lavie CJ, Bittner V, Virani SS, Slipczuk L. New approaches to triglyceride reduction: Is there any hope left? Am J Prev Cardiol 2024; 18:100648. [PMID: 38584606 PMCID: PMC10998004 DOI: 10.1016/j.ajpc.2024.100648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/29/2024] [Accepted: 03/09/2024] [Indexed: 04/09/2024] Open
Abstract
Triglycerides play a crucial role in the efficient storage of energy in the body. Mild and moderate hypertriglyceridemia (HTG) is a heterogeneous disorder with significant association with atherosclerotic cardiovascular disease (ASCVD), including myocardial infarction, ischemic stroke, and peripheral artery disease and represents an important component of the residual ASCVD risk in statin treated patients despite optimal low-density lipoprotein cholesterol reduction. Individuals with severe HTG (>1,000 mg/dL) rarely develop atherosclerosis but have an incremental incidence of acute pancreatitis with significant morbidity and mortality. HTG can occur from a combination of genetic (both mono and polygenic) and environmental factors including poor diet, low physical activity, obesity, medications, and diseases like insulin resistance and other endocrine pathologies. HTG represents a potential target for ASCVD risk and pancreatitis risk reduction, however data on ASCVD reduction by treating HTG is still lacking and HTG-associated acute pancreatitis occurs too rarely to effectively demonstrate treatment benefit. In this review, we address the key aspects of HTG pathophysiology and examine the mechanisms and background of current and emerging therapies in the management of HTG.
Collapse
Affiliation(s)
- Annalisa Filtz
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Siddhant Parihar
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Garred S Greenberg
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Christine M Park
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrea Scotti
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel Lorenzatti
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Juan J Badimon
- Cardiology Department, Hospital General Jaen, Jaen, Spain
- Atherothrombosis Research Unit, Mount Sinai School of Medicine, New York, New York, USA
| | - Daniel E Soffer
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter P Toth
- CGH Medical Center, Sterling, Illinois
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-the UQ School of Medicine, New Orleans, Louisiana, USA
| | - Vera Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Salim S Virani
- Section of Cardiology, Department of Medicine, The Aga Khan University, Karachi, Pakistan
- Section of Cardiology, Texas Heart Institute & Baylor College of Medicine, Houston, TX, USA
| | - Leandro Slipczuk
- Cardiology Division, Montefiore-Einstein Center for Heart and Vascular Care, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Yeoh YQ, Amin A, Cuic B, Tomas D, Turner BJ, Shabanpoor F. Efficient systemic CNS delivery of a therapeutic antisense oligonucleotide with a blood-brain barrier-penetrating ApoE-derived peptide. Biomed Pharmacother 2024; 175:116737. [PMID: 38749176 DOI: 10.1016/j.biopha.2024.116737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 06/03/2024] Open
Abstract
Antisense oligonucleotide (ASO) has emerged as a promising therapeutic approach for treating central nervous system (CNS) disorders by modulating gene expression with high selectivity and specificity. However, the poor permeability of ASO across the blood-brain barrier (BBB) diminishes its therapeutic success. Here, we designed and synthesized a series of BBB-penetrating peptides (BPP) derived from either the receptor-binding domain of apolipoprotein E (ApoE) or a transferrin receptor-binding peptide (THR). The BPPs were conjugated to phosphorodiamidate morpholino oligomers (PMO) that are chemically analogous to the 2'-O-(2-methoxyethyl) (MOE)-modified ASO approved by the FDA for treating spinal muscular atrophy (SMA). The BPP-PMO conjugates significantly increased the level of full-length SMN2 in the patient-derived SMA fibroblasts in a concentration-dependent manner with minimal to no toxicity. Furthermore, the systemic administration of the most potent BPP-PMO conjugates significantly increased the expression of full-length SMN2 in the brain and spinal cord of SMN2 transgenic adult mice. Notably, BPP8-PMO conjugate showed a 1.25-fold increase in the expression of full-length functional SMN2 in the brain. Fluorescence imaging studies confirmed that 78% of the fluorescently (Cy7)-labelled BPP8-PMO reached brain parenchyma, with 11% uptake in neuronal cells. Additionally, the BPP-PMO conjugates containing retro-inverso (RI) D-BPPs were found to possess extended half-lives compared to their L-counterparts, indicating increased stability against protease degradation while preserving the bioactivity. This delivery platform based on BPP enhances the CNS bioavailability of PMO targeting the SMN2 gene, paving the way for the development of systemically administered neurotherapeutics for CNS disorders.
Collapse
Affiliation(s)
- Yuan Qi Yeoh
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Azin Amin
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Brittany Cuic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Doris Tomas
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Fazel Shabanpoor
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia; School of Chemistry, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
3
|
Milman S, Barzilai N. Discovering Biological Mechanisms of Exceptional Human Health Span and Life Span. Cold Spring Harb Perspect Med 2023; 13:a041204. [PMID: 37137499 PMCID: PMC10513160 DOI: 10.1101/cshperspect.a041204] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Humans age at different rates and families with exceptional longevity provide an opportunity to understand why some people age slower than others. Unique features exhibited by centenarians include a family history of extended life span, compression of morbidity with resultant extension of health span, and longevity-associated biomarker profiles. These biomarkers, including low-circulating insulin-like growth factor 1 (IGF-1) and elevated high-density lipoprotein (HDL) cholesterol levels, are associated with functional genotypes that are enriched in centenarians, suggesting that they may be causative for longevity. While not all genetic discoveries from centenarians have been validated, in part due to exceptional life span being a rare phenotype in the general population, the APOE2 and FOXO3a genotypes have been confirmed in a number of populations with exceptional longevity. However, life span is now recognized as a complex trait and genetic research methods to study longevity are rapidly extending beyond classical Mendelian genetics to polygenic inheritance methodologies. Moreover, newer approaches are suggesting that pathways that have been recognized for decades to control life span in animals may also regulate life span in humans. These discoveries led to strategic development of therapeutics that may delay aging and prolong health span.
Collapse
Affiliation(s)
- Sofiya Milman
- Institute for Aging Research, Department of Medicine, Divisions of Endocrinology and Geriatrics, Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Nir Barzilai
- Institute for Aging Research, Department of Medicine, Divisions of Endocrinology and Geriatrics, Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
4
|
Friedman L, Avitzur OB, Galai EO, Ferrari N, Choen A, Dahan S, Mordechai T, Hart G. The safety and toxicity profile of SPL84, an inhaled antisense oligonucleotide for treatment of cystic fibrosis patients with the 3849 +10kb C->T mutation, supports a Phase 1/2 clinical study. Expert Opin Drug Metab Toxicol 2023; 19:709-720. [PMID: 37799089 DOI: 10.1080/17425255.2023.2266361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION SPL84 is an inhaled antisense oligonucleotide (ASO) in development for the treatment of cystic fibrosis (CF) patients carrying the 3849 + 10kb C->T (3849) mutation. To support the initiation of the first clinical study, a full battery of safety and toxicology studies were performed. RESEARCH DESIGN AND METHODS SPL84 was administered by inhalation to mice and monkeys to determine the no observed adverse effect level (NOAEL) and establish sufficient safety margins for the starting clinical dose. RESULTS There were no preclinical safety findings with SPL84; no related clinical signs, nor any effect on body weight, food consumption, or clinical pathology. The microscopic changes in the lungs were regarded as non-adverse and reflected a normal clearance process for inhaled compounds. Systemic exposure in both species was low. The NOAEL for mice and monkeys was the highest administered dose in both species, resulting in safety margins ~ 40X the proposed starting clinical dose. CONCLUSION These successful results supported the initiation of a phase 1/2 clinical study of SPL84 (ongoing), assessing the safety, tolerability, and pharmacokinetics of a single ascending dose in healthy subjects to be followed by assessment of safety, tolerability, pharmacokinetics, and preliminary efficacy of multiple ascending doses in CF patients carrying the 3849 mutation.
Collapse
Affiliation(s)
- Lital Friedman
- SpliSense, BiohouseLabs, Haddasah Ein Kerem, Jerusalem, Israel
| | | | | | | | - Asa Choen
- SpliSense, BiohouseLabs, Haddasah Ein Kerem, Jerusalem, Israel
| | - Sara Dahan
- SpliSense, BiohouseLabs, Haddasah Ein Kerem, Jerusalem, Israel
| | - Tamar Mordechai
- SpliSense, BiohouseLabs, Haddasah Ein Kerem, Jerusalem, Israel
| | - Gili Hart
- SpliSense, BiohouseLabs, Haddasah Ein Kerem, Jerusalem, Israel
| |
Collapse
|
5
|
Chen R, Lin S, Chen X. The promising novel therapies for familial hypercholesterolemia. J Clin Lab Anal 2022; 36:e24552. [PMID: 35712827 PMCID: PMC9279988 DOI: 10.1002/jcla.24552] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Background The incidence of premature atherosclerotic cardiovascular disease in familial hypercholesterolemia (FH) is high. In recent years, novel therapeutic modalities have shown significant lipid‐lowering ability. In this paper, we summarize the recent developments in novel therapies for FH via the treatment of different targets and discuss the characteristics of each targeted therapy. Based on the process of protein synthesis, we attempt to summarize the direct‐effect targets including protein, RNA, and DNA. Methods For this systematic review, relevant studies are assessed by searching in several databases including PubMed, Web of Science, Scopus, and Google Scholar. The publications of original researches are considered for screening. Results Most drugs are protein‐targeted such as molecule‐based and monoclonal antibodies, including statins, ezetimibe, alirocumab, evolocumab, and evinacumab. Both antisense oligonucleotide (ASO) and small interfering RNA (siRNA) approaches, such as mipomersen, vupanorsen, inclisiran, and ARO‐ANG3, are designed to reduce the number of mRNA transcripts and then degrade proteins. DNA‐targeted therapies such as adeno‐associated virus or CRISPR–Cas9 modification could be used to deliver or edit genes to address a genetic deficiency and improve the related phenotype. Conclusion While the therapies based on different targets including protein, RNA, and DNA are on different stages of development, the mechanisms of these novel therapies may provide new ideas for precision medicine.
Collapse
Affiliation(s)
- Ruoyu Chen
- School of Medicine of Ningbo University, Ningbo, China
| | - Shaoyi Lin
- The Affiliated Ningbo First Hospital, School of Medicine of Ningbo University, Ningbo, China
| | - Xiaomin Chen
- The Affiliated Ningbo First Hospital, School of Medicine of Ningbo University, Ningbo, China.,Ningbo First Hospital Affiliated to School of Medicine of Zhejiang University, Ningbo, China
| |
Collapse
|
6
|
Zhang Y, Lai L, Liu Y, Chen B, Yao J, Zheng P, Pan Q, Zhu W. Biomineralized Cascade Enzyme-Encapsulated ZIF-8 Nanoparticles Combined with Antisense Oligonucleotides for Drug-Resistant Bacteria Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6453-6464. [PMID: 35094518 DOI: 10.1021/acsami.1c23808] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The unrestrained use of antibiotics accelerates the development of drug-resistant bacteria and leads to an increasing threat to human health. Therefore, there is an urgent need to explore novel and effective strategies for the treatment of bacterial infections. Herein, zeolite imidazole framework-8 (ZIF-8) material was utilized to construct biomineralized nanomaterial (GOx&HRP@ZIF-8/ASO) by encapsulating biological cascade enzymes and combining with antisense oligonucleotides (ASOs), which achieved effective and synergistic antidrug-resistant bacteria therapy. Various in vitro assays confirmed that GOx&HRP@ZIF-8/ASO exhibited excellent antibacterial properties against Escherichia coli, Staphylococcus aureus, methicillin-resistant S. aureus (MRSA) during catalysis of glucose (Glu), especially the minimum inhibitory concentration (MIC) against MRSA was only 16 μg/mL. Compared with simple ZIF-8 (32.85%) and ftsZ ASO (58.65%), GOx&HRP@ZIF-8/ASO+Glu exhibited superb biofilm destruction ability, and the bacteria removal efficiency of the MRSA biofilm could be as high as 88.2%, indicating that the reactive oxygen species (ROS) produced by the cascade enzyme reaction imparted the main synergistic antibacterial capability, and simultaneously, ftsZ ASO significantly enhanced the antibacterial effect by inhibiting the expression of the ftsZ gene. In vivo anti-infection treatment experiments revealed that GOx&HRP@ZIF-8/ASO exhibited the best wound repairing performance and excellent biocompatibility in the presence of Glu. These findings suggested that GOx&HRP@ZIF-8/ASO has favorably realized high-efficiency treatment of MRSA infection and filled the gap in the antibacterial application of biological enzymes.
Collapse
Affiliation(s)
- Yan Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Luogen Lai
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Yijun Liu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Beini Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Jing Yao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Qingshan Pan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, Jiangxi, China
| |
Collapse
|
7
|
Noh S, Mai K, Shaver M, Yong S, Mostaghimi M, Oh G, Radwan MM. Emerging Cholesterol Modulators for Atherosclerotic Cardiovascular Disease. Am J Med Sci 2022; 363:373-387. [DOI: 10.1016/j.amjms.2021.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/07/2021] [Accepted: 12/07/2021] [Indexed: 12/01/2022]
|
8
|
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: 347] [Impact Index Per Article: 86.8] [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
|
9
|
He L, Chang H, Qi Y, Zhang B, Shao Q. ceRNA Networks: The Backbone Role in Neoadjuvant Chemoradiotherapy Resistance/Sensitivity of Locally Advanced Rectal Cancer. Technol Cancer Res Treat 2021; 20:15330338211062313. [PMID: 34908512 PMCID: PMC8689620 DOI: 10.1177/15330338211062313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/21/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022] Open
Abstract
Approximately 40% of rectal cancers during initial diagnosis are identified as locally advanced rectal cancers (LARCs), for which the standardized treatment scenario is total mesorectal excision following neoadjuvant chemoradiotherapy (nCRT). nCRT can lead to discernible reductions in local relapse rate and distant metastasis rate in LARC patients, in whom previously inoperable tumors may potentially be surgically removed. However, only 4% to 20% cases can attain pathological complete response, and the remaining patients who are unresponsive to nCRT have to suffer from the side effects plus toxicities and may encounter poor survival outcomes due to the late surgical intervention. As such, employing potential biomarkers to differentiate responders from nonresponders before nCRT implementation appears to be the overarching goal. Well-defined competing endogenous RNA (ceRNA) networks include long noncoding RNA (lncRNA)-microRNA (miRNA)-mRNA and circRNA-miRNA-mRNA networks. As ceRNAs, lncRNAs, and circRNAs sponge miRNAs to indirectly suppress miRNAs downstream of oncogenic mRNAs or tumor-suppressive mRNAs. The abnormal expression of mRNAs regulates the nCRT-induced DNA damage repair process through pluralistic carcinogenic signaling pathways, thereby bringing about alterations in the nCRT resistance/sensitivity of tumors. Moreover, many molecular mechanisms relevant to cell proliferation, metastasis, or apoptosis of cancers (eg, epithelial-mesenchymal transition and caspase-9-caspase-3 pathway) are influenced by ceRNA networks. Herein, we reviewed a large group of abnormally expressed mRNAs and noncoding RNAs that are associated with nCRT resistance/sensitivity in LARC patients and ultimately pinpointed the backbone role of ceRNA networks in the molecular mechanisms of nCRT resistance/sensitivity.
Collapse
Affiliation(s)
- Lin He
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Hao Chang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| | - Yuhong Qi
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| | - Bing Zhang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| | - Qiuju Shao
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University, Xi’an, Shaanxi Province, China
| |
Collapse
|
10
|
Abstract
Diabetes mellitus is a major risk factor for coronary heart disease (CHD). The major form of diabetes mellitus is type 2 diabetes mellitus (T2D), which is thus largely responsible for the CHD association in the general population. Recent years have seen major advances in the genetics of T2D, principally through ever-increasing large-scale genome-wide association studies. This article addresses the question of whether this expanding knowledge of the genomics of T2D provides insight into the etiologic relationship between T2D and CHD. We will investigate this relationship by reviewing the evidence for shared genetic loci between T2D and CHD; by examining the formal testing of this interaction (Mendelian randomization studies assessing whether T2D is causal for CHD); and then turn to the implications of this genetic relationship for therapies for CHD, for therapies for T2D, and for therapies that affect both. In conclusion, the growing knowledge of the genetic relationship between T2D and CHD is beginning to provide the promise for improved prevention and treatment of both disorders.
Collapse
Affiliation(s)
- Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
11
|
Zhou X, Wang S, Zhu Y, Pan Y, Zhang L, Yang Z. Overcoming the delivery barrier of oligonucleotide drugs and enhancing nucleoside drug efficiency: The use of nucleolipids. Med Res Rev 2019; 40:1178-1199. [PMID: 31820472 DOI: 10.1002/med.21652] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
With the rapid development of synthetic technology and biological technology, many nucleic acid-based drugs have entered the clinical trials. However, their inherent disabilities in actively and efficiently penetrating cell membranes still severely restrict their further application. The main drawback of cationic lipids, which have been widely used as nonviral vectors of nucleic acids, is their high cytotoxicity. A series of nucleoside-based or nucleotide-based nucleolipids have been reported in recent years, due to their oligonucleotide delivery capacity and low toxicity in comparison with cationic lipids. Lipophilic prodrugs of nucleoside analogs have extremely similar structures with nucleolipid vectors and are thus helpful for improving the transmembrane ability. This review introduces the progress of nucleolipids and provides new strategies for improving the delivery efficiency of nucleic acid-based drugs, as well as lipophilic prodrugs of nucleosides or nucleotides for antiviral or anticancer therapies.
Collapse
Affiliation(s)
- Xinyang Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Haidian, China
| | - Shuhe Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Haidian, China
| | - Yuejie Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Haidian, China
| | - Yufei Pan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Haidian, China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Haidian, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Haidian, China
| |
Collapse
|
12
|
Pirie E, Cauntay P, Fu W, Ray S, Pan C, Lusis AJ, Hsiao J, Burel SA, Narayanan P, Crooke RM, Lee RG. Hybrid Mouse Diversity Panel Identifies Genetic Architecture Associated with the Acute Antisense Oligonucleotide-Mediated Inflammatory Response to a 2'- O-Methoxyethyl Antisense Oligonucleotide. Nucleic Acid Ther 2019; 29:266-277. [PMID: 31368839 PMCID: PMC6765210 DOI: 10.1089/nat.2019.0797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/04/2019] [Indexed: 01/04/2023] Open
Abstract
Although antisense oligonucleotides (ASOs) are well tolerated preclinically and in the clinic, some sequences of ASOs can trigger an inflammatory response leading to B cell and macrophage activation in rodents. This prompted our investigation into the contribution of genetic architecture to the ASO-mediated inflammatory response. Genome-wide association (GWA) and transcriptomic analysis in a hybrid mouse diversity panel (HMDP) were used to identify and validate novel genes involved in the acute and delayed inflammatory response to a single 75 mg/kg dose of an inflammatory 2'-O-methoxyethyl (2'MOE) modified ASO. The acute response was measured 6 h after ASO administration, via evaluation for increased plasma production of interleukin 6 (IL6), IL10, monocyte chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein-1β (MIP-1β). Delayed inflammation was evaluated by spleen weight increases after 96 h. We identified single nucleotide polymorphisms (SNPs) on chromosomes 16 and 17 associated with plasma MIP-1β, IL6, and MCP-1 levels, and one on chromosome 8 associated with increases in spleen weight. Systems genetic analysis utilizing transcriptomic data from HMDP strain macrophages determined that the acute inflammatory SNPs were expression quantitative trait locis (eQTLs) for CCAAT/enhancer-binding protein beta (Cebpb) and salt inducible kinase 1 (Sik1). The delayed inflammatory SNP was an eQTL for Rho guanine nucleotide exchange factor 10 (Arhgef10). In vitro assays in mouse primary cells and human cell lines have confirmed the HMDP finding that lower Sik1 expression increases the acute inflammatory response. Our results demonstrate the utility of using mouse GWA study (GWAS) and the HMDP for detecting genes modulating the inflammatory response to pro-inflammatory ASOs in a pharmacological setting.
Collapse
Affiliation(s)
- Elaine Pirie
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California
| | - Patrick Cauntay
- Preclinical Development, Ionis Pharmaceuticals, Carlsbad, California
| | - Wuxia Fu
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California
| | - Shayoni Ray
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California
| | - Calvin Pan
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
| | - Aldonis J. Lusis
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
| | - Jill Hsiao
- Preclinical Development, Ionis Pharmaceuticals, Carlsbad, California
| | | | - Padma Narayanan
- Preclinical Development, Ionis Pharmaceuticals, Carlsbad, California
| | - Rosanne M. Crooke
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California
| | - Richard G. Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW High lipoprotein(a) levels are observationally and causally, from human genetics, associated with increased risk of cardiovascular disease including myocardial infarction and aortic valve stenosis. The European Atherosclerosis Society recommends screening for elevated lipoprotein(a) levels in high-risk patients. Different therapies have been suggested and some are used to treat elevated lipoprotein(a) levels such as niacin, PCSK9 inhibitors, and CETP inhibitors; however, to date, no randomized controlled trial has demonstrated that lowering of lipoprotein(a) leads to lower risk of cardiovascular disease. RECENT FINDINGS Synthetic oligonucleotides can be used to inactivate genes involved in disease processes. To lower lipoprotein(a), two antisense oligonucleotides have been developed, one targeting apolipoprotein B and one targeting apolipoprotein(a). Mipomersen is an antisense oligonucleotide targeting apolipoprotein B and thereby reducing levels of all apolipoprotein B containing lipoproteins in the circulation. Mipomersen has been shown to lower lipoprotein(a) by 20-50% in phase 3 studies. AKCEA-APO(a)-LRx is the most recent antisense oligonucleotide targeting apolipoprotein(a) and thereby uniquely targeting lipoprotein(a). It has been tested in a phase 2 study and has shown to lower lipoprotein(a) levels by 50-80%. The treatment of elevated lipoprotein(a) levels with the newest antisense oligonucleotides seems promising; however, no improvement in cardiovascular disease risk has yet been shown. However, a phase 3 study of AKCEA-APO(a)-LRx is being planned with cardiovascular disease as outcome, and results are awaited with great anticipation.
Collapse
|
14
|
Vahdat Lasemi F, Mahjoubin Tehran M, Aghaee-Bakhtiari SH, Jalili A, Jaafari MR, Sahebkar A. Harnessing nucleic acid-based therapeutics for atherosclerotic cardiovascular disease: state of the art. Drug Discov Today 2019; 24:1116-1131. [PMID: 30980904 DOI: 10.1016/j.drudis.2019.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/16/2019] [Accepted: 04/05/2019] [Indexed: 12/19/2022]
Abstract
Dyslipidemia is one of the major but modifiable risk factors for atherosclerotic cardiovascular disease (ACVD). Despite the accessibility of statins and other lipid-lowering drugs, the burden of ACVD is still high globally, highlighting the need for new therapeutic approaches. Nucleic acid-based technologies, including antisense oligonucleotides (ASOs), small interfering (si)RNAs, miRNAs, and decoys, are emerging therapeutic modalities for the treatment of ACVD. These technologies aim to degrade gene mRNA transcripts to decrease the levels of atherogenic lipoproteins. Using gene-silencing approaches, the levels of atherogenic lipoproteins can be decreased by targeting proteins that have key roles in lipoprotein metabolism. Here, we highlight preclinical and clinical findings using these approaches for the development of novel therapies against ACVD.
Collapse
Affiliation(s)
- Fatemeh Vahdat Lasemi
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahjoubin Tehran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Jalili
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Pirie E, Ray S, Pan C, Fu W, Powers AF, Polikoff D, Miller CM, Kudrna KM, Harris EN, Lusis AJ, Crooke RM, Lee RG. Mouse genome-wide association studies and systems genetics uncover the genetic architecture associated with hepatic pharmacokinetic and pharmacodynamic properties of a constrained ethyl antisense oligonucleotide targeting Malat1. PLoS Genet 2018; 14:e1007732. [PMID: 30372444 PMCID: PMC6224167 DOI: 10.1371/journal.pgen.1007732] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 11/08/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022] Open
Abstract
Antisense oligonucleotides (ASOs) have demonstrated variation of efficacy in patient populations. This has prompted our investigation into the contribution of genetic architecture to ASO pharmacokinetics (PK) and pharmacodynamics (PD). Genome wide association (GWA) and transcriptomic analysis in a hybrid mouse diversity panel (HMDP) were used to identify and validate novel genes involved in the uptake and efficacy of a single dose of a Malat1 constrained ethyl (cEt) modified ASO. The GWA of the HMDP identified two significant associations on chromosomes 4 and 10 with hepatic Malat1 ASO concentrations. Stabilin 2 (Stab2) and vesicle associated membrane protein 3 (Vamp3) were identified by cis-eQTL analysis. HMDP strains with lower Stab2 expression and Stab2 KO mice displayed significantly lower PK than strains with higher Stab2 expression and the wild type (WT) animals respectively, confirming the role of Stab2 in regulating hepatic Malat1 ASO uptake. GWA examining ASO efficacy uncovered three loci associated with Malat1 potency: Small Subunit Processome Component (Utp11l) on chromosome 4, Rho associated coiled-coil containing protein kinase 2 (Rock2) and Aci-reductone dioxygenase (Adi1) on chromosome 12. Our results demonstrate the utility of mouse GWAS using the HMDP in detecting genes capable of impacting the uptake of ASOs, and identifies genes critical for the activity of ASOs in vivo.
Collapse
Affiliation(s)
- Elaine Pirie
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Shayoni Ray
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Calvin Pan
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Wuxia Fu
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Andrew F. Powers
- Exploratory Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Danielle Polikoff
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Colton M. Miller
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Katrina M. Kudrna
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Edward N. Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Aldons J. Lusis
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Rosanne M. Crooke
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Richard G. Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| |
Collapse
|
16
|
Weng SY, Wang X, Vijayan S, Tang Y, Kim YO, Padberg K, Regen T, Molokanova O, Chen T, Bopp T, Schild H, Brombacher F, Crosby JR, McCaleb ML, Waisman A, Bockamp E, Schuppan D. IL-4 Receptor Alpha Signaling through Macrophages Differentially Regulates Liver Fibrosis Progression and Reversal. EBioMedicine 2018; 29:92-103. [PMID: 29463471 PMCID: PMC5925448 DOI: 10.1016/j.ebiom.2018.01.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic hepatitis leads to liver fibrosis and cirrhosis. Cirrhosis is a major cause of worldwide morbidity and mortality. Macrophages play a key role in fibrosis progression and reversal. However, the signals that determine fibrogenic vs fibrolytic macrophage function remain ill defined. We studied the role of interleukin-4 receptor α (IL-4Rα), a potential central switch of macrophage polarization, in liver fibrosis progression and reversal. We demonstrate that inflammatory monocyte infiltration and liver fibrogenesis were suppressed in general IL-4Rα−/− as well as in macrophage-specific IL-4Rα−/− (IL-4RαΔLysM) mice. However, with deletion of IL-4RαΔLysM spontaneous fibrosis reversal was retarded. Results were replicated by pharmacological intervention using IL-4Rα-specific antisense oligonucleotides. Retarded resolution was linked to the loss of M2-type resident macrophages, which secreted MMP-12 through IL-4 and IL-13-mediated phospho-STAT6 activation. We conclude that IL-4Rα signaling regulates macrophage functional polarization in a context-dependent manner. Pharmacological targeting of macrophage polarization therefore requires disease stage-specific treatment strategies. Research in Context Alternative (M2-type) macrophage activation through IL-4Rα promotes liver inflammation and fibrosis progression but speeds up fibrosis reversal. This demonstrates context dependent, opposing roles of M2-type macrophages. During reversal IL-4Rα induces fibrolytic MMPs, especially MMP-12, through STAT6. Liver-specific antisense oligonucleotides efficiently block IL-4Rα expression and attenuate fibrosis progression. IL-4Rα is considered a central switch for alternative macrophage polarization. IL-4Rα on macrophages is shown to differentially regulate liver fibrosis progression vs reversal. Therapeutic IL-4Rα antisense oligonucleotides replicate these findings. MMP-12 induced via macrophage IL-4Rα is a key promoter of fibrosis reversal.
Collapse
Affiliation(s)
- Shih-Yen Weng
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Xiaoyu Wang
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Santosh Vijayan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Yilang Tang
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Yong Ook Kim
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Kornelius Padberg
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tommy Regen
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Olena Molokanova
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tao Chen
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hansjörg Schild
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology, Institute of Infectious Disease and Molecular Medicine, South African Medical Research Council, Cape Town, South Africa
| | | | | | - Ari Waisman
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ernesto Bockamp
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
17
|
Nordestgaard BG, Nicholls SJ, Langsted A, Ray KK, Tybjærg-Hansen A. Advances in lipid-lowering therapy through gene-silencing technologies. Nat Rev Cardiol 2018; 15:261-272. [PMID: 29417937 DOI: 10.1038/nrcardio.2018.3] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
New treatment opportunities are emerging in the field of lipid-lowering therapy through gene-silencing approaches. Both antisense oligonucleotide inhibition and small interfering RNA technology aim to degrade gene mRNA transcripts to reduce protein production and plasma lipoprotein levels. Elevated levels of LDL, remnant lipoproteins, and lipoprotein(a) all cause cardiovascular disease, whereas elevated levels of triglyceride-rich lipoproteins in some patients can cause acute pancreatitis. The levels of each of these lipoproteins can be reduced using gene-silencing therapies by targeting proteins that have an important role in lipoprotein production or removal (for example, the protein products of ANGPTL3, APOB, APOC3, LPA, and PCSK9). Using this technology, plasma levels of these lipoproteins can be reduced by 50-90% with 2-12 injections per year; such dramatic reductions are likely to reduce the incidence of cardiovascular disease or acute pancreatitis in at-risk patients. The reported adverse effects of these new therapies include injection-site reactions, flu-like symptoms, and low blood platelet counts. However, newer-generation drugs are more efficiently delivered to liver cells, requiring lower drug doses, which leads to fewer adverse effects. Although these findings are promising, robust evidence of cardiovascular disease reduction and long-term safety is needed before these gene-silencing technologies can have widespread implementation. Before the availability of such evidence, these drugs might have roles in patients with unmet medical needs through orphan indications.
Collapse
Affiliation(s)
- Børge G Nordestgaard
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, North Terrace, Adelaide 5000, South Australia, Australia
| | - Anne Langsted
- Department of Clinical Biochemistry and The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, Reynolds Building, St Dunstan's Road, London W6 8RP, UK
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsveg 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
18
|
Moschovis D, Gazouli M, Tzouvala M, Vezakis A, Karamanolis G. Long non-coding RNA in pancreatic adenocarcinoma and pancreatic neuroendocrine tumors. Ann Gastroenterol 2017; 30:622-628. [PMID: 29118556 PMCID: PMC5670281 DOI: 10.20524/aog.2017.0185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022] Open
Abstract
Interest in non-coding regions of DNA has been increasing since the mapping of the human genome revealed that human DNA contains far fewer genes encoding proteins than previously expected. However, analysis of the derivatives of DNA transcription (transcriptomics) revealed that the majority of the genetic material is transcribed into non-coding RNA (ncRNA), indicating that these molecules probably provide the functional diversity and complexity of the physiology of the human body that cannot be attributed to the proteins. Of these ncRNA, long ncRNA (lncRNA) have a length greater than 200 nucleotides and share many common components with the coding messenger RNA (mRNA): They are transcribed by RNA polymerase II, comprised of multiple exons and subjected to normal RNA splicing giving RNA products of several kilobases. Scientific data reveal the regulatory role of lncRNA in the control of gene expression during cell development and homeostasis. However, to date, very few lncRNAs have been characterized in depth, and lncRNAs are thought to have a wide range of functions in cellular and developmental processes. These molecules will have the possibility to be used as biomarkers and contribute to the development of targeted therapies. Concerning pancreatic cancer, there are limited data in the literature that correlate the growth of these tumors with deregulation of various lncRNA. We herein review the literature regarding the role of lncRNA as a diagnostic and prognostic biomarker and possible therapeutic target in the neoplasms of the pancreas, particularly pancreatic adenocarcinoma and pancreatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Dimitrios Moschovis
- Department of Gastroenterology, Agios Panteleimon General Hospital, Nikea (Dimitrios Moschovis, Maria Tzouvala), Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (Maria Gazouli), Greece
| | - Maria Tzouvala
- Department of Gastroenterology, Agios Panteleimon General Hospital, Nikea (Dimitrios Moschovis, Maria Tzouvala), Greece
| | - Antonios Vezakis
- 2 Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens (Antonios Vezakis), Greece
| | - George Karamanolis
- Gastroenterology Unit, 2 Department of Surgery, Aretaieio University Hospital, National and Kapodistrian University of Athens (George Karamanolis), Greece
| |
Collapse
|
19
|
Deshpande D, Janero DR, Segura-Ibarra V, Blanco E, Amiji MM. Nucleic Acid Delivery for Endothelial Dysfunction in Cardiovascular Diseases. Methodist Debakey Cardiovasc J 2017; 12:134-140. [PMID: 27826366 DOI: 10.14797/mdcj-12-3-134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction has been implicated in the pathophysiology of multiple cardiovascular diseases and involves components of both innate and acquired immune mechanisms. Identifying signature patterns and targets associated with endothelial dysfunction can help in the development of novel nanotherapeutic platforms for treatment of vascular diseases. This review discusses nucleic acid-based regulation of endothelial function and the different nucleic acid-based nanotherapeutic approaches designed to target endothelial dysfunction in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | | | - Elvin Blanco
- Houston Methodist Research Institute, Houston, Texas
| | - Mansoor M Amiji
- Northeastern University, Boston, Massachusetts; King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
20
|
Tai L, Liu C, Jiang K, Chen X, Wei G, Lu W, Pan W. Noninvasive delivery of oligonucleotide by penetratin-modified polyplexes to inhibit protein expression of intraocular tumor. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2091-2100. [PMID: 28435135 DOI: 10.1016/j.nano.2017.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/08/2017] [Accepted: 04/11/2017] [Indexed: 10/19/2022]
Abstract
Our present study aimed to develop an antisense oligonucleotide (ASO) delivery system to achieve gene silencing in intraocular tumor via topical instillation. ASO specific for luciferase was chosen as model drug, polyamidoamine (PG5) was employed to condense ASO, and penetratin (Pene) was used to enhance cellular uptake. Nanoscale PG5/ASO/Pene polyplex was stabilized via noncovalent bonding. In vitro evaluations indicated that PG5/ASO/Pene exhibited improved cell-penetrating and gene silencing ability compared with naked ASO and PG5/ASO. Subcutaneous and orthotopic tumor models expressing luciferase were established in nude mice. After treated by PG5/ASO/Pene, immunohistochemical results of subcutaneous tumors showed significant inhibition of luciferase expression via peritumoral injection, and bioluminescence from orthotopic tumor was obviously weakened via topical instillation. To date, few works were successful in noninvasive treatment of intraocular diseases using antisense strategy, this penetratin-modified polyplex could be a promising vector to inhibit protein expression by effectively delivering ASOs into the eye.
Collapse
Affiliation(s)
- Lingyu Tai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China; Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Chang Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Kuan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Xishan Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China.
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Weisan Pan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
21
|
Somatic genome editing with CRISPR/Cas9 generates and corrects a metabolic disease. Sci Rep 2017; 7:44624. [PMID: 28300165 PMCID: PMC5353616 DOI: 10.1038/srep44624] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/09/2017] [Indexed: 12/14/2022] Open
Abstract
Germline manipulation using CRISPR/Cas9 genome editing has dramatically accelerated the generation of new mouse models. Nonetheless, many metabolic disease models still depend upon laborious germline targeting, and are further complicated by the need to avoid developmental phenotypes. We sought to address these experimental limitations by generating somatic mutations in the adult liver using CRISPR/Cas9, as a new strategy to model metabolic disorders. As proof-of-principle, we targeted the low-density lipoprotein receptor (Ldlr), which when deleted, leads to severe hypercholesterolemia and atherosclerosis. Here we show that hepatic disruption of Ldlr with AAV-CRISPR results in severe hypercholesterolemia and atherosclerosis. We further demonstrate that co-disruption of Apob, whose germline loss is embryonically lethal, completely prevented disease through compensatory inhibition of hepatic LDL production. This new concept of metabolic disease modeling by somatic genome editing could be applied to many other systemic as well as liver-restricted disorders which are difficult to study by germline manipulation.
Collapse
|
22
|
Cellular uptake and trafficking of antisense oligonucleotides. Nat Biotechnol 2017; 35:230-237. [PMID: 28244996 DOI: 10.1038/nbt.3779] [Citation(s) in RCA: 399] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 12/22/2016] [Indexed: 01/08/2023]
Abstract
Antisense oligonucleotides (ASOs) modified with phosphorothioate (PS) linkages and different 2' modifications can be used either as drugs (e.g., to treat homozygous familial hypercholesterolemia and spinal muscular atrophy) or as research tools to alter gene expression. PS-ASOs can enter cells without additional modification or formulation and can be designed to mediate sequence-specific cleavage of different types of RNA (including mRNA and non-coding RNA) targeted by endogenous RNase H1. Although PS-ASOs function in both the cytoplasm and nucleus, localization to different subcellular regions can affect their therapeutic potency. Cellular uptake and intracellular distribution of PS ASOs are mediated by protein interactions. The main proteins involved in these processes have been identified, and intracellular sites in which PS ASOs are active, or inactive, cataloged.
Collapse
|
23
|
Cervantes Gracia K, Llanas-Cornejo D, Husi H. CVD and Oxidative Stress. J Clin Med 2017; 6:E22. [PMID: 28230726 PMCID: PMC5332926 DOI: 10.3390/jcm6020022] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
Nowadays, it is known that oxidative stress plays at least two roles within the cell, the generation of cellular damage and the involvement in several signaling pathways in its balanced normal state. So far, a substantial amount of time and effort has been expended in the search for a clear link between cardiovascular disease (CVD) and the effects of oxidative stress. Here, we present an overview of the different sources and types of reactive oxygen species in CVD, highlight the relationship between CVD and oxidative stress and discuss the most prominent molecules that play an important role in CVD pathophysiology. Details are given regarding common pharmacological treatments used for cardiovascular distress and how some of them are acting upon ROS-related pathways and molecules. Novel therapies, recently proposed ROS biomarkers, as well as future challenges in the field are addressed. It is apparent that the search for a better understanding of how ROS are contributing to the pathophysiology of CVD is far from over, and new approaches and more suitable biomarkers are needed for the latter to be accomplished.
Collapse
Affiliation(s)
- Karla Cervantes Gracia
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| | - Daniel Llanas-Cornejo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
24
|
LOX-1 and Its Splice Variants: A New Challenge for Atherosclerosis and Cancer-Targeted Therapies. Int J Mol Sci 2017; 18:ijms18020290. [PMID: 28146073 PMCID: PMC5343826 DOI: 10.3390/ijms18020290] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/15/2017] [Accepted: 01/23/2017] [Indexed: 12/13/2022] Open
Abstract
Alternative splicing (AS) is a process in which precursor messenger RNA (pre-mRNA) splicing sites are differentially selected to diversify the protein isoform population. Changes in AS patterns have an essential role in normal development, differentiation and response to physiological stimuli. It is documented that AS can generate both “risk” and “protective” splice variants that can contribute to the pathogenesis of several diseases including atherosclerosis. The main endothelial receptor for oxidized low-density lipoprotein (ox-LDLs) is LOX-1 receptor protein encoded by the OLR1 gene. When OLR1 undergoes AS events, it generates three variants: OLR1, OLR1D4 and LOXIN. The latter lacks exon 5 and two-thirds of the functional domain. Literature data demonstrate a protective role of LOXIN in pathologies correlated with LOX-1 overexpression such as atherosclerosis and tumors. In this review, we summarize recent developments in understanding of OLR1 AS while also highlighting data warranting further investigation of this process as a novel therapeutic target.
Collapse
|
25
|
Mitsuoka Y, Yamamoto T, Kugimiya A, Waki R, Wada F, Tahara S, Sawamura M, Noda M, Fujimura Y, Kato Y, Hari Y, Obika S. Triazole- and Tetrazole-Bridged Nucleic Acids: Synthesis, Duplex Stability, Nuclease Resistance, and in Vitro and in Vivo Antisense Potency. J Org Chem 2016; 82:12-24. [PMID: 27936689 DOI: 10.1021/acs.joc.6b02417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Antisense oligonucleotides are attractive therapeutic agents for several types of disease. One of the most promising modifications of antisense oligonucleotides is the introduction of bridged nucleic acids. As we report here, we designed novel bridged nucleic acids, triazole-bridged nucleic acid (TrNA), and tetrazole-bridged nucleic acid (TeNA), whose sugar conformations are restricted to N-type by heteroaromatic ring-bridged structures. We then successfully synthesized TrNA and TeNA and introduced these monomers into oligonucleotides. In UV-melting experiments, TrNA-modified oligonucleotides exhibited increased binding affinity toward complementary RNA and decreased binding affinity toward complementary DNA, although TeNA-modified oligonucleotides were decomposed under the annealing conditions. Enzymatic degradation experiments demonstrated that introduction of TrNA at the 3'-terminus rendered oligonucleotides resistant to nuclease digestion. Furthermore, we tested the silencing potencies of TrNA-modified antisense oligonucleotides using in vitro and in vivo assays. These experiments revealed that TrNA-modified antisense oligonucleotides induced potent downregulation of gene expression in liver. In addition, TrNA-modified antisense oligonucleotides showed a tendency for increased liver biodistribution. Taken together, our findings indicate that TrNA is a good candidate for practical application in antisense methodology.
Collapse
Affiliation(s)
- Yasunori Mitsuoka
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan.,Discovery Research Laboratory for Innovative Frontier Medicines, Shionogi & Co., Ltd. , 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Tsuyoshi Yamamoto
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| | - Akira Kugimiya
- Discovery Research Laboratory for Innovative Frontier Medicines, Shionogi & Co., Ltd. , 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Reiko Waki
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| | - Fumito Wada
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| | - Saori Tahara
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| | - Motoki Sawamura
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| | - Mio Noda
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| | - Yuko Fujimura
- Discovery Research Laboratory for Innovative Frontier Medicines, Shionogi & Co., Ltd. , 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Yuki Kato
- Research Laboratory for Development, Shionogi & Co., Ltd. , 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825, Japan
| | - Yoshiyuki Hari
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan.,Faculty of Pharmaceutical Sciences, Tokushima Bunri University , Nishihama, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Satoshi Obika
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita Osaka 565-0871, Japan
| |
Collapse
|
26
|
Reyes-Soffer G, Moon B, Hernandez-Ono A, Dionizovik-Dimanovski M, Dionizovick-Dimanovski M, Jimenez J, Obunike J, Thomas T, Ngai C, Fontanez N, Donovan DS, Karmally W, Holleran S, Ramakrishnan R, Mittleman RS, Ginsberg HN. Complex effects of inhibiting hepatic apolipoprotein B100 synthesis in humans. Sci Transl Med 2016; 8:323ra12. [PMID: 26819195 DOI: 10.1126/scitranslmed.aad2195] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mipomersen is a 20mer antisense oligonucleotide (ASO) that inhibits apolipoprotein B (apoB) synthesis; its low-density lipoprotein (LDL)-lowering effects should therefore result from reduced secretion of very-low-density lipoprotein (VLDL). We enrolled 17 healthy volunteers who received placebo injections weekly for 3 weeks followed by mipomersen weekly for 7 to 9 weeks. Stable isotopes were used after each treatment to determine fractional catabolic rates and production rates of apoB in VLDL, IDL (intermediate-density lipoprotein), and LDL, and of triglycerides in VLDL. Mipomersen significantly reduced apoB in VLDL, IDL, and LDL, which was associated with increases in fractional catabolic rates of VLDL and LDL apoB and reductions in production rates of IDL and LDL apoB. Unexpectedly, the production rates of VLDL apoB and VLDL triglycerides were unaffected. Small interfering RNA-mediated knockdown of apoB expression in human liver cells demonstrated preservation of apoB secretion across a range of apoB synthesis. Titrated ASO knockdown of apoB mRNA in chow-fed mice preserved both apoB and triglyceride secretion. In contrast, titrated ASO knockdown of apoB mRNA in high-fat-fed mice resulted in stepwise reductions in both apoB and triglyceride secretion. Mipomersen lowered all apoB lipoproteins without reducing the production rate of either VLDL apoB or triglyceride. Our human data are consistent with long-standing models of posttranscriptional and posttranslational regulation of apoB secretion and are supported by in vitro and in vivo experiments. Targeting apoB synthesis may lower levels of apoB lipoproteins without necessarily reducing VLDL secretion, thereby lowering the risk of steatosis associated with this therapeutic strategy.
Collapse
Affiliation(s)
- Gissette Reyes-Soffer
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA.
| | - Byoung Moon
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Antonio Hernandez-Ono
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | | | | | - Jhonsua Jimenez
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Joseph Obunike
- Biological Sciences Department, New York City College of Technology, 300 Jay Street, Brooklyn, NY 11201, USA
| | - Tiffany Thomas
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Colleen Ngai
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Nelson Fontanez
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Daniel S Donovan
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Wahida Karmally
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Stephen Holleran
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Rajasekhar Ramakrishnan
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | | | - Henry N Ginsberg
- Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
27
|
Cardon LR, Harris T. Precision medicine, genomics and drug discovery. Hum Mol Genet 2016; 25:R166-R172. [PMID: 27538422 DOI: 10.1093/hmg/ddw246] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022] Open
Abstract
The hope for precision medicine has long been on the drug discovery horizon, well before the Human Genome Project gave it promise at the turn of the 21st century. In oncology, the concept has finally been realized and is now firmly embedded in ongoing drug discovery programs, and with many recent therapies involving some level of patient/disease stratification, including some highly personalized treatments. In addition, several drugs for rare diseases have been recently approved or are in late-stage clinical development, and new delivery modalities in cell and gene therapy and oligonucleotide approaches are yielding exciting new medicines for rare diseases of unmet need. For common complex diseases, however, the GWAS-driven advances in annotation of the genetic architecture over the past decade have not led to a concomitant shift in refined treatments. Similarly, attempts to disentangle treatment responders from non-responders via genetic predictors in pharmacogenetics studies have not met their anticipated success. It is possible that common diseases are simply lagging behind due to the inherent time lag with drug discovery, but it is also possible that their inherent multifactorial nature and their etiological and clinical heterogeneity will prove more resistant to refined treatment paradigms. The emergence of population-based resources in electronic health records, coupled with the rapid expansion of mobile devices and digital health may help to refine the measurement of phenotypic outcomes to match the exquisite detail emerging at the molecular level.
Collapse
Affiliation(s)
- Lon R Cardon
- Target Sciences, GlaxoSmithKline, King of Prussia, PA, USA
| | - Tim Harris
- Venture Partner SV Life Sciences, Boston, MA, USA
| |
Collapse
|
28
|
Integrated Safety Assessment of 2'-O-Methoxyethyl Chimeric Antisense Oligonucleotides in NonHuman Primates and Healthy Human Volunteers. Mol Ther 2016; 24:1771-1782. [PMID: 27357629 PMCID: PMC5112040 DOI: 10.1038/mt.2016.136] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023] Open
Abstract
The common chemical and biological properties of antisense oligonucleotides provide the opportunity to identify and characterize chemical class effects across species. The chemical class that has proven to be the most versatile and best characterized is the 2′-O-methoxyethyl chimeric antisense oligonucleotides. In this report we present an integrated safety assessment of data obtained from controlled dose-ranging studies in nonhuman primates (macaques) and healthy human volunteers for 12 unique 2′-O-methoxyethyl chimeric antisense oligonucleotides. Safety was assessed by the incidence of safety signals in standardized laboratory tests for kidney and liver function, hematology, and complement activation; as well as by the mean test results as a function of dose level over time. At high doses a number of toxicities were observed in nonhuman primates. However, no class safety effects were identified in healthy human volunteers from this integrated data analysis. Effects on complement in nonhuman primates were not observed in humans. Nonhuman primates predicted safe doses in humans, but over predicted risk of complement activation and effects on platelets. Although limited to a single chemical class, comparisons from this analysis are considered valid and accurate based on the carefully controlled setting for the specified study populations and within the total exposures studied.
Collapse
|
29
|
Fox RG, Lytle NK, Jaquish DV, Park FD, Ito T, Bajaj J, Koechlein CS, Zimdahl B, Yano M, Kopp J, Kritzik M, Sicklick J, Sander M, Grandgenett PM, Hollingsworth MA, Shibata S, Pizzo D, Valasek M, Sasik R, Scadeng M, Okano H, Kim Y, MacLeod AR, Lowy AM, Reya T. Image-based detection and targeting of therapy resistance in pancreatic adenocarcinoma. Nature 2016; 534:407-411. [PMID: 27281208 PMCID: PMC4998062 DOI: 10.1038/nature17988] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 04/07/2016] [Indexed: 01/08/2023]
Abstract
Pancreatic intraepithelial neoplasia (PanIN) is a premalignant lesion that can progress to pancreatic ductal adenocarcinoma, a highly lethal malignancy marked by its late stage at clinical presentation and profound drug resistance1. The genomic alterations that commonly occur in pancreatic cancer include activation of KRAS2 and inactivation of p53, and SMAD42-4. To date, however, it has been challenging to target these pathways therapeutically; thus the search for other key mediators of pancreatic cancer growth remains an important endeavor. Here we show that the stem cell determinant Musashi (Msi) is a critical element of pancreatic cancer progression in both genetic models and patient derived xenografts. Specifically, we developed Msi reporter mice that allowed image based tracking of stem cell signals within cancers, revealing that Msi expression rises as PanIN progresses to adenocarcinoma, and that Msi-expressing cells are key drivers of pancreatic cancer: they preferentially harbor the capacity to propagate adenocarcinoma, are enriched in circulating tumor cells, and are markedly drug resistant. This population could be effectively targeted by deletion of either Msi1 or Msi2, which led to a striking defect in PanIN progression to adenocarcinoma and an improvement in overall survival. Msi inhibition also blocked the growth of primary patient-derived tumors, suggesting that this signal is required for human disease. To define the translational potential of this work we developed antisense oligonucleotides against Msi; these showed reliable tumor penetration, uptake and target inhibition, and effectively blocked pancreatic cancer growth. Collectively, these studies highlight Msi reporters as a unique tool to identify therapy resistance, and define Msi signaling as a central regulator of pancreatic cancer.
Collapse
Affiliation(s)
- Raymond G Fox
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Nikki K Lytle
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Dawn V Jaquish
- Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA.,Department of Surgery, Division of Surgical Oncology, University of California San Diego School of Medicine, La Jolla, CA
| | - Frederick D Park
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA.,Department of Medicine, Division of Gastroenterology, University of California San Diego School of Medicine, La Jolla, CA
| | - Takahiro Ito
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Jeevisha Bajaj
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Claire S Koechlein
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Bryan Zimdahl
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Masato Yano
- Department of Physiology, Graduate School of Medicine, Keio University, Keio, Japan
| | - Janel Kopp
- Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine, La Jolla, CA
| | - Marcie Kritzik
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| | - Jason Sicklick
- Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA.,Department of Surgery, Division of Surgical Oncology, University of California San Diego School of Medicine, La Jolla, CA
| | - Maike Sander
- Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine, La Jolla, CA
| | - Paul M Grandgenett
- Eppley Institute For Research in Cancer and Allied Diseases, Department of Pathology, University of Nebraska Medical Center, Omaha, NE
| | - Michael A Hollingsworth
- Eppley Institute For Research in Cancer and Allied Diseases, Department of Pathology, University of Nebraska Medical Center, Omaha, NE
| | - Shinsuke Shibata
- Department of Physiology, Graduate School of Medicine, Keio University, Keio, Japan
| | - Donald Pizzo
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA
| | - Mark Valasek
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, CA
| | - Roman Sasik
- Center for Computational Biology and Bioinformatics, University of California San Diego School of Medicine, La Jolla, CA
| | - Miriam Scadeng
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA
| | - Hideyuki Okano
- Department of Physiology, Graduate School of Medicine, Keio University, Keio, Japan
| | - Youngsoo Kim
- Department of Oncology Drug Discovery, Ionis pharmaceuticals, Carlsbad, CA
| | - A Robert MacLeod
- Department of Oncology Drug Discovery, Ionis pharmaceuticals, Carlsbad, CA
| | - Andrew M Lowy
- Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA.,Department of Surgery, Division of Surgical Oncology, University of California San Diego School of Medicine, La Jolla, CA
| | - Tannishtha Reya
- Departments of Pharmacology and Medicine, University of California San Diego School of Medicine La Jolla, CA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA.,Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA
| |
Collapse
|
30
|
Roth CM. Delivery of Genes and Oligonucleotides. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
31
|
Fang Y, Fullwood MJ. Roles, Functions, and Mechanisms of Long Non-coding RNAs in Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2016; 14:42-54. [PMID: 26883671 PMCID: PMC4792843 DOI: 10.1016/j.gpb.2015.09.006] [Citation(s) in RCA: 746] [Impact Index Per Article: 82.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/31/2015] [Accepted: 09/17/2015] [Indexed: 12/28/2022]
Abstract
Long non-coding RNAs (lncRNAs) play important roles in cancer. They are involved in chromatin remodeling, as well as transcriptional and post-transcriptional regulation, through a variety of chromatin-based mechanisms and via cross-talk with other RNA species. lncRNAs can function as decoys, scaffolds, and enhancer RNAs. This review summarizes the characteristics of lncRNAs, including their roles, functions, and working mechanisms, describes methods for identifying and annotating lncRNAs, and discusses future opportunities for lncRNA-based therapies using antisense oligonucleotides.
Collapse
Affiliation(s)
- Yiwen Fang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Melissa J Fullwood
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore; Yale-NUS Liberal Arts College, Singapore 138527, Singapore.
| |
Collapse
|
32
|
Evaluation of the effects of chemically different linkers on hepatic accumulations, cell tropism and gene silencing ability of cholesterol-conjugated antisense oligonucleotides. J Control Release 2016; 226:57-65. [PMID: 26855051 DOI: 10.1016/j.jconrel.2016.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/14/2016] [Accepted: 02/04/2016] [Indexed: 01/04/2023]
Abstract
Cholesterol conjugation of oligonucleotides is an attractive way to deliver the oligonucleotides specifically to the liver. However cholesterol-conjugated antisense oligonucleotides (ASOs) mainly accumulate in non-parenchymal cells (NPCs) such as Kupffer cells. In this study, to increase the hepatic accumulation of cholesterol-conjugated ASOs, we prepared a variety of linkers for cholesterol conjugation to anti-Pcsk9 ASOs and examined their effects on pharmacological parameters. Hepatic accumulation of ASO was dramatically increased with cholesterol conjugation. The increase in hepatic accumulation depended largely on the linker chemistry of each cholesterol-conjugated ASO. In addition to hepatic accumulation, the cell tropism of each cholesterol-conjugated ASO tended to depend on their linker. Although a linker bearing a disulfide bond accumulated mainly in NPCs, hexamethylene succinimide linker accumulated mainly in hepatocytes. To estimate the benefits of releasing ASO from the conjugated cholesterol in hepatocyte, we designed another linker based on hexamethylene succinimide, which has a phosphodiester bond between the linker and the ASO. The cholesterol-conjugated ASO bearing such a phosphodiester bond showed a significantly improved Pcsk9 mRNA inhibitory effect compared to its counterpart, cholesterol-conjugated ASO with a phosphorothioate bond, while the hepatic accumulation of both cholesterol-conjugated ASOs was comparable, indicating the effectiveness of removing the conjugated cholesterol for ASO activity. In toxicity analysis, some of the linkers induced lethal toxicities when they were injected at high concentrations (>600μM). These toxicities were attributed to decreased platelet levels in the blood, suggesting an interaction between cholesterol-conjugated ASO and platelets. Our findings may provide a guideline for the design of molecule-conjugated ASOs.
Collapse
|
33
|
Zhou T, Kim Y, MacLeod AR. Targeting Long Noncoding RNA with Antisense Oligonucleotide Technology as Cancer Therapeutics. Methods Mol Biol 2016; 1402:199-213. [PMID: 26721493 DOI: 10.1007/978-1-4939-3378-5_16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Recent annotation of the human transcriptome revealed that only 2 % of the genome encodes proteins while the majority of human genome is transcribed into noncoding RNAs. Although we are just beginning to understand the diverse roles long noncoding RNAs (lncRNAs) play in molecular and cellular processes, they have potentially important roles in human development and pathophysiology. However, targeting of RNA by traditional structure-based design of small molecule inhibitors has been difficult, due to a lack of understanding of the dynamic tertiary structures most RNA molecules adopt. Antisense oligonucleotides (ASOs) are capable of targeting specific genes or transcripts directly through Watson-Crick base pairing and thus can be designed based on sequence information alone. These agents have made possible specific targeting of "non-druggable targets" including RNA molecules. Here we describe how ASOs can be applied in preclinical studies to reduce levels of lncRNAs of interest.
Collapse
Affiliation(s)
- Tianyuan Zhou
- Antisense Drug Discovery, Oncology, Isis Pharmaceuticals, Inc, 2855 Gazelle Court, Carlsbad, CA, 92010, USA.
| | - Youngsoo Kim
- Antisense Drug Discovery, Oncology, Isis Pharmaceuticals, Inc, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - A Robert MacLeod
- Antisense Drug Discovery, Oncology, Isis Pharmaceuticals, Inc, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| |
Collapse
|
34
|
Milman S, Barzilai N. Dissecting the Mechanisms Underlying Unusually Successful Human Health Span and Life Span. Cold Spring Harb Perspect Med 2015; 6:a025098. [PMID: 26637439 DOI: 10.1101/cshperspect.a025098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Humans age at different rates and families with exceptional survival provide the opportunity to understand why some people age slower than others. Unique features exhibited by centenarians include a family history of longevity, compression of morbidity with resultant extension of health span, and biomarkers such as low-circulating insulin-like growth factor 1 (IGF-1) and elevated high-density lipoprotein (HDL) cholesterol levels. Given the rarity of the centenarian phenotype, it has not been surprising that the use of discovery methods that relied on common population single nucleotide polymorphisms (SNPs) to unlock the genetic determinants of exceptional longevity have not yielded significant results. Conversely, gene sequencing has resulted in discoveries of functional gene variants that support several of the centenarian phenotypes. These discoveries have led to the strategic developments of drugs that may delay aging and prolong health span.
Collapse
Affiliation(s)
- Sofiya Milman
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York 10461 Institute for Aging Research, Albert Einstein College of Medicine, New York, New York 10461
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York 10461 Institute for Aging Research, Albert Einstein College of Medicine, New York, New York 10461 Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
35
|
Niemietz C, Chandhok G, Schmidt H. Therapeutic Oligonucleotides Targeting Liver Disease: TTR Amyloidosis. Molecules 2015; 20:17944-75. [PMID: 26437390 PMCID: PMC6332041 DOI: 10.3390/molecules201017944] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/13/2022] Open
Abstract
The liver has become an increasingly interesting target for oligonucleotide therapy. Mutations of the gene encoding transthyretin (TTR), expressed in vast amounts by the liver, result in a complex degenerative disease, termed familial amyloid polyneuropathy (FAP). Misfolded variants of TTR are linked to the establishment of extracellular protein deposition in various tissues, including the heart and the peripheral nervous system. Recent progress in the chemistry and formulation of antisense (ASO) and small interfering RNA (siRNA) designed for a knockdown of TTR mRNA in the liver has allowed to address the issue of gene-specific molecular therapy in a clinical setting of FAP. The two therapeutic oligonucleotides bind to RNA in a sequence specific manner but exploit different mechanisms. Here we describe major developments that have led to the advent of therapeutic oligonucleotides for treatment of TTR-related disease.
Collapse
MESH Headings
- Amyloid Neuropathies, Familial/genetics
- Amyloid Neuropathies, Familial/therapy
- Animals
- Clinical Studies as Topic
- Drug Evaluation, Preclinical
- Gene Silencing
- Genetic Therapy
- Humans
- Liver Diseases/genetics
- Liver Diseases/therapy
- Mutation
- Oligonucleotides/administration & dosage
- Oligonucleotides/chemistry
- Oligonucleotides/genetics
- Oligonucleotides/therapeutic use
- Oligonucleotides, Antisense/administration & dosage
- Oligonucleotides, Antisense/chemistry
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/therapeutic use
- Prealbumin/genetics
- RNA Interference
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- Treatment Outcome
Collapse
Affiliation(s)
- Christoph Niemietz
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, D-48149 Münster, Germany.
| | - Gursimran Chandhok
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, D-48149 Münster, Germany.
| | - Hartmut Schmidt
- Klinik für Transplantationsmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, D-48149 Münster, Germany.
| |
Collapse
|
36
|
Pandey SK, Wheeler TM, Justice SL, Kim A, Younis HS, Gattis D, Jauvin D, Puymirat J, Swayze EE, Freier SM, Bennett CF, Thornton CA, MacLeod AR. Identification and characterization of modified antisense oligonucleotides targeting DMPK in mice and nonhuman primates for the treatment of myotonic dystrophy type 1. J Pharmacol Exp Ther 2015; 355:329-40. [PMID: 26330536 DOI: 10.1124/jpet.115.226969] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/31/2015] [Indexed: 01/07/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults. DM1 is caused by an expanded CTG repeat in the 3'-untranslated region of DMPK, the gene encoding dystrophia myotonica protein kinase (DMPK). Antisense oligonucleotides (ASOs) containing 2',4'-constrained ethyl-modified (cEt) residues exhibit a significantly increased RNA binding affinity and in vivo potency relative to those modified with other 2'-chemistries, which we speculated could translate to enhanced activity in extrahepatic tissues, such as muscle. Here, we describe the design and characterization of a cEt gapmer DMPK ASO (ISIS 486178), with potent activity in vitro and in vivo against mouse, monkey, and human DMPK. Systemic delivery of unformulated ISIS 486718 to wild-type mice decreased DMPK mRNA levels by up to 90% in liver and skeletal muscle. Similarly, treatment of either human DMPK transgenic mice or cynomolgus monkeys with ISIS 486178 led to up to 70% inhibition of DMPK in multiple skeletal muscles and ∼50% in cardiac muscle in both species. Importantly, inhibition of DMPK was well tolerated and was not associated with any skeletal muscle or cardiac toxicity. Also interesting was the demonstration that the inhibition of DMPK mRNA levels in muscle was maintained for up to 16 and 13 weeks post-treatment in mice and monkeys, respectively. These results demonstrate that cEt-modified ASOs show potent activity in skeletal muscle, and that this attractive therapeutic approach warrants further clinical investigation to inhibit the gain-of-function toxic RNA underlying the pathogenesis of DM1.
Collapse
Affiliation(s)
- Sanjay K Pandey
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Thurman M Wheeler
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Samantha L Justice
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Aneeza Kim
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Husam S Younis
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Danielle Gattis
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Dominic Jauvin
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Jack Puymirat
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Eric E Swayze
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Susan M Freier
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - C Frank Bennett
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - Charles A Thornton
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| | - A Robert MacLeod
- Isis Pharmaceuticals Inc., Carlsbad, CA (S.K.P., S.L.J., A.K., H.S.Y., D.G., E.E.S., S.M.F., C.F.B., A.R.M.); Department of Neurology and Center of Neural Development and Disease, University of Rochester, Rochester, New York (T.M.W., C.A.T.); Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts (T.M.W.); and Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Canada (D.J., J.P.)
| |
Collapse
|
37
|
Bell TA, Graham MJ, Baker BF, Crooke RM. Therapeutic inhibition of apoC-III for the treatment of hypertriglyceridemia. ACTA ACUST UNITED AC 2015. [DOI: 10.2217/clp.15.7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Abstract
Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. The disease originates from low levels of SMN protein due to deletion and/or mutations of SMN1 coupled with the inability of SMN2 to compensate for the loss of SMN1. While SMN1 and SMN2 are nearly identical, SMN2 predominantly generates a truncated protein (SMNΔ7) due to skipping of exon 7, the last coding exon. Several avenues for SMA therapy are being explored, including means to enhance SMN2 transcription, correct SMN2 exon 7 splicing, stabilize SMN/SMNΔ7 protein, manipulate SMN-regulated pathways and SMN1 gene delivery by viral vectors. This review focuses on the aspects of target discovery, validations and outcome measures for a promising therapy of SMA.
Collapse
|
39
|
Advances in induced pluripotent stem cells, genomics, biomarkers, and antiplatelet therapy highlights of the year in JCTR 2013. J Cardiovasc Transl Res 2015; 7:518-25. [PMID: 24659088 DOI: 10.1007/s12265-014-9555-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/19/2014] [Indexed: 12/22/2022]
Abstract
The Journal provides the clinician and scientist with the latest advances in discovery research, emerging technologies, preclinical research design and testing, and clinical trials. We highlight advances in areas of induced pluripotent stem cells, genomics, biomarkers, multimodality imaging, and antiplatelet biology and therapy. The top publications are critically discussed and presented along with anatomical reviews and FDA insight to provide context.
Collapse
|
40
|
He X, Wu DF, Ji J, Ling WP, Chen XL, Chen YX. Ultrasound microbubble-carried PNA targeting to c-myc mRNA inhibits the proliferation of rabbit iliac arterious smooth muscle cells and intimal hyperplasia. Drug Deliv 2015; 23:2482-2487. [PMID: 25726989 DOI: 10.3109/10717544.2015.1014947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To elucidate the transfected effect of albumin ultrasound microbubbles carrying peptide nucleic acids (PNAs) against c-myc gene to the vascular walls and their effect on the intimal proliferation induced by vascular denudation. METHODS A rabbit iliac artery intimal proliferation model was constructed and PNA against c-myc mRNA was designed and synthesized and was added to albumin solution before ultrasound microbubbles were prepared and encapsulated in matrix of albumin. The ultrasound microbubbles carrying PNA were transfected to intima under ultrasound exposure. The transfected effect was identified by a histochemical method and the expression of c-myc was detected by in situ hybridization. The proliferation of intimal smooth muscle cells was estimated by the expression of proliferative cell nuclear antigen (PCNA) of them. The intimal area and thickness were judged morphologically for intimal hyperplasia. RESULTS The ultrasound microbubbles with PNA were successfully prepared and c-myc PNA was transfected to vascular intimal cells. The expression of c-myc and PCNA by intimal vascular smooth muscle cells (vSMCs) was inhibited significantly and the intimal thickness and area were reduced remarkably. CONCLUSION Transfection of c-myc PNA could inhibit proliferartion of vSMCs and intima in the rabbit iliac artery intimal proliferation model and the targeted transfection of albumin ultrasound microbubbles carrying PNA offers a feasible way to facilitate its access to specific cells in vivo and produce bioavailability.
Collapse
Affiliation(s)
- Xia He
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Da-Fang Wu
- b Department of Endocrinology , 451 Hospital of PLA , Xi'an, Shanxi , PR China
| | - Jun Ji
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Wen-Ping Ling
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Xiao-Ling Chen
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| | - Yue-Xuan Chen
- a The Department of Pathology , Shenzhen Sun Yet-Sen Cardiovascular Hospital , Shenzhen , PR China and
| |
Collapse
|
41
|
Judge DP. Why should cardiologists consider genetic testing for hypertrophic cardiomyopathy? JACC-HEART FAILURE 2014; 3:189-91. [PMID: 25543975 DOI: 10.1016/j.jchf.2014.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 09/18/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Daniel P Judge
- Center for Inherited Heart Disease, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
42
|
Rigo F, Seth PP, Bennett CF. Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 825:303-52. [PMID: 25201110 DOI: 10.1007/978-1-4939-1221-6_9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Before a messenger RNA (mRNA) is translated into a protein in the cytoplasm, its pre-mRNA precursor is extensively processed through capping, splicing and polyadenylation in the nucleus. Defects in the processing of pre-mRNAs due to mutations in RNA sequences often cause disease. Traditional small molecules or protein-based therapeutics are not well suited for correcting processing defects by targeting RNA. However, antisense oligonucleotides (ASOs) designed to bind RNA by Watson-Crick base pairing can target most RNA transcripts and have emerged as the ideal therapeutic agents for diseases that are caused by pre-mRNA processing defects. Here we review the diverse ASO-based mechanisms that can be exploited to modulate the expression of RNA. We also discuss how advancements in medicinal chemistry and a deeper understanding of the pharmacokinetic and toxicological properties of ASOs have enabled their use as therapeutic agents. We end by describing how ASOs have been used successfully to treat various pre-mRNA processing diseases in animal models.
Collapse
Affiliation(s)
- Frank Rigo
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, USA,
| | | | | |
Collapse
|
43
|
Gaudet D, Brisson D, Tremblay K, Alexander VJ, Singleton W, Hughes SG, Geary RS, Baker BF, Graham MJ, Crooke RM, Witztum JL. Targeting APOC3 in the familial chylomicronemia syndrome. N Engl J Med 2014; 371:2200-6. [PMID: 25470695 DOI: 10.1056/nejmoa1400284] [Citation(s) in RCA: 356] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The familial chylomicronemia syndrome is a genetic disorder characterized by severe hypertriglyceridemia and recurrent pancreatitis due to a deficiency in lipoprotein lipase (LPL). Currently, there are no effective therapies except for extreme restriction in the consumption of dietary fat. Apolipoprotein C-III (APOC3) is known to inhibit LPL, although there is also evidence that APOC3 increases the level of plasma triglycerides through an LPL-independent mechanism. We administered an inhibitor of APOC3 messenger RNA (mRNA), called ISIS 304801, to treat three patients with the familial chylomicronemia syndrome and triglyceride levels ranging from 1406 to 2083 mg per deciliter (15.9 to 23.5 mmol per liter). After 13 weeks of study-drug administration, plasma APOC3 levels were reduced by 71 to 90% and triglyceride levels by 56 to 86%. During the study, all patients had a triglyceride level of less than 500 mg per deciliter (5.7 mmol per liter) with treatment. These data support the role of APOC3 as a key regulator of LPL-independent pathways of triglyceride metabolism.
Collapse
Affiliation(s)
- Daniel Gaudet
- From the ECOGENE-21 Clinical Research Center, Chicoutimi Hospital, Chicoutimi, and the Department of Medicine, Université de Montréal, Montreal - both in Canada (D.G., D.B., K.T.); and Isis Pharmaceuticals, Carlsbad (V.J.A., W.S., S.G.H., R.S.G., B.F.B., M.J.G., R.M.C.), and the Department of Medicine, Division of Endocrinology-Metabolism, University California, San Diego, School of Medicine, La Jolla (J.L.W.) - both in California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pharmacokinetic-pharmacodynamic modeling for reduction of hepatic apolipoprotein B mRNA and plasma total cholesterol after administration of antisense oligonucleotide in mice. J Pharmacokinet Pharmacodyn 2014; 42:67-77. [PMID: 25376372 DOI: 10.1007/s10928-014-9398-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 11/01/2014] [Indexed: 10/24/2022]
Abstract
Second-generation antisense oligonucleotides (ASOs) demonstrate excellent biological stability and in vitro/in vivo potency, and thus are considered to be attractive candidates for drugs to treat various diseases. A pharmacokinetic-pharmacodynamic (PK-PD) model of ASOs is desired for the design of appropriate PK and pharmacological studies. The objective of this study was to develop a PK-PD model to accurately simulate hepatic ASO concentration and its efficacy from plasma ASO concentration. After single subcutaneous administration of an ASO targeting hepatic apolipoprotein B (Apo-B) mRNA to mice, the ASO was absorbed rapidly and showed biphasic decline with time from the plasma and liver (t1/2: 1-3 and 81-183 h, Tmax: 0.25-0.50 and 4-8 h). After administration, hepatic Apo-B mRNA and plasma total cholesterol began decreasing at 4-8 and 8-24 h, and their Tmax values were observed at 24-72 and 72 h. To develop the PK-PD model based on the mechanisms of ASOs, we described the plasma and hepatic ASO concentration with linear two-compartment models. In addition, we inserted two indirect response models for mRNA and plasma total cholesterol. Model predictions from plasma ASO concentration gave excellent fits to the observed values of hepatic ASO concentration, Apo-B mRNA and plasma total cholesterol after single or multiple subcutaneous administrations. Our PK-PD model could accurately predict hepatic ASO concentrations and their efficacies from plasma ASO concentrations. This PK-PD model could be a useful tool for suggesting PK and pharmacological study protocols for various liver-targeted second-generation ASOs.
Collapse
|
45
|
Mitsuoka Y, Fujimura Y, Waki R, Kugimiya A, Yamamoto T, Hari Y, Obika S. Sulfonamide-bridged nucleic acid: synthesis, high RNA selective hybridization, and high nuclease resistance. Org Lett 2014; 16:5640-3. [PMID: 25341881 DOI: 10.1021/ol503029v] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
2'-N,4'-C-(N-methylamino)sulfonylmethylene-bridged thymidine (SuNA), which has a six-membered linkage including a sulfonamide moiety, was synthesized and introduced into oligonucleotides. The oligonucleotides containing SuNA exhibited excellent nuclease resistance, a high affinity toward single-stranded RNA, and a low affinity toward single-stranded DNA compared to the natural oligonucleotide.
Collapse
Affiliation(s)
- Yasunori Mitsuoka
- Graduate School of Pharmaceutical Sciences, Osaka University , 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Abendroth F, Seitz O. Double-Clicking Peptides onto Phosphorothioate Oligonucleotides: Combining Two Proapoptotic Agents in One Molecule. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201406674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
47
|
Abendroth F, Seitz O. Double-clicking peptides onto phosphorothioate oligonucleotides: combining two proapoptotic agents in one molecule. Angew Chem Int Ed Engl 2014; 53:10504-9. [PMID: 25138283 DOI: 10.1002/anie.201406674] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Indexed: 11/05/2022]
Abstract
Described here is a method for the conjugation of phosphorothioate oligonucleotides (PSOs) with peptides. PSOs are key to antisense technology. Peptide-PSO conjugates may improve target specificity, tissue distribution, and cellular uptake of PSOs. However, the highly nucleophilic phosphorothioate structure poses a challenge to conjugation chemistry. Herein, we introduce a new method which involves a sequence of oxime ligation and strain-promoted [2+3] cycloaddition. The usefulness of the method was demonstrated in the synthesis of peptide-PSO conjugates that targeted two suppressors of both the intrinsic and the extrinsic pathway of apoptosis. It is shown that the activity of a PSO sequence targeted against mRNA from c-Flip can be enhanced by conjugation with a peptide mimetic designed to inhibit the X-linked inhibitor of apoptosis protein (XIAP).
Collapse
Affiliation(s)
- Frank Abendroth
- Department of Chemistry, Humboldt University Berlin, Brook-Taylor-Strasse 2, 12489 Berlin (Germany)
| | | |
Collapse
|
48
|
Vitiello M, Tuccoli A, Poliseno L. Long non-coding RNAs in cancer: implications for personalized therapy. Cell Oncol (Dordr) 2014; 38:17-28. [PMID: 25113790 DOI: 10.1007/s13402-014-0180-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2014] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs, pseudogenes and circRNAs) have recently come into light as powerful players in cancer pathogenesis and it is becoming increasingly clear that they have the potential of greatly contributing to the spread and success of personalized cancer medicine. In this concise review, we briefly introduce these three classes of long non-coding RNAs. We then discuss their applications as diagnostic and prognostic biomarkers. Finally, we describe their appeal as targets and as drugs, while pointing out the limitations that still lie ahead of their definitive entry into clinical practice.
Collapse
Affiliation(s)
- Marianna Vitiello
- Oncogenomics Unit, Core Research Laboratory, Istituto Toscano Tumori c/o IFC-CNR, via Moruzzi 1, 56124, Pisa, Italy
| | | | | |
Collapse
|
49
|
George M, Selvarajan S, Muthukumar R, Elangovan S. Looking into the Crystal Ball—Upcoming Drugs for Dyslipidemia. J Cardiovasc Pharmacol Ther 2014; 20:11-20. [DOI: 10.1177/1074248414545127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dyslipidaemia is a critical risk factor for the development of cardiovascular complications such as ischemic heart disease and stroke. Although statins are effective anti-dyslipidemic drugs, their usage is fraught with issues such as failure of adequate lipid control in 30% of cases and intolerance in select patients. The limited potential of other alternatives such as fibrates, bile acid sequestrants and niacin has spurred the search for novel drug molecules with better efficacy and safety. CETP inhibitors such as evacetrapib and anacetrapib have shown promise in raising HDL besides LDL lowering property. Microsomal triglyceride transfer protein (MTP) inhibitors such as lomitapide and Apo CIII inhibitors such as mipomersen have recently been approved in Familial Hypercholesterolemia but experience in the non-familial setting is pretty much limited. One of the novel anti-dyslipidemic drugs which is greatly anticipated to make a mark in LDL-C control is the PCSK9 inhibitors. Some of the anti-dyslipidemic drugs which work by PCSK9 inhibition include evolocumab, alirocumab and ALN-PCS. Other approaches that are being given due consideration include farnesoid X receptor modulation and Lp-PLA2 inhibition. While it may not be an easy proposition to dismantle statins from their current position as a cholesterol reducing agent and as a drug to reduce coronary and cerebro-vascular atherosclerosis, our improved understanding of the disease and appropriate harnessing of resources using sound and robust technology could make rapid in-roads in our pursuit of the ideal anti-dyslipidemic drug.
Collapse
Affiliation(s)
- Melvin George
- Department of Cardiology, SRM Medical College Hospital & Research Centre, Kattankulathur, Kancheepuram, Chennai, India
| | - Sandhiya Selvarajan
- Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, India
| | - Rajaram Muthukumar
- Department of Cardiology, SRM Medical College Hospital & Research Centre, Kattankulathur, Kancheepuram, Chennai, India
| | - Shanmugam Elangovan
- Department of Cardiology, SRM Medical College Hospital & Research Centre, Kattankulathur, Kancheepuram, Chennai, India
| |
Collapse
|
50
|
Yu J, Pandey SK, Khatri H, Prakash TP, Swayze EE, Seth PP. Synthesis and antisense properties of 2'-O-(2S-methoxypropyl)-RNA-modified gapmer antisense oligonucleotides. ChemMedChem 2014; 9:2040-4. [PMID: 24891270 DOI: 10.1002/cmdc.201402099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Indexed: 11/06/2022]
Abstract
To ascertain whether increasing hydrophobicity can enhance the activity of second-generation antisense oligonucleotides (ASOs) in muscle, we investigated the antisense properties of 2'-O-(2S-methoxypropyl)-RNA (2S-MOP)-modified ASOs. Synthesis of the 2S-MOP 5-methyl uridine phosphoramidite was accomplished on a multi-gram scale by Lewis-acid-catalyzed ring opening of 5'-O-tert-butyldiphenylsilyl ether-protected 2,2'-anhydro-5-methyl uridine with 2S-methoxy-1-propanol. Synthesis of the 2S-MOP 5-methyl cytidine nucleoside from the corresponding 5-methyl uridine nucleoside was accomplished by formation and displacement of a 4-triazolide intermediate with aqueous ammonia. 2S-MOP-modified oligonucleotides were prepared on an automated DNA synthesizer and showed similar enhancements in duplex thermal stability as 2'-O-methoxyethyl RNA (MOE)-modified oligonucleotides. 2S-MOP-containing antisense oligonucleotides were evaluated in Balb-c mice and showed good activity for decreasing the expression levels of scavenger receptor B1 (Srb1) and phosphatase and tensin homologue (PTEN) mRNA in liver and muscle tissue.
Collapse
Affiliation(s)
- Jinghua Yu
- Isis Pharmaceuticals, Inc. 2855 Gazelle Court, Carlsbad, CA 92010 (USA)
| | | | | | | | | | | |
Collapse
|