201
|
deRonde BM, Posey ND, Otter R, Minter LM, Tew GN. Optimal Hydrophobicity in Ring-Opening Metathesis Polymerization-Based Protein Mimics Required for siRNA Internalization. Biomacromolecules 2016; 17:1969-77. [PMID: 27103189 PMCID: PMC4964964 DOI: 10.1021/acs.biomac.6b00138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Exploring the role of polymer structure for the internalization of biologically relevant cargo, specifically siRNA, is of critical importance to the development of improved delivery reagents. Herein, we report guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold containing tunable hydrophobic moieties that promote siRNA internalization. Structure-activity relationships using Jurkat T cells and HeLa cells were explored to determine how the length of the hydrophobic block and the hydrophobic side chain compositions of these PTDMs impacted siRNA internalization. To explore the hydrophobic block length, two different series of diblock copolymers were synthesized: one series with symmetric block lengths and one with asymmetric block lengths. At similar cationic block lengths, asymmetric and symmetric PTDMs promoted siRNA internalization in the same percentages of the cell population regardless of the hydrophobic block length; however, with 20 repeat units of cationic charge, the asymmetric block length had greater siRNA internalization, highlighting the nontrivial relationships between hydrophobicity and overall cationic charge. To further probe how the hydrophobic side chains impacted siRNA internalization, an additional series of asymmetric PTDMs was synthesized that featured a fixed hydrophobic block length of five repeat units that contained either dimethyl (dMe), methyl phenyl (MePh), or diphenyl (dPh) side chains and varied cationic block lengths. This series was further expanded to incorporate hydrophobic blocks consisting of diethyl (dEt), diisobutyl (diBu), and dicyclohexyl (dCy) based repeat units to better define the hydrophobic window for which our PTDMs had optimal activity. High-performance liquid chromatography retention times quantified the relative hydrophobicities of the noncationic building blocks. PTDMs containing the MePh, diBu, and dPh hydrophobic blocks were shown to have superior siRNA internalization capabilities compared to their more and less hydrophobic counterparts, demonstrating a critical window of relative hydrophobicity for optimal internalization. This better understanding of how hydrophobicity impacts PTDM-induced internalization efficiencies will help guide the development of future delivery reagents.
Collapse
Affiliation(s)
- Brittany M. deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
| | - Nicholas D. Posey
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
| | - Ronja Otter
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| | - Lisa M. Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| | - Gregory N. Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
202
|
Witzigmann D, Quagliata L, Schenk SH, Quintavalle C, Terracciano LM, Huwyler J. Variable asialoglycoprotein receptor 1 expression in liver disease: Implications for therapeutic intervention. Hepatol Res 2016; 46:686-96. [PMID: 26422581 DOI: 10.1111/hepr.12599] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/07/2015] [Accepted: 09/19/2015] [Indexed: 02/08/2023]
Abstract
AIM One of the most promising strategies for the treatment of liver diseases is targeted drug delivery via the asialoglycoprotein receptor (ASGPR). The success of this approach heavily depends on the ASGPR expression level on parenchymal liver cells. In this study, we assessed the mRNA and protein expression levels of the major receptor subunit, ASGR1, in hepatocytes both in vitro and in vivo. METHODS In vitro, various liver cancer-derived cell lines were evaluated. In vivo, we screened the ASGR1 mRNA on 59 hepatocellular carcinoma and matched non-neoplastic tissue using RNA microarray. In addition, 350 human liver specimens of patients with hepatocellular carcinoma or non-neoplastic liver diseases were screened for ASGR1 protein level using tissue microarray analysis. RESULTS Our data reveal that the ASGR1 mRNA expression directly correlates with the protein level. We demonstrate that the ASGR1 expression is upregulated in cirrhotic specimens and is significantly decreased with increasing hepatocellular carcinoma grade. CONCLUSION Because the ASGR1 expression levels are variable between patients, our findings suggest that ASGPR-based targeting strategies should be combined with ASGPR-companion diagnostics to maximize clinical benefit.
Collapse
Affiliation(s)
- Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, Basel, Switzerland
| | - Susanne H Schenk
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Cristina Quintavalle
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, Basel, Switzerland
| | - Luigi M Terracciano
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, Basel, Switzerland
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
203
|
Yang Y, Yang Y, Xie X, Xu X, Xia X, Wang H, Li L, Dong W, Ma P, Liu Y. Dual stimulus of hyperthermia and intracellular redox environment triggered release of siRNA for tumor-specific therapy. Int J Pharm 2016; 506:158-73. [DOI: 10.1016/j.ijpharm.2016.04.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/31/2016] [Accepted: 04/15/2016] [Indexed: 01/06/2023]
|
204
|
Müller K, Kessel E, Klein PM, Höhn M, Wagner E. Post-PEGylation of siRNA Lipo-oligoamino Amide Polyplexes Using Tetra-glutamylated Folic Acid as Ligand for Receptor-Targeted Delivery. Mol Pharm 2016; 13:2332-45. [DOI: 10.1021/acs.molpharmaceut.6b00102] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Katharina Müller
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Eva Kessel
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse 4, D-80799 Munich, Germany
| | - Philipp M. Klein
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research, Ludwig-Maximilians-Universität München, Butenandtstrasse 5-13, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse 4, D-80799 Munich, Germany
| |
Collapse
|
205
|
Amsalem O, Nassar T, Benhamron S, Lazarovici P, Benita S, Yavin E. Solid nano-in-nanoparticles for potential delivery of siRNA. J Control Release 2016; 257:144-155. [PMID: 27215702 DOI: 10.1016/j.jconrel.2016.05.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/24/2016] [Accepted: 05/16/2016] [Indexed: 12/22/2022]
Abstract
siRNA-based therapeutics possess great potential to treat a wide variety of genetic disorders. However, they suffer from low cellular uptake and short half-lives in blood circulation; issues that remain to be addressed. This work is, to the best of our knowledge, the first to report the production of solid nano-in-nanoparticles, termed double nano carriers (DNCs) by means of the innovative technology of nano spray drying. DNCs (with a median size of 580-770nm) were produced by spraying at low temperatures (50°C) to prevent damage to heat-sensitive biomacromolecules like siRNA. DNCs consisting of Poly (d,l-lactide-co-glycolide) used as a wall material, encapsulating 20% human serum albumin primary nanoparticles (PNPs) loaded with siRNA, were obtained as a dry nanoparticulate powder with smooth spherical surfaces and a unique inner morphology. Incubation of pegylated or non-pegylated DNCs under sink conditions at 37°C, elicited a controlled release profile of the siRNA for up to 12 or 24h, respectively, with a minimal burst effect. Prolonged incubation of pegylated DNCs loaded with active siRNA (anti EGFR) in an A549 epithelial cell culture monolayer did not induce any apparent cytotoxicity. A slow degradation of the internalized DNCs by the cells was also observed resulting in the progressive release of the siRNA for up to 6days, as corroborated by laser confocal microscopy. The structural integrity and silencing activity of the double encapsulated siRNA were fully preserved, as demonstrated by HPLC, gel electrophoresis, and potent RNAi activity of siRNA extracted from DNCs. These results demonstrate the potential use of DNCs as a nano drug delivery system for systemic administration and controlled release of siRNA and potentially other sensitive bioactive macromolecules.
Collapse
Affiliation(s)
- Orit Amsalem
- The Institute for Drug Research of the School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Taher Nassar
- The Institute for Drug Research of the School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Sandrine Benhamron
- The Institute for Drug Research of the School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Philip Lazarovici
- The Institute for Drug Research of the School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Simon Benita
- The Institute for Drug Research of the School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel.
| | - Eylon Yavin
- The Institute for Drug Research of the School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
206
|
Fragment-based solid-phase assembly of oligonucleotide conjugates with peptide and polyethylene glycol ligands. Eur J Med Chem 2016; 121:132-142. [PMID: 27236069 DOI: 10.1016/j.ejmech.2016.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/25/2016] [Accepted: 05/01/2016] [Indexed: 12/26/2022]
Abstract
Ligand conjugation to oligonucleotides is an attractive strategy for enhancing the therapeutic potential of antisense and siRNA agents by inferring properties such as improved cellular uptake or better pharmacokinetic properties. Disulfide linkages enable dissociation of ligands and oligonucleotides in reducing environments found in endosomal compartments after cellular uptake. Solution-phase fragment coupling procedures for producing oligonucleotide conjugates are often tedious, produce moderate yields and reaction byproducts are frequently difficult to remove. We have developed an improved method for solid-phase coupling of ligands to oligonucleotides via disulfides directly after solid-phase synthesis. A 2'-thiol introduced using a modified nucleotide building block was orthogonally deprotected on the controlled pore glass solid support with N-butylphosphine. Oligolysine peptides and a short monodisperse ethylene glycol chain were successfully coupled to the deprotected thiol. Cleavage from the resin and full removal of oligonucleotide protection groups were achieved using methanolic ammonia. After standard desalting, and without further purification, homogenous conjugates were obtained as demonstrated by HPLC, gel electrophoresis, and mass spectrometry. The attachment of both amphiphilic and cationic ligands proves the versatility of the conjugation procedure. An antisense oligonucleotide conjugate with hexalysine showed pronounced gene silencing in a cell culture tumor model in the absence of a transfection reagent and the corresponding ethylene glycol conjugate resulted in down regulation of the target gene to nearly 50% after naked application.
Collapse
|
207
|
Conjugates of small targeting molecules to non-viral vectors for the mediation of siRNA. Acta Biomater 2016; 36:21-41. [PMID: 27045350 DOI: 10.1016/j.actbio.2016.03.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
Abstract
UNLABELLED To use siRNA (small interfering RNA) for gene therapy, a gene delivery system is often necessary to overcome several challenging requirements including rapid excretion, low stability in blood serum, non-specific accumulation in tissues, poor cellular uptake and inefficient intracellular release. Active and/or passive targeting should help the delivery system to reach the desired tissue or cell, to be internalized, and to deliver siRNA to the cytoplasm so that siRNA can inhibit protein synthesis. This review covers conjugates of small targeting molecules and non-viral delivery systems for the mediation of siRNA, with a focus on their transfection properties in order to help the development of new and efficient siRNA delivery systems, as the therapeutic solutions of tomorrow. STATEMENT OF SIGNIFICANCE The delivery of siRNA into cells or tissues remains to be a challenge for its applications, an alternative strategy for siRNA delivery systems is direct conjugation of non-viral vectors with targeting moieties for cellular delivery. In comparison to macromolecules, small targeting molecules have attracted great attention due to their many potential advantages including significant simplicity and ease of production, good repeatability and biodegradability. This review will focus on the most recent advances in the delivery of siRNA using conjugates of small targeting molecules and non-viral delivery systems. Based the editor's suggestions, we hope the revised manuscript could provide more profound understanding to the conjugates of targeting molecules to vectors for mediation of siRNA.
Collapse
|
208
|
Yang J, Li Y, Zhang T, Zhang X. Development of bioactive materials for glioblastoma therapy. Bioact Mater 2016; 1:29-38. [PMID: 29744393 PMCID: PMC5883963 DOI: 10.1016/j.bioactmat.2016.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma is the most common and deadly human brain cancers. Unique barriers hinder the drug delivering pathway due to the individual position of glioblastoma, including blood-brain barrier and blood-brain tumor barrier. Numerous bioactive materials have been exploited and applied as the transvascular delivery carriers of therapeutic drugs. They promote site-specific accumulation and long term release of the encapsulated drugs at the tumor sites and reduce side effects with systemic delivery. And the delivery systems exhibit a certain extent of anti-glioblastoma effect and extend the median survival time. However, few of them step into the clinical trials. In this review, we will investigate the recent studies of bioactive materials for glioblastoma chemotherapy, including the inorganic materials, lipids and polymers. These bioactive materials construct diverse delivery vehicles to trigger tumor sites in brain intravenously. Herein, we exploit their functionality in drug delivery and discuss the deficiency for the featured tumors, to provide guidance for establishing optimized therapeutic drug formulation for anti-glioblastoma therapy and pave the way for clinical application. Numerous bioactive materials have been exploited as delivery carriers of therapeutic drugs for glioblastoma chemotherapy. The functionality and deficiency of the bioactive materials are discussed. Combing the chemo- and immunotherapy will provide a promising strategy for glioblastoma therapy and inhibiting recurrence.
Collapse
Key Words
- ALA, α-lipoic acid
- BAG3, Bcl-2 associated athanogene 3
- BBB, blood-brain barrier
- BTB, blood-brain tumor barrier
- Bioactive material
- Blood-brain barrier
- Blood-brain tumor barrier
- CNS, central nervous system
- CPT, camptothecin
- Chemotherapy
- DACHPt, dichloro-(1,2-diaminocyclohexane)platinum (II)
- DCs, dendritic cells
- DHA, dehydroascorbic acid
- DOX, doxorubicin
- DPPC, 1,2-dihexadecanoyl-rac-glycero-3-phosphocholine
- FA, folate
- GCV, ganciclovir
- GLUT1, glucose transporter isoform 1
- Glioblastoma
- IL, interleukin
- MMPs, matrix metalloproteinases
- PTX, paclitaxel
- ROS, reactive oxygen species
- SN38, 7-ethyl-10-hydroxy-camptothecin
- TAT, transactivator of transcription
- TEG, tetra(ethylene glycol)
- TMZ, temozolomide
- TNF, tumor necrosis factor
- TfR, transferrin receptor
- cRGD, cyclic Arg-Gly-Asp
Collapse
Affiliation(s)
- Jun Yang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yan Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Tianlu Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xin Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
209
|
Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Acids Res 2016; 44:6518-48. [PMID: 27084936 PMCID: PMC5001581 DOI: 10.1093/nar/gkw236] [Citation(s) in RCA: 616] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/28/2016] [Indexed: 12/14/2022] Open
Abstract
The oligonucleotide therapeutics field has seen remarkable progress over the last few years with the approval of the first antisense drug and with promising developments in late stage clinical trials using siRNA or splice switching oligonucleotides. However, effective delivery of oligonucleotides to their intracellular sites of action remains a major issue. This review will describe the biological basis of oligonucleotide delivery including the nature of various tissue barriers and the mechanisms of cellular uptake and intracellular trafficking of oligonucleotides. It will then examine a variety of current approaches for enhancing the delivery of oligonucleotides. This includes molecular scale targeted ligand-oligonucleotide conjugates, lipid- and polymer-based nanoparticles, antibody conjugates and small molecules that improve oligonucleotide delivery. The merits and liabilities of these approaches will be discussed in the context of the underlying basic biology.
Collapse
Affiliation(s)
- Rudolph L Juliano
- UNC Eshelman School of Pharmacy and UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
210
|
Moon JS, Lee SH, Han SH, Kim EJ, Cho H, Lee W, Kim MK, Kim TE, Park HJ, Rhee JK, Kim SJ, Cho SW, Han SH, Oh JW. Inhibition of hepatitis C virus in mouse models by lipidoid nanoparticle-mediated systemic delivery of siRNA against PRK2. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1489-98. [PMID: 27013134 DOI: 10.1016/j.nano.2016.02.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 02/10/2016] [Accepted: 02/15/2016] [Indexed: 12/12/2022]
Abstract
Host-targeting antivirals have an advantage over direct-acting antivirals in that they have a high genetic barrier to resistance. Here, we describe in vivo anti-hepatitis C virus (HCV) efficacy of a potent siRNA targeting the protein kinase C-related kinase 2 (PRK2), which phosphorylates HCV NS5B RNA-dependent RNA polymerase and promotes HCV replication. PRK2-silencing reduced the phosphorylated NS5B level and resulted in inhibition of NS5B RdRp activity to decrease HCV genome abundance. Systemic administration of lipidoid nanoparticle-formulated PRK2 siRNA (once every three days for a total of three injections at a dose of 3mgkg(-1)) resulted in a 3.72 and 1.96 log10 reduction in serum HCV RNA titer, in mouse subcutaneous and orthotopic xenograft models for HCV replication, respectively. Our results verify the essential role of PRK2 in HCV replication and offer a host-targeting anti-HCV siRNA therapy that might be beneficial for non-responders to current treatment regimens.
Collapse
Affiliation(s)
- Jae-Su Moon
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Seung-Hoon Lee
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Song-Hee Han
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Eun-Jung Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Hee Cho
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Wooseong Lee
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Mi-Kyung Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Tae-Eun Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Hyun-Ji Park
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Jin-Kyu Rhee
- Western Seoul Center of Korea Basic Science Institute, Seoul, Korea
| | - Seong-Jun Kim
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Jong-Won Oh
- Department of Biotechnology, Yonsei University, Seoul, Korea.
| |
Collapse
|
211
|
Conceição M, Mendonça L, Nóbrega C, Gomes C, Costa P, Hirai H, Moreira JN, Lima MC, Manjunath N, Pereira de Almeida L. Intravenous administration of brain-targeted stable nucleic acid lipid particles alleviates Machado-Joseph disease neurological phenotype. Biomaterials 2016; 82:124-37. [DOI: 10.1016/j.biomaterials.2015.12.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/13/2015] [Accepted: 12/16/2015] [Indexed: 12/25/2022]
|
212
|
Ngamcherdtrakul W, Castro DJ, Gu S, Morry J, Reda M, Gray JW, Yantasee W. Current development of targeted oligonucleotide-based cancer therapies: Perspective on HER2-positive breast cancer treatment. Cancer Treat Rev 2016; 45:19-29. [PMID: 26930249 DOI: 10.1016/j.ctrv.2016.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/13/2016] [Accepted: 02/15/2016] [Indexed: 12/24/2022]
Abstract
This Review discusses the various types of non-coding oligonucleotides, which have garnered extensive interest as new alternatives for targeted cancer therapies over small molecule inhibitors and monoclonal antibodies. These oligonucleotides can target any hallmark of cancer, no longer limited to so-called "druggable" targets. Thus, any identified gene that plays a key role in cancer progression or drug resistance can be exploited with oligonucleotides. Among them, small-interfering RNAs (siRNAs) are frequently utilized for gene silencing due to the robust and well established mechanism of RNA interference. Despite promising advantages, clinical translation of siRNAs is hindered by the lack of effective delivery platforms. This Review provides general criteria and consideration of nanoparticle development for systemic siRNA delivery. Different classes of nanoparticle candidates for siRNA delivery are discussed, and the progress in clinical trials for systemic cancer treatment is reviewed. Lastly, this Review presents HER2 (human epidermal growth factor receptor type 2)-positive breast cancer as one example that could benefit significantly from siRNA technology. How siRNA-based therapeutics can overcome cancer resistance to such therapies is discussed.
Collapse
Affiliation(s)
- Worapol Ngamcherdtrakul
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - David J Castro
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Shenda Gu
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Jingga Morry
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA.
| | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA.
| |
Collapse
|
213
|
Silencing CCR2 in Macrophages Alleviates Adipose Tissue Inflammation and the Associated Metabolic Syndrome in Dietary Obese Mice. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e280. [PMID: 26812653 PMCID: PMC5012549 DOI: 10.1038/mtna.2015.51] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/23/2015] [Indexed: 12/17/2022]
Abstract
Adipose tissue macrophage (ATM)-mediated inflammation is a key feature contributing to the adverse metabolic outcomes of dietary obesity. Recruitment of macrophages to obese adipose tissues (AT) can occur through the engagement of CCR2, the receptor for MCP-1 (monocyte chemoattractant protein-1), which is expressed on peripheral monocytes/macrophages. Here, we show that i.p. administration of a rabies virus glycoprotein-derived acetylcholine receptor-binding peptide effectively delivers complexed siRNA into peritoneal macrophages and ATMs in a mouse model of high-fat diet-induced obesity. Treatment with siRNA against CCR2 inhibited macrophage infiltration and accumulation in AT and, therefore, proinflammatory cytokines produced by macrophages. Consequently, the treatment significantly improved glucose tolerance and insulin sensitivity profiles, and also alleviated the associated symptoms of hepatic steatosis and reduced hepatic triglyceride production. These results demonstrate that disruption of macrophage chemotaxis to the AT through cell-targeted gene knockdown strategies can provide a therapeutic intervention for obesity-related metabolic diseases. The study also highlights a siRNA delivery approach for targeting specific monocyte subsets that contribute to obesity-associated inflammation without affecting the function of other tissue-resident macrophages that are essential for host homeostasis and survival.
Collapse
|
214
|
Simion V, Stan D, Constantinescu CA, Deleanu M, Dragan E, Tucureanu MM, Gan AM, Butoi E, Constantin A, Manduteanu I, Simionescu M, Calin M. Conjugation of curcumin-loaded lipid nanoemulsions with cell-penetrating peptides increases their cellular uptake and enhances the anti-inflammatory effects in endothelial cells. ACTA ACUST UNITED AC 2016; 68:195-207. [PMID: 26748549 DOI: 10.1111/jphp.12513] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/29/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVES To prepare and characterize in vitro and in vivo lipid nanoemulsions (LN) loaded with curcumin (Cm) and functionalized with a cell-penetrating peptide. METHODS Curcumin-loaded lipid nanoemulsions (CmLN) functionalized with a nona-arginine peptide (R9-CmLN) have been obtained, characterized and optimized for size, entrapment efficiency and in vitro Cm release. The interaction of R9-CmLN with human endothelial cells (HEC) was investigated using cultured EA.hy926 cells, and in vivo biodistribution studies were performed using C57BL6 mice. KEY FINDINGS When used in therapeutically relevant concentration, R9-CmLN have low haemolytic activity, low cytotoxicity on HEC, and show anti-inflammatory effects by reducing the monocytes adhesion to TNF-α activated HEC. Moreover, HEC uptake and internalization of R9-CmLN was significantly higher compared to the non-functionalized CmLN. In vivo biodistribution studies in mice revealed a higher accumulation of R9-CmLN in the liver and the lungs compared to CmLN and the body clearance of the both nanoformulations after 72 h. CONCLUSIONS Cell-penetrating peptides-functionalized CmLN have superior characteristics compared to their non-functionalized counterparts: are more efficiently internalized by the cells, produces anti-inflammatory effects in HEC and when administrated intravenously in mice exhibit increased accumulation in the liver and the lungs, suggesting their potential therapeutic applications in different inflammatory pathologies localized in the liver or the lungs.
Collapse
Affiliation(s)
- Viorel Simion
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Daniela Stan
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Cristina Ana Constantinescu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.,Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| | - Mariana Deleanu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.,Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| | - Emanuel Dragan
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Monica Madalina Tucureanu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Ana-Maria Gan
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Elena Butoi
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Alina Constantin
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Ileana Manduteanu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Manuela Calin
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
215
|
Affiliation(s)
- Mark W. Tibbitt
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department
of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - James E. Dahlman
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Robert Langer
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department
of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Harvard-MIT
Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
216
|
Tay J, Tiao J, Hughes Q, Gilmore G, Baker R. Therapeutic Potential of miR-494 in Thrombosis and Other Diseases: A Review. Aust J Chem 2016. [DOI: 10.1071/ch16020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Functional nucleic acids, such as microRNAs (miRNAs), have been implicated in the pathophysiology of many diseases. The miRNA expression profiles of various cancers including haematological malignancies are well defined, but the role of miRNAs in haemostasis and the regulation of coagulation is poorly understood. We identified that miR-494 is oestrogen responsive and directly targets the anticoagulant protein, Protein S, as a mechanism for acquiring Protein S deficiency under high oestrogenic conditions such as during pregnancy and oral contraceptive use. Furthermore, previous studies have also characterised miR-494 to be involved in many biological processes. This paper reviews the current knowledge in the role of miRNAs in regulating haemostatic proteins and the known biological functions of miR-494, highlighting miR-494 as an emerging therapeutic target, with an overview of the strategy we have employed in identifying functional nucleic acids such as miRNAs that target haemostatic factors and the therapeutic potential of miR-494-directed therapy for the treatment of thrombotic disorders.
Collapse
|
217
|
Synthesis of multifunctional bovine serum albumin microcapsules by the sonochemical method for targeted drug delivery and controlled drug release. Colloids Surf B Biointerfaces 2015; 136:470-8. [DOI: 10.1016/j.colsurfb.2015.09.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/02/2015] [Accepted: 09/27/2015] [Indexed: 02/07/2023]
|
218
|
Liu Y, Kang Y, Wang J, Wang Z, Chen G, Jiang M. Sequence-Defined Peptidocopolymers: The Effect of Small Molecular Linkers. Biomacromolecules 2015; 16:3995-4003. [DOI: 10.1021/acs.biomac.5b01348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yijiang Liu
- The
State Key Laboratory of Molecular Engineering of Polymers and Department
of Macromolecular Science, Fudan University, Shanghai, 200433 China
| | - Yu Kang
- College
of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, People’s Republic of China
| | - Jue Wang
- The
State Key Laboratory of Molecular Engineering of Polymers and Department
of Macromolecular Science, Fudan University, Shanghai, 200433 China
| | - Zheyu Wang
- The
State Key Laboratory of Molecular Engineering of Polymers and Department
of Macromolecular Science, Fudan University, Shanghai, 200433 China
| | - Guosong Chen
- The
State Key Laboratory of Molecular Engineering of Polymers and Department
of Macromolecular Science, Fudan University, Shanghai, 200433 China
| | - Ming Jiang
- The
State Key Laboratory of Molecular Engineering of Polymers and Department
of Macromolecular Science, Fudan University, Shanghai, 200433 China
| |
Collapse
|
219
|
Kaspar RL, Hickerson RP, González-González E, Flores MA, Speaker TP, Rogers FA, Milstone LM, Contag CH. Imaging Functional Nucleic Acid Delivery to Skin. Methods Mol Biol 2015; 1372:1-24. [PMID: 26530911 DOI: 10.1007/978-1-4939-3148-4_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Monogenic skin diseases arise from well-defined single gene mutations, and in some cases a single point mutation. As the target cells are superficial, these diseases are ideally suited for treatment by nucleic acid-based therapies as well as monitoring through a variety of noninvasive imaging technologies. Despite the accessibility of the skin, there remain formidable barriers for functional delivery of nucleic acids to the target cells within the dermis and epidermis. These barriers include the stratum corneum and the layered structure of the skin, as well as more locally, the cellular, endosomal and nuclear membranes. A wide range of technologies for traversing these barriers has been described and moderate success has been reported for several approaches. The lessons learned from these studies include the need for combinations of approaches to facilitate nucleic acid delivery across these skin barriers and then functional delivery across the cellular and nuclear membranes for expression (e.g., reporter genes, DNA oligonucleotides or shRNA) or into the cytoplasm for regulation (e.g., siRNA, miRNA, antisense oligos). The tools for topical delivery that have been evaluated include chemical, physical and electrical methods, and the development and testing of each of these approaches has been greatly enabled by imaging tools. These techniques allow delivery and real time monitoring of reporter genes, therapeutic nucleic acids and also triplex nucleic acids for gene editing. Optical imaging is comprised of a number of modalities based on properties of light-tissue interaction (e.g., scattering, autofluorescence, and reflectance), the interaction of light with specific molecules (e.g., absorbtion, fluorescence), or enzymatic reactions that produce light (bioluminescence). Optical imaging technologies operate over a range of scales from macroscopic to microscopic and if necessary, nanoscopic, and thus can be used to assess nucleic acid delivery to organs, regions, cells and even subcellular structures. Here we describe the animal models, reporter genes, imaging approaches and general strategies for delivery of nucleic acids to cells in the skin for local expression (e.g., plasmid DNA) or gene silencing (e.g., siRNA) with the intent of developing nucleic acid-based therapies to treat diseases of the skin.
Collapse
Affiliation(s)
- Roger L Kaspar
- TransDerm Inc., 2161 Delaware Ave, Santa Cruz, CA, 95060, USA.
| | - Robyn P Hickerson
- Centre for Dermatology and Genetic Medicine, University of Dundee, Dundee, UK
| | | | - Manuel A Flores
- TransDerm Inc., 2161 Delaware Ave, Santa Cruz, CA, 95060, USA
| | - Tycho P Speaker
- TransDerm Inc., 2161 Delaware Ave, Santa Cruz, CA, 95060, USA
| | - Faye A Rogers
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Leonard M Milstone
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher H Contag
- Molecular Imaging Program at Stanford (MIPS), E150 Clark Center, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA, 94305, USA. .,Department of Pediatrics, E150 Clark Center, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA, 94305, USA. .,Department of Radiology, E150 Clark Center, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA, 94305, USA. .,Microbiology and Immunology, E150 Clark Center, Stanford University School of Medicine, 318 Campus Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
220
|
Singh Y, Tomar S, Khan S, Meher JG, Pawar VK, Raval K, Sharma K, Singh PK, Chaurasia M, Surendar Reddy B, Chourasia MK. Bridging small interfering RNA with giant therapeutic outcomes using nanometric liposomes. J Control Release 2015; 220:368-387. [PMID: 26528900 DOI: 10.1016/j.jconrel.2015.10.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 01/04/2023]
Abstract
The scope of RNAi based therapeutics is unquestionable. However, if we dissect the current trend of clinical trials for afore mentioned drug class, some stark trends appear: 1) naked siRNA only exerts influence in topical mode whilst systemic delivery requires a carrier and 2) even after two decades of extensive efforts, not even a single siRNA containing product is commercially available. It was therefore felt that a perspective simplifying the unique intricacies of working with a merger of siRNA and liposomes from a pharmaceutical viewpoint could draw the attention of a wider array of interested researchers. We begin from the beginning and attempt to conduit the gap between theoretical logic and experimental/actual constraints. This, in turn could stimulate the next generation of investigators, gearing them to tackle the conundrum, which is siRNA delivery.
Collapse
Affiliation(s)
- Yuvraj Singh
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sandeep Tomar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shariq Khan
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jaya Gopal Meher
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Vivek K Pawar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kavit Raval
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Komal Sharma
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pankaj K Singh
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Mohini Chaurasia
- Amity Institute of Pharmacy, Amity University, Lucknow, UP 226028, India
| | - B Surendar Reddy
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Manish K Chourasia
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
221
|
Childs-Disney JL, Disney MD. Approaches to Validate and Manipulate RNA Targets with Small Molecules in Cells. Annu Rev Pharmacol Toxicol 2015; 56:123-40. [PMID: 26514201 DOI: 10.1146/annurev-pharmtox-010715-103910] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RNA has become an increasingly important target for therapeutic interventions and for chemical probes that dissect and manipulate its cellular function. Emerging targets include human RNAs that have been shown to directly cause cancer, metabolic disorders, and genetic disease. In this review, we describe various routes to obtain bioactive compounds that target RNA, with a particular emphasis on the development of small molecules. We use these cases to describe approaches that are being developed for target validation, which include target-directed cleavage, classic pull-down experiments, and covalent cross-linking. Thus, tools are available to design small molecules to target RNA and to identify the cellular RNAs that are their targets.
Collapse
Affiliation(s)
| | - Matthew D Disney
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458; ,
| |
Collapse
|
222
|
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
|
223
|
Therapeutic oligonucleotides with polyethylene glycol modifications. Future Med Chem 2015; 7:1721-31. [PMID: 26465713 DOI: 10.4155/fmc.15.94] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In the field of oligonucleotide drugs, the attachment of PEG is a well-established strategy to prevent enzymatic degradation and avoid renal elimination. Pegaptanib and other oligonucleotides in clinical development utilize the attachment of linear or branched high molecular weight PEG chains for increase of accumulation and duration of the effect after local or systemic application. The length of PEG chains is decisive for the pharmacokinetic and pharmacodynamic effects. Longer chains increase circulation times, but generally decrease gene-silencing efficiencies for antisense and siRNA agents and binding affinities for aptamers. Shorter chains are less efficient in preventing renal filtration, but have also less impact on the gene-silencing machinery and binding kinetics.
Collapse
|
224
|
Concise postsynthetic preparation of oligonucleotide-oligopeptide conjugates through facile disulfide bond formation. Future Med Chem 2015; 7:1657-73. [PMID: 26381134 DOI: 10.4155/fmc.15.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Despite recent advances, major hurdles still need to be cleared for widespread application of therapeutic antisense technologies. In particular, pharmacokinetic properties and efficient cellular uptake need to be improved through chemical derivatization or bioconjugation. RESULTS The 2'-O-thioethylene nucleotide building block affords easy implementation into standard oligonucleotide synthesis protocols and was used to attach oligolysine chains to phosphodiester oligonucleotides by direct reaction with S-sulfonate protected peptides. Efficient gene silencing was induced in a cell culture model after transfection reagent-free application of the conjugates. CONCLUSION A facile optimized procedure for generating oligonucleotide-peptide conjugates was established. The attachment of short basic peptides via a labile linker is sufficient to enhance membrane permeability of oligonucleotides and result in successful gene silencing.
Collapse
|
225
|
Liposomes as carriers: not as innocent as one would like. Arch Toxicol 2015; 89:1399-400. [PMID: 26245945 DOI: 10.1007/s00204-015-1574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
|
226
|
Lundin KE, Gissberg O, Smith CE. Oligonucleotide Therapies: The Past and the Present. Hum Gene Ther 2015; 26:475-85. [PMID: 26160334 PMCID: PMC4554547 DOI: 10.1089/hum.2015.070] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/04/2015] [Indexed: 12/19/2022] Open
Abstract
In this review we address the development of oligonucleotide (ON) medicines from a historical perspective by listing the landmark discoveries in this field. The various biological processes that have been targeted and the corresponding ON interventions found in the literature are discussed together with brief updates on some of the more recent developments. Most ON therapies act through antisense mechanisms and are directed against various RNA species, as exemplified by gapmers, steric block ONs, antagomirs, small interfering RNAs (siRNAs), micro-RNA mimics, and splice switching ONs. However, ONs binding to Toll-like receptors and those forming aptamers have completely different modes of action. Similar to other novel medicines, the path to success has been lined with numerous failures, where different therapeutic ONs did not stand the test of time. Since the first ON drug was approved for clinical use in 1998, the therapeutic landscape has changed considerably, but many challenges remain until the expectations for this new form of medicine are met. However, there is room for cautious optimism.
Collapse
Affiliation(s)
- Karin E. Lundin
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Olof Gissberg
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - C.I. Edvard Smith
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
227
|
Criscione F, O'Brochta DA, Reid W. Genetic technologies for disease vectors. CURRENT OPINION IN INSECT SCIENCE 2015; 10:90-97. [PMID: 29588019 DOI: 10.1016/j.cois.2015.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/17/2015] [Accepted: 04/20/2015] [Indexed: 06/08/2023]
Abstract
The first genetic technologies for insect vectors of disease were introduced 20 years ago. As of today there are 12 classes of genetic technologies used as functional genomic tools for insect vectors of important diseases. Although the applications of genetic technologies in insect disease vectors have been conducted primarily in mosquitoes, other insect systems could benefit from current technologies. While the various technological platforms are likely to function in diverse arthropods, the delivery of these technologies to cells and tissues of interest is the major technical constraint that limits their widespread adoption. Increased community resources of various types would enhance the adoption of these technologies and potentially eliminate technical limitations.
Collapse
Affiliation(s)
- Frank Criscione
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD 20850, United States.
| | - David A O'Brochta
- Institute for Bioscience and Biotechnology Research, Department of Entomology, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD 20850, United States.
| | - William Reid
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD 20850, United States.
| |
Collapse
|
228
|
Micellar carriers for the delivery of multiple therapeutic agents. Colloids Surf B Biointerfaces 2015; 135:291-308. [PMID: 26263217 DOI: 10.1016/j.colsurfb.2015.07.046] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 07/16/2015] [Accepted: 07/19/2015] [Indexed: 12/27/2022]
Abstract
Multi-drug therapy is described as a simultaneous or sequential administration of two or more drugs with similar or different mechanisms of action and is recognized as a more efficient solution to combat successfully, various ailments. Polymeric micelles (PMs) are self-assemblies of block copolymers providing numerous opportunities for drug delivery. To date various micellar formulations were studied for delivery of drugs, nutraceuticals and genes; a few of them are in clinical trials. It was observed that there is an immense need for the development of PMs embedding multiple therapeutic agents to combat various ailments, including cancers, HIV/AIDS, malaria, multiple sclerosis, hypertension, infectious diseases, cardiovascular and metabolic diseases, immune disorders and many psychiatric disorders. Several combinations of drug-drug, drug-nutraceutical, drug-gene and drug-siRNA explored to date are detailed in this review, with a special emphasis on their potential and future perspectives. A summary of various preparation methods, characterization techniques and applications of PMs are also provided. This review presents a holistic approach on multi-drug delivery using micellar carriers and emphasizes on the development of therapeutic hybrids embedding novel combinations for safer and effective therapy.
Collapse
|
229
|
Gish RG, Yuen MF, Chan HLY, Given BD, Lai CL, Locarnini SA, Lau JYN, Wooddell CI, Schluep T, Lewis DL. Synthetic RNAi triggers and their use in chronic hepatitis B therapies with curative intent. Antiviral Res 2015; 121:97-108. [PMID: 26129970 DOI: 10.1016/j.antiviral.2015.06.019] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/27/2015] [Accepted: 06/27/2015] [Indexed: 02/06/2023]
Abstract
Current therapies for chronic hepatitis B virus infection (CHB) - nucleos(t)ide analogue reverse transcriptase inhibitors and interferons - result in low rates of functional cure defined as sustained off-therapy seroclearance of hepatitis B surface antigen (HBsAg). One likely reason is the inability of these therapies to consistently and substantially reduce the levels of viral antigen production. Accumulated evidence suggests that high serum levels of HBsAg result in exhaustion of the host immune system, rendering it unable to mount the effective antiviral response required for HBsAg clearance. New mechanistic approaches are required to produce high rates of HBsAg seroclearance in order to greatly reduce off-treatment disease progression. Already shown to be a clinically viable means of reducing gene expression in a number of other diseases, therapies based on RNA interference (RNAi) can directly target hepatitis B virus transcripts with high specificity, profoundly reducing the production of viral proteins. The fact that the viral RNA transcripts contain overlapping sequences means that a single RNAi trigger can result in the degradation of all viral transcripts, including all messenger RNAs and pregenomic RNA. Advances in the design of RNAi triggers have increased resistance to degradation and reduced nonspecific innate immune stimulation. Additionally, new methods to effectively deliver the trigger to liver hepatocytes, and specifically to the cytoplasmic compartment, have resulted in increased efficacy and tolerability. An RNAi-based drug currently in clinical trials is ARC-520, a dynamic polyconjugate in which the RNAi trigger is conjugated to cholesterol, which is coinjected with a hepatocyte-targeted, membrane-active peptide. Phase 2a clinical trial results indicate that ARC-520 was well tolerated and resulted in significant, dose-dependent reduction in HBsAg for up to 57days in CHB patients. RNAi-based therapies may play an important role in future therapeutic regimes aimed at improving HBsAg seroclearance and eliminating the need for lifelong therapy. This paper forms part of a symposium in Antiviral Research on "An unfinished story: from the discovery of the Australia antigen to the development of new curative therapies for hepatitis B."
Collapse
Affiliation(s)
- Robert G Gish
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA; Hepatitis B Foundation, Doylestown, PA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|