151
|
Hashiba K, Sato Y, Harashima H. pH-labile PEGylation of siRNA-loaded lipid nanoparticle improves active targeting and gene silencing activity in hepatocytes. J Control Release 2017; 262:239-246. [PMID: 28774839 DOI: 10.1016/j.jconrel.2017.07.046] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/03/2017] [Accepted: 07/31/2017] [Indexed: 11/17/2022]
Abstract
Lipid nanoparticles (LNPs) are one of the promising technologies for the in vivo delivery of short interfering RNA (siRNA). Modifying LNPs with polyethyleneglycol (PEG) is widely used to inhibit non-specific interactions with serum components in the blood stream, and is a useful strategy for maximizing the efficiency of active targeting. However, it is a widely accepted fact that PEGylation of the LNP surface strongly inhibits fusion between LNPs and endosomal membranes, resulting in poor cytosolic siRNA delivery, a process that is referred to as the 'PEG-dilemma'. In the present study, in an attempt to overcome this problem, siRNA-loaded LNPs were modified with PEG through maleic anhydride, a pH-labile linkage. The in vitro, suppression of cationic charge, stealth function at physiological pH up to 1h and the rapid desorption of PEG and restoration of fusogenic activity under slightly acidic conditions (within only 2min) were achieved by PEG modification of the LNPs through maleic anhydride. In vivo, PEG modification through maleic anhydride resulted in a dramatic improvement in the targeting capability of the active targeting of ligand (N-acetyl-d-galactosamine)-modified LNPs to hepatocytes, with an approximately 14-fold increase in gene silencing activity in factor 7 model mice. Taken together, the maleic anhydride-mediated pH-labile PEGylation of the active targeting LNPs is a useful strategy for achieving the specific and efficient delivery of siRNAs in vivo.
Collapse
Affiliation(s)
- Kazuki Hashiba
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
152
|
Engineered polymeric nanoparticles to guide the cellular internalization and trafficking of small interfering ribonucleic acids. J Control Release 2017; 259:3-15. [DOI: 10.1016/j.jconrel.2017.02.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 12/29/2022]
|
153
|
Darvishi B, Farahmand L, Majidzadeh-A K. Stimuli-Responsive Mesoporous Silica NPs as Non-viral Dual siRNA/Chemotherapy Carriers for Triple Negative Breast Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 7:164-180. [PMID: 28624192 PMCID: PMC5415966 DOI: 10.1016/j.omtn.2017.03.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 12/31/2022]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive and lethal subtype of breast cancer. It is associated with a very poor prognosis and intrinsically resistant to several conventional and targeted chemotherapy agents and has a 5-year survival rate of less than 25%. Because the treatment options for TNBC are very limited and not efficient enough for achieving minimum desired goals, shifting toward a new generation of anti-cancer agents appears to be very critical. Among recent alternative approaches being proposed, small interfering RNA (siRNA) gene therapy can potently suppress Bcl-2 proto-oncogene and p-glycoprotein gene expression, the most important chemotherapy resistance inducers in TNBC. When resensitized, primarily ineffective chemotherapy drugs turn back into valuable sources for further intensive chemotherapy. Regrettably, siRNA's poor stability, rapid clearance in the circulatory system, and poor cellular uptake mostly hampers the beneficial outcomes of siRNA therapy. Considering these drawbacks, dual siRNA/chemotherapy drug encapsulation in targeted delivery vehicles, especially mesoporous silica nanoparticles (MSNs) appears to be the most reasonable solution. The literature is full of reports of successful treatments of multi-drug-resistant cancer cells by administration of dual drug/siRNA-loaded MSNs. Here we tried to answer the question of whether application of a similar approach with identical delivery devices in TNBC is rational.
Collapse
Affiliation(s)
- Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, 1517964311 Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, 1517964311 Tehran, Iran
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, 1517964311 Tehran, Iran.
| |
Collapse
|
154
|
Quantitative determination of a siRNA (AD00370) in rat plasma using peptide nucleic acid probe and HPLC with fluorescence detection. Bioanalysis 2017; 9:861-872. [PMID: 28617037 DOI: 10.4155/bio-2017-0017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIM Toxicokinetic and pharmacokinetic studies of therapeutic oligonucleotides require validated bioanalytical methods for sensitive and specific quantification of oligonucleotide drug candidates in biological samples. RESULTS A peptide nucleic acid (PNA) hybridization-based HPLC-fluorescence assay was developed and validated for quantification of Arrowhead Pharmaceuticals' proprietary siRNA in rat plasma samples via hybridization and anion-exchange-HPLC (AEX-HPLC) with fluorescence detection. CONCLUSION The validated method provided a sensitive and selective approach for quantification of siRNA in biological samples at a linear quantitation range of 1-1000 ng/ml. The assay requires only 25 μl of plasma sample and shows excellent accuracy and precision even without using an internal standard, providing a useful quantification method for siRNA determination in biological matrix with limited sample volume.
Collapse
|
155
|
Dana H, Chalbatani GM, Mahmoodzadeh H, Karimloo R, Rezaiean O, Moradzadeh A, Mehmandoost N, Moazzen F, Mazraeh A, Marmari V, Ebrahimi M, Rashno MM, Abadi SJ, Gharagouzlo E. Molecular Mechanisms and Biological Functions of siRNA. INTERNATIONAL JOURNAL OF BIOMEDICAL SCIENCE : IJBS 2017; 13:48-57. [PMID: 28824341 PMCID: PMC5542916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
One of the most important advances in biology has been the discovery that siRNA (small interfering RNA) is able to regulate the expression of genes, by a phenomenon known as RNAi (RNA interference). The discovery of RNAi, first in plants and Caenorhabditis elegans and later in mammalian cells, led to the emergence of a transformative view in biomedical research. siRNA has gained attention as a potential therapeutic reagent due to its ability to inhibit specific genes in many genetic diseases. siRNAs can be used as tools to study single gene function both in vivo and in-vitro and are an attractive new class of therapeutics, especially against undruggable targets for the treatment of cancer and other diseases. The siRNA delivery systems are categorized as non-viral and viral delivery systems. The non-viral delivery system includes polymers; Lipids; peptides etc. are the widely studied delivery systems for siRNA. Effective pharmacological use of siRNA requires 'carriers' that can deliver the siRNA to its intended site of action. The carriers assemble the siRNA into supramolecular complexes that display functional properties during the delivery process.
Collapse
Affiliation(s)
- Hassan Dana
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | | | - Habibollah Mahmoodzadeh
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Rezvan Karimloo
- Department of Medicine, Zahedan Medical Science, Zahedan, Iran
| | - Omid Rezaiean
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Amirreza Moradzadeh
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Narges Mehmandoost
- Department of chemistry, University of Sistan and Baluchestan, Zahedan, Iran
| | - Fateme Moazzen
- Department of Laboratory sciences, Zahedan Branch, Islamic Azad University, Zahedan, Iran
| | - Ali Mazraeh
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Vahid Marmari
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | | | | | - Saeid Jan Abadi
- Department of Microbiology, Shiraz Medical Science, Shiraz, Iran
| | - Elahe Gharagouzlo
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
156
|
Tatiparti K, Sau S, Kashaw SK, Iyer AK. siRNA Delivery Strategies: A Comprehensive Review of Recent Developments. NANOMATERIALS (BASEL, SWITZERLAND) 2017; 7:E77. [PMID: 28379201 PMCID: PMC5408169 DOI: 10.3390/nano7040077] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/07/2017] [Accepted: 03/31/2017] [Indexed: 01/01/2023]
Abstract
siRNA is a promising therapeutic solution to address gene overexpression or mutations as a post-transcriptional gene regulation process for several pathological conditions such as viral infections, cancer, genetic disorders, and autoimmune disorders like arthritis. This therapeutic method is currently being actively pursued in cancer therapy because siRNA has been found to suppress the oncogenes and address mutations in tumor suppressor genes and elucidate the key molecules in cellular pathways in cancer. It is also effective in personalized gene therapy for several diseases due to its specificity, adaptability, and broad targeting capability. However, naked siRNA is unstable in the bloodstream and cannot efficiently cross cell membranes besides being immunogenic. Therefore, careful design of the delivery systems is essential to fully utilize the potential of this therapeutic solution. This review presents a comprehensive update on the challenges of siRNA delivery and the current strategies used to develop nanoparticulate delivery systems.
Collapse
Affiliation(s)
- Katyayani Tatiparti
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Samaresh Sau
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Sushil Kumar Kashaw
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar 470003, India.
| | - Arun K Iyer
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
157
|
Polymers in the Delivery of siRNA for the Treatment of Virus Infections. Top Curr Chem (Cham) 2017; 375:38. [PMID: 28324594 PMCID: PMC7100576 DOI: 10.1007/s41061-017-0127-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/22/2017] [Indexed: 01/13/2023]
Abstract
Viral diseases remain a major cause of death worldwide. Despite advances in vaccine and antiviral drug technology, each year over three million people die from a range of viral infections. Predominant viruses include human immunodeficiency virus, hepatitis viruses, and gastrointestinal and respiratory viruses. Now more than ever, robust, easily mobilised and cost-effective antiviral strategies are needed to combat both known and emerging disease threats. RNA interference and small interfering (si)RNAs were initially hailed as a “magic bullet”, due to their ability to inhibit the synthesis of any protein via the degradation of its complementary messenger RNA sequence. Of particular interest was the potential for attenuating viral mRNAs contributing to the pathogenesis of disease that were not able to be targeted by vaccines or antiviral drugs. However, it was soon discovered that delivery of active siRNA molecules to the infection site in vivo was considerably more difficult than anticipated, due to a number of physiological barriers in the body. This spurred a new wave of investigation into nucleic acid delivery vehicles which could facilitate safe, targeted and effective administration of the siRNA as therapy. Amongst these, cationic polymer delivery vehicles have emerged as a promising candidate as they are low-cost and easy to produce at an industrial scale, and bind to the siRNA by non-specific electrostatic interactions. These nanoparticles (NPs) can be functionally designed to target the infection site, improve uptake in infected cells, release the siRNA inside the endosome and facilitate delivery into the cell cytoplasm. They may also have the added benefit of acting as adjuvants. This chapter provides a background around problems associated with the translation of siRNA as antiviral treatments, reviews the progress made in nucleic acid therapeutics and discusses current methods and progress in overcoming these challenges. It also addresses the importance of combining physicochemical characterisation of the NPs with in vitro and in vivo data.
Collapse
|
158
|
Huang Y. Preclinical and Clinical Advances of GalNAc-Decorated Nucleic Acid Therapeutics. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 6:116-132. [PMID: 28325278 PMCID: PMC5363494 DOI: 10.1016/j.omtn.2016.12.003] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/04/2016] [Accepted: 12/04/2016] [Indexed: 01/03/2023]
Abstract
A main challenge in realizing the full potential of nucleic acid therapeutics is efficient delivery of them into targeted tissues and cells. N-acetylgalactosamine (GalNAc) is a well-defined liver-targeted moiety benefiting from its high affinity with asialoglycoprotein receptor (ASGPR). By conjugating it directly to the oligonucleotides or decorating it to a certain delivery system as a targeting moiety, GalNAc has achieved compelling successes in the development of nucleic acid therapeutics in recent years. Several oligonucleotide modalities are undergoing pivotal clinical studies, followed by a blooming pipeline in the preclinical stage. This review covers the progress of GalNAc-decorated oligonucleotide drugs, including siRNAs, anti-miRs, and ASOs, which provides a panorama for this field.
Collapse
Affiliation(s)
- Yuanyu Huang
- Advanced Research Institute for Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China; Institute of Molecular Medicine, Peking University, Beijing 100871, China.
| |
Collapse
|
159
|
Sanhueza CA, Baksh MM, Thuma B, Roy MD, Dutta S, Préville C, Chrunyk BA, Beaumont K, Dullea R, Ammirati M, Liu S, Gebhard D, Finley JE, Salatto CT, King-Ahmad A, Stock I, Atkinson K, Reidich B, Lin W, Kumar R, Tu M, Menhaji-Klotz E, Price DA, Liras S, Finn MG, Mascitti V. Efficient Liver Targeting by Polyvalent Display of a Compact Ligand for the Asialoglycoprotein Receptor. J Am Chem Soc 2017; 139:3528-3536. [PMID: 28230359 PMCID: PMC6991140 DOI: 10.1021/jacs.6b12964] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A compact and stable bicyclic bridged ketal was developed as a ligand for the asialoglycoprotein receptor (ASGPR). This compound showed excellent ligand efficiency, and the molecular details of binding were revealed by the first X-ray crystal structures of ligand-bound ASGPR. This analogue was used to make potent di- and trivalent binders of ASGPR. Extensive characterization of the function of these compounds showed rapid ASGPR-dependent cellular uptake in vitro and high levels of liver/plasma selectivity in vivo. Assessment of the biodistribution in rodents of a prototypical Alexa647-labeled trivalent conjugate showed selective hepatocyte targeting with no detectable distribution in nonparenchymal cells. This molecule also exhibited increased ASGPR-directed hepatocellular uptake and prolonged retention compared to a similar GalNAc derived trimer conjugate. Selective release in the liver of a passively permeable small-molecule cargo was achieved by retro-Diels-Alder cleavage of an oxanorbornadiene linkage, presumably upon encountering intracellular thiol. Therefore, the multicomponent construct described here represents a highly efficient delivery vehicle to hepatocytes.
Collapse
Affiliation(s)
- Carlos A. Sanhueza
- School of Chemistry & Biochemistry, Georgia Institute of Technology, 901 Atlantic Avenue, Atlanta, Georgia 30332, United States
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Michael M. Baksh
- School of Chemistry & Biochemistry, Georgia Institute of Technology, 901 Atlantic Avenue, Atlanta, Georgia 30332, United States
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Benjamin Thuma
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Marc D. Roy
- Pfizer Drug Safety R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sanjay Dutta
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Cathy Préville
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Boris A. Chrunyk
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin Beaumont
- Pfizer Medicine Design, Main Street, Cambridge, Massachusetts 02139, United States
| | - Robert Dullea
- Pfizer CVMET Biology, Main Street, Cambridge, Massachusetts 02139, United States
| | - Mark Ammirati
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Shenping Liu
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - David Gebhard
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - James E. Finley
- Pfizer Drug Safety R&D, Eastern Point Road, Groton, Connecticut 06340, United States
| | | | - Amanda King-Ahmad
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ingrid Stock
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Karen Atkinson
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Benjamin Reidich
- Pfizer CVMET Biology, Main Street, Cambridge, Massachusetts 02139, United States
| | - Wen Lin
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Rajesh Kumar
- Pfizer Medicinal Sciences, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Meihua Tu
- Pfizer Medicine Design, Main Street, Cambridge, Massachusetts 02139, United States
| | - Elnaz Menhaji-Klotz
- Pfizer Medicine Design, Main Street, Cambridge, Massachusetts 02139, United States
| | - David A. Price
- Pfizer Medicine Design, Main Street, Cambridge, Massachusetts 02139, United States
| | - Spiros Liras
- Pfizer Medicine Design, Main Street, Cambridge, Massachusetts 02139, United States
| | - M. G. Finn
- School of Chemistry & Biochemistry, Georgia Institute of Technology, 901 Atlantic Avenue, Atlanta, Georgia 30332, United States
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Vincent Mascitti
- Pfizer Medicine Design, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
160
|
Cheng Y, Sellers DL, Tan JKY, Peeler DJ, Horner PJ, Pun SH. Development of switchable polymers to address the dilemma of stability and cargo release in polycationic nucleic acid carriers. Biomaterials 2017; 127:89-96. [PMID: 28284104 DOI: 10.1016/j.biomaterials.2017.02.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/15/2017] [Accepted: 02/26/2017] [Indexed: 10/20/2022]
Abstract
Cationic polymer gene delivery vehicles that effectively resist premature serum degradation often have difficulty releasing their nucleic acid cargoes. In this work, we report a pH-sensitive polymer (SP), poly(oligo(ethylene glycol) monomethyl ether methacrylate)-co-poly(2-(dimethylamino)ethyl methacrylate)-block- poly(propargyl methacrylate-graft-propyl-(4-methoxy-benzylidene)-amine) (p(PMA-PMBA)-b-(p(OEGMA-DMAEMA)), for successful in vitro and in vivo gene transfer. In the physiological condition, the hydrophobization of p(OEGMA-DMAEMA) polycations by p(PMA-PMBA) significantly enhanced the stability of its polyplexes counterpart. In endosomes, the polymer undergoes an acid-triggered hydrophilic transition through the cleavage of benzoic imines, thus allowing the vector to quickly release nucleic acid cargo due to the loss of hydrophobic functionalization. Compared to a pH-insensitive polymer (IP), SP exhibited more significant luciferase plasmid delivery efficiency with HeLa cells in vitro and with in vivo intraventricular brain injections. Therefore, the polymer designed here is a good solution to address the dilemma of stability and cargo release in gene delivery, and may have broad potential applications in therapeutic agent delivery.
Collapse
Affiliation(s)
- Yilong Cheng
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - Drew L Sellers
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - James-Kevin Y Tan
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - David J Peeler
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, United States
| | - Philip J Horner
- Center for Neuroregeneration and Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX 77030, United States.
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
161
|
Trubetskoy VS, Griffin JB, Nicholas AL, Nord EM, Xu Z, Peterson RM, Wooddell CI, Rozema DB, Wakefield DH, Lewis DL, Kanner SB. Phosphorylation-specific status of RNAi triggers in pharmacokinetic and biodistribution analyses. Nucleic Acids Res 2017; 45:1469-1478. [PMID: 28180327 PMCID: PMC5388421 DOI: 10.1093/nar/gkw828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 01/23/2023] Open
Abstract
The RNA interference (RNAi)-based therapeutic ARC-520 for chronic hepatitis B virus (HBV) infection consists of a melittin-derived peptide conjugated to N-acetylgalactosamine for hepatocyte targeting and endosomal escape, and cholesterol-conjugated RNAi triggers, which together result in HBV gene silencing. To characterize the kinetics of RNAi trigger delivery and 5΄-phosphorylation of guide strands correlating with gene knockdown, we employed a peptide-nucleic acid (PNA) hybridization assay. A fluorescent sense strand PNA probe binding to RNAi duplex guide strands was coupled with anion exchange high performance liquid chromatography to quantitate guide strands and metabolites. Compared to PCR- or ELISA-based methods, this assay enables separate quantitation of non-phosphorylated full-length guide strands from 5΄-phosphorylated forms that may associate with RNA-induced silencing complexes (RISC). Biodistribution studies in mice indicated that ARC-520 guide strands predominantly accumulated in liver. 5΄-phosphorylation of guide strands was observed within 5 min after ARC-520 injection, and was detected for at least 4 weeks corresponding to the duration of HBV mRNA silencing. Guide strands detected in RISC by AGO2 immuno-isolation represented 16% of total 5΄-phosphorylated guide strands in liver, correlating with a 2.7 log10 reduction of HBsAg. The PNA method enables pharmacokinetic analysis of RNAi triggers, elucidates potential metabolic processing events and defines pharmacokinetic-pharmacodynamic relationships.
Collapse
MESH Headings
- Animals
- Argonaute Proteins/genetics
- Argonaute Proteins/metabolism
- Female
- Gene Knockdown Techniques
- Hepatitis B Surface Antigens/blood
- Hepatitis B Surface Antigens/genetics
- Hepatitis B virus/genetics
- Hepatitis B virus/metabolism
- Hepatitis B, Chronic/metabolism
- Hepatitis B, Chronic/therapy
- Hepatitis B, Chronic/virology
- Humans
- Kinetics
- Liver/metabolism
- Liver/virology
- Mice
- Mice, Inbred ICR
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Peptide Nucleic Acids/genetics
- Peptide Nucleic Acids/metabolism
- Phosphorylation
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- RNA, Viral/genetics
- RNA, Viral/metabolism
- RNA-Induced Silencing Complex/genetics
- RNA-Induced Silencing Complex/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
| | - Jacob B. Griffin
- Department of Biology, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | - Anthony L. Nicholas
- Department of Chemistry, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | - Eric M. Nord
- Department of Chemistry, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | - Zhao Xu
- Department of Biology, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | - Ryan M. Peterson
- Department of Biology, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | | | - David B. Rozema
- Department of Chemistry, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | - Darren H. Wakefield
- Department of Chemistry, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| | | | - Steven B. Kanner
- Department of Biology, Arrowhead Pharmaceuticals, Inc., Madison, WI 53711, USA
| |
Collapse
|
162
|
Zhang P, Wagner E. History of Polymeric Gene Delivery Systems. Top Curr Chem (Cham) 2017; 375:26. [PMID: 28181193 DOI: 10.1007/s41061-017-0112-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 01/24/2017] [Indexed: 12/16/2022]
Abstract
As an option for genetic disease treatment and an alternative for traditional cancer chemotherapy, gene therapy achieves significant attention. Nucleic acid delivery, however, remains a main challenge in human gene therapy. Polymer-based delivery systems offer a safer and promising route for therapeutic gene delivery. Over the past five decades, various cationic polymers have been optimized for increasingly effective nucleic acid transfer. This resulted in a chemical evolution of cationic polymers from the first-generation polycations towards bioinspired multifunctional sequence-defined polymers and nanocomposites. With the increasing of knowledge in molecular biological processes and rapid progress of macromolecular chemistry, further improvement of polymeric nucleic acid delivery systems will provide effective tool for gene-based therapy in the near future.
Collapse
Affiliation(s)
- Peng Zhang
- Pharmaceutical Biotechnology, Center for System-Based Drug Research Ludwig-Maximilians-Universität, 81377, Munich, Germany. .,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany.
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-Based Drug Research Ludwig-Maximilians-Universität, 81377, Munich, Germany.,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany.,Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, 80799, Munich, Germany
| |
Collapse
|
163
|
Ly S, Navaroli DM, Didiot MC, Cardia J, Pandarinathan L, Alterman JF, Fogarty K, Standley C, Lifshitz LM, Bellve KD, Prot M, Echeverria D, Corvera S, Khvorova A. Visualization of self-delivering hydrophobically modified siRNA cellular internalization. Nucleic Acids Res 2017; 45:15-25. [PMID: 27899655 PMCID: PMC5224471 DOI: 10.1093/nar/gkw1005] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 10/07/2016] [Accepted: 10/24/2016] [Indexed: 01/07/2023] Open
Abstract
siRNAs are a new class of therapeutic modalities with promising clinical efficacy that requires modification or formulation for delivery to the tissue and cell of interest. Conjugation of siRNAs to lipophilic groups supports efficient cellular uptake by a mechanism that is not well characterized. Here we study the mechanism of internalization of asymmetric, chemically stabilized, cholesterol-modified siRNAs (sd-rxRNAs®) that efficiently enter cells and tissues without the need for formulation. We demonstrate that uptake is rapid with significant membrane association within minutes of exposure followed by the formation of vesicular structures and internalization. Furthermore, sd-rxRNAs are internalized by a specific class of early endosomes and show preferential association with epidermal growth factor (EGF) but not transferrin (Tf) trafficking pathways as shown by live cell TIRF and structured illumination microscopy (SIM). In fixed cells, we observe ∼25% of sd-rxRNA co-localizing with EGF and <5% with Tf, which is indicative of selective endosomal sorting. Likewise, preferential sd-rxRNA co-localization was demonstrated with EEA1 but not RBSN-containing endosomes, consistent with preferential EGF-like trafficking through EEA1-containing endosomes. sd-rxRNA cellular uptake is a two-step process, with rapid membrane association followed by internalization through a selective, saturable subset of the endocytic process. However, the mechanistic role of EEA1 is not yet known. This method of visualization can be used to better understand the kinetics and mechanisms of hydrophobic siRNA cellular uptake and will assist in further optimization of these types of compounds for therapeutic intervention.
Collapse
Affiliation(s)
- Socheata Ly
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Deanna M Navaroli
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Marie-Cécile Didiot
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | - Julia F Alterman
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Kevin Fogarty
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Clive Standley
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Lawrence M Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Karl D Bellve
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Matthieu Prot
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Dimas Echeverria
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Anastasia Khvorova
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
164
|
Recent advances in the design, development, and targeting mechanisms of polymeric micelles for delivery of siRNA in cancer therapy. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
165
|
Ganbold T, Gerile G, Xiao H, Baigude H. Efficient in vivo siRNA delivery by stabilized d-peptide-based lipid nanoparticles. RSC Adv 2017. [DOI: 10.1039/c6ra25862j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A lipid functionalized d-dipeptide has shown remarkable biocompatibility and tissue targeting as well as excellent RNAi delivery efficiency in vivo.
Collapse
Affiliation(s)
- Tsogzolmaa Ganbold
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| | - Gerile Gerile
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| | - Hai Xiao
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| | - Huricha Baigude
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- PR China
| |
Collapse
|
166
|
Zhang A, Yao L, An M. Reversing the undesirable pH-profile of doxorubicin via activation of a di-substituted maleamic acid prodrug at tumor acidity. Chem Commun (Camb) 2017; 53:12826-12829. [DOI: 10.1039/c7cc06843c] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Selective drug release at pH 6.7 over pH 7.4 is achieved through a combination of ‘switch-on’ and ‘switch-off’ mechanisms.
Collapse
Affiliation(s)
- Anqi Zhang
- Department of Chemistry
- State University of New York (SUNY)
- Binghamton University
- Binghamton
- USA
| | - Lan Yao
- Department of Physics
- Applied Physics and Astronomy
- SUNY
- Binghamton University
- Binghamton
| | - Ming An
- Department of Chemistry
- State University of New York (SUNY)
- Binghamton University
- Binghamton
- USA
| |
Collapse
|
167
|
Huang X, Leroux JC, Castagner B. Well-Defined Multivalent Ligands for Hepatocytes Targeting via Asialoglycoprotein Receptor. Bioconjug Chem 2016; 28:283-295. [DOI: 10.1021/acs.bioconjchem.6b00651] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Xiangang Huang
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute
of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Bastien Castagner
- Department
of Pharmacology and Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
168
|
Munsell EV, Ross NL, Sullivan MO. Journey to the Center of the Cell: Current Nanocarrier Design Strategies Targeting Biopharmaceuticals to the Cytoplasm and Nucleus. Curr Pharm Des 2016; 22:1227-44. [PMID: 26675220 DOI: 10.2174/1381612822666151216151420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/15/2015] [Indexed: 01/06/2023]
Abstract
New biopharmaceutical molecules, potentially able to provide more personalized and effective treatments, are being identified through the advent of advanced synthetic biology strategies, sophisticated chemical synthesis approaches, and new analytical methods to assess biological potency. However, translation of many of these structures has been significantly limited due to the need for more efficient strategies to deliver macromolecular therapeutics to desirable intracellular sites of action. Engineered nanocarriers that encapsulate peptides, proteins, or nucleic acids are generally internalized into target cells via one of several endocytic pathways. These nanostructures, entrapped within endosomes, must navigate the intracellular milieu to orchestrate delivery to the intended destination, typically the cytoplasm or nucleus. For therapeutics active in the cytoplasm, endosomal escape continues to represent a limiting step to effective treatment, since a majority of nanocarriers trapped within endosomes are ultimately marked for enzymatic degradation in lysosomes. Therapeutics active in the nucleus have the added challenges of reaching and penetrating the nuclear envelope, and nuclear delivery remains a preeminent challenge preventing clinical translation of gene therapy applications. Herein, we review cutting-edge peptide- and polymer-based design strategies with the potential to enable significant improvements in biopharmaceutical efficacy through improved intracellular targeting. These strategies often mimic the activities of pathogens, which have developed innate and highly effective mechanisms to penetrate plasma membranes and enter the nucleus of host cells. Understanding these mechanisms has enabled advances in synthetic peptide and polymer design that may ultimately improve intracellular trafficking and bioavailability, leading to increased access to new classes of biotherapeutics.
Collapse
Affiliation(s)
| | | | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, Delaware.
| |
Collapse
|
169
|
Schluep T, Lickliter J, Hamilton J, Lewis DL, Lai CL, Lau JY, Locarnini SA, Gish RG, Given BD. Safety, Tolerability, and Pharmacokinetics of ARC-520 Injection, an RNA Interference-Based Therapeutic for the Treatment of Chronic Hepatitis B Virus Infection, in Healthy Volunteers. Clin Pharmacol Drug Dev 2016; 6:350-362. [PMID: 27739230 PMCID: PMC5516171 DOI: 10.1002/cpdd.318] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/05/2016] [Indexed: 12/18/2022]
Abstract
ARC‐520 Injection, an RNA interference drug for the treatment of hepatitis B that targets cccDNA‐derived viral mRNA transcripts with high specificity, effectively reduces the production of viral proteins and HBV DNA. In this phase 1 randomized, double‐blind, placebo‐controlled study, 54 healthy volunteers (half male, half female) received a single, intravenous dose of 0.01–4.0 mg/kg ARC‐520 Injection (n = 36) or placebo (n = 18). Assessments included safety, tolerability, pharmacokinetics, and pharmacodynamics (cytokines and complement). Pharmacokinetics of the siRNA and peptide excipient components contained in ARC‐520 Injection showed a relatively short half‐life of 3–5 and 8–10 hours, respectively. Dose exposure linearity was demonstrated within the dose range. ARC‐520 Injection was well tolerated, with adverse‐event frequency the same as placebo and no serious adverse events. ARC‐520 Injection was initially found to induce histamine release through mast cell degranulation, resulting in 2 moderate hypersensitivity reactions. However, after initiation of pretreatment with oral antihistamine, no further hypersensitivity reactions occurred. Low‐level, transient complement induction and sporadic, mild, and transient elevations of several cytokines were observed but not associated with any symptoms. ARC‐520 Injection showed a favorable tolerability profile in this single‐dose study in healthy volunteers. Oral antihistamine pretreatment is recommended in the future to offset mast cell degranulation stimulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robert G Gish
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University Medical Center, Stanford, CA, USA
| | | |
Collapse
|
170
|
Smekalova EM, Kotelevtsev YV, Leboeuf D, Shcherbinina EY, Fefilova AS, Zatsepin TS, Koteliansky V. lncRNA in the liver: Prospects for fundamental research and therapy by RNA interference. Biochimie 2016; 131:159-172. [DOI: 10.1016/j.biochi.2016.06.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
|
171
|
Huang CH, Takemoto H, Nomoto T, Tomoda K, Matsui M, Nishiyama N. Utility of the 2-Nitrobenzenesulfonamide Group as a Chemical Linker for Enhanced Extracellular Stability and Cytosolic Cleavage in siRNA-Conjugated Polymer Systems. ChemMedChem 2016; 12:19-22. [DOI: 10.1002/cmdc.201600488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/26/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Chih Hao Huang
- Laboratory for Chemistry and Life Science; Institute of Innovative Research; Tokyo Institute of Technology; R1-11, 4259 Nagatsuta-cho, Midori-ku Yokohama Kanagawa 226-8503 Japan
| | - Hiroyasu Takemoto
- Laboratory for Chemistry and Life Science; Institute of Innovative Research; Tokyo Institute of Technology; R1-11, 4259 Nagatsuta-cho, Midori-ku Yokohama Kanagawa 226-8503 Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science; Institute of Innovative Research; Tokyo Institute of Technology; R1-11, 4259 Nagatsuta-cho, Midori-ku Yokohama Kanagawa 226-8503 Japan
| | - Keishiro Tomoda
- Laboratory for Chemistry and Life Science; Institute of Innovative Research; Tokyo Institute of Technology; R1-11, 4259 Nagatsuta-cho, Midori-ku Yokohama Kanagawa 226-8503 Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science; Institute of Innovative Research; Tokyo Institute of Technology; R1-11, 4259 Nagatsuta-cho, Midori-ku Yokohama Kanagawa 226-8503 Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science; Institute of Innovative Research; Tokyo Institute of Technology; R1-11, 4259 Nagatsuta-cho, Midori-ku Yokohama Kanagawa 226-8503 Japan
| |
Collapse
|
172
|
Zhou J, Wu Y, Wang C, Cheng Q, Han S, Wang X, Zhang J, Deng L, Zhao D, Du L, Cao H, Liang Z, Huang Y, Dong A. pH-Sensitive Nanomicelles for High-Efficiency siRNA Delivery in Vitro and in Vivo: An Insight into the Design of Polycations with Robust Cytosolic Release. NANO LETTERS 2016; 16:6916-6923. [PMID: 27748606 DOI: 10.1021/acs.nanolett.6b02915] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The extremely low efficient cytosolic release of the internalized siRNA has emerged recently as a central issue for siRNA delivery, while there is a lack of guidelines to facilitate the cytosolic release of internalized siRNA. To address these concerns, we studied the contribution of the pH-sensitive inner core on handling the cytosolic release of siRNA delivered by a series of PG-P(DPAx-co-DMAEMAy)-PCB amphiphilic polycation nanomicelles (GDDC-Ms) with extremely low internalization (<1/4 of lipofactamine 2000 (Lipo2000)). Significantly, just by varying the mole ratio of DPA and DMAEMA to adjust the initial disassembly pH (pHdis) of the core near to 6.8, GDDC4-Ms/siRNA could get nearly 98.8% silencing efficiency at w/w = 12 with 50 nM siRNA and ∼78% silencing efficiency at w/w = 30 with a very low dose of 5 nM siRNA in HepG-2 cell lines, while Lipo2000 only got 65.7% with 50 nM siRNA. Furthermore, ∼98.4% silencing efficiency was also realized in the hard-to-transfect human acute monoblastic leukemia cell line U937 by GDDC4-Ms/siRNA (at w/w = 15, 50 nM siRNA), in the inefficient case for Lipo2000. Additionally, the high silencing efficiency (∼80%) in skin tissue in vivo was discovered. Undoubtedly, the robust potential of GDDC4-Ms in handling the cytosolic release paves a simple but efficient new way for the design of the nonviral siRNA vector.
Collapse
Affiliation(s)
- Junhui Zhou
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Yidi Wu
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Changrong Wang
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | - Qiang Cheng
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Shangcong Han
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
- Department of Pharmaceutics, School of Pharmacy, Qingdao University , Qingdao 266021, China
| | - Xiaoxia Wang
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Jianhua Zhang
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | - Liandong Deng
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | - Deyao Zhao
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Lili Du
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Huiqing Cao
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Zicai Liang
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Yuanyu Huang
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University , Beijing 100871, China
| | - Anjie Dong
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| |
Collapse
|
173
|
Dengl S, Sustmann C, Brinkmann U. Engineered hapten-binding antibody derivatives for modulation of pharmacokinetic properties of small molecules and targeted payload delivery. Immunol Rev 2016; 270:165-77. [PMID: 26864111 PMCID: PMC4755198 DOI: 10.1111/imr.12386] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Hapten‐binding antibodies have for more than 50 years played a pivotal role in immunology, paving the way to antibody generation (as haptens are very important and robust immunogens), to antibody characterization (as the first structures generated more than 40 years ago were those of hapten binders), and enabled and expanded antibody engineering technologies. The latter field of engineered antibodies evolved over many years and many steps resulting in recombinant humanized or human‐derived antibody derivatives in multiple formats. Today, hapten‐binding antibodies are applied not only as reagents and tools (where they still play an important part) but evolved also to engineered targeting and pretargeting vehicles for disease diagnosis and therapy. Here we describe recent applications of hapten‐binding antibodies and of engineered mono‐ and bispecific hapten‐binding antibody derivatives. We have designed and applied these molecules for the modulation of the pharmacokinetic properties of small compounds or peptides. They are also integrated as additional binding entities into bispecific antibody formats. Here they serve as non‐covalent or covalent coupling modules to haptenylated compounds, to enable targeted payload delivery to disease tissues or cells.
Collapse
Affiliation(s)
- Stefan Dengl
- 1Roche Pharma Research & Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Penzberg, Germany
| | - Claudio Sustmann
- 1Roche Pharma Research & Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Penzberg, Germany
| | - Ulrich Brinkmann
- 1Roche Pharma Research & Early Development, Large Molecule Research, Roche Innovation Center Penzberg, Penzberg, Germany
| |
Collapse
|
174
|
Li H, Chen G, Das S. Electric double layer electrostatics of pH-responsive spherical polyelectrolyte brushes in the decoupled regime. Colloids Surf B Biointerfaces 2016; 147:180-190. [DOI: 10.1016/j.colsurfb.2016.07.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 11/29/2022]
|
175
|
Ho W, Zhang XQ, Xu X. Biomaterials in siRNA Delivery: A Comprehensive Review. Adv Healthc Mater 2016; 5:2715-2731. [PMID: 27700013 DOI: 10.1002/adhm.201600418] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/07/2016] [Indexed: 01/31/2023]
Abstract
With the dearth of effective treatment options for prominent diseases including Ebola and cancer, RNA interference (RNAi), a sequence-specific mechanism for genetic regulation that can silence nearly any gene, holds the promise of unlimited potential in treating illness ever since its discovery in 1999. Given the large size, unstable tertiary structure in physiological conditions and negative charge of small interfering RNAs (siRNAs), the development of safe and effective delivery vehicles is of critical importance in order to drive the widespread use of RNAi therapeutics into clinical settings. Immense amounts of time and billions of dollars have been devoted into the design of novel and diverse delivery strategies, and there are a handful of delivery systems that have been successfully translated into clinic. This review provides an introduction to the in vivo barriers that need to be addressed by siRNA delivery systems. We also discuss the progress up to the most effective and clinically advanced siRNA delivery systems including liposomal, polymeric and siRNA conjugate delivery systems, as well as their design to overcome the challenges.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| | - Xue-Qing Zhang
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical, Biological and Pharmaceutical Engineering; Newark School of Engineering; New Jersey Institute of Technology; Newark NJ 07102 USA
| |
Collapse
|
176
|
Sato Y, Sakurai Y, Kajimoto K, Nakamura T, Yamada Y, Akita H, Harashima H. Innovative Technologies in Nanomedicines: From Passive Targeting to Active Targeting/From Controlled Pharmacokinetics to Controlled Intracellular Pharmacokinetics. Macromol Biosci 2016; 17. [PMID: 27797146 DOI: 10.1002/mabi.201600179] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yu Sakurai
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Kazuaki Kajimoto
- Health Research Institute; National Institute of Advanced Industrial Science and Technology (AIST); 2217-14 Hayashi-cho Takamatsu, Kagawa 761-0395 Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences; Chiba University; 1-8-1 Inohana Chuo-ku, Chiba-shi, Chiba 260-8675 Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| |
Collapse
|
177
|
DeRosa F, Guild B, Karve S, Smith L, Love K, Dorkin JR, Kauffman KJ, Zhang J, Yahalom B, Anderson DG, Heartlein MW. Therapeutic efficacy in a hemophilia B model using a biosynthetic mRNA liver depot system. Gene Ther 2016; 23:699-707. [PMID: 27356951 PMCID: PMC5059749 DOI: 10.1038/gt.2016.46] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/06/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022]
Abstract
DNA-based gene therapy has considerable therapeutic potential, but the challenges associated with delivery continue to limit progress. Messenger RNA (mRNA) has the potential to provide for transient production of therapeutic proteins, without the need for nuclear delivery and without the risk of insertional mutagenesis. Here we describe the sustained delivery of therapeutic proteins in vivo in both rodents and non-human primates via nanoparticle-formulated mRNA. Nanoparticles formulated with lipids and lipid-like materials were developed for delivery of two separate mRNA transcripts encoding either human erythropoietin (hEPO) or factor IX (hFIX) protein. Dose-dependent protein production was observed for each mRNA construct. Upon delivery of hEPO mRNA in mice, serum EPO protein levels reached several orders of magnitude (>125 000-fold) over normal physiological values. Further, an increase in hematocrit (Hct) was established, demonstrating that the exogenous mRNA-derived protein maintained normal activity. The capacity of producing EPO in non-human primates via delivery of formulated mRNA was also demonstrated as elevated EPO protein levels were observed over a 72-h time course. Exemplifying the possible broad utility of mRNA drugs, therapeutically relevant amounts of human FIX (hFIX) protein were achieved upon a single intravenous dose of hFIX mRNA-loaded lipid nanoparticles in mice. In addition, therapeutic value was established within a hemophilia B (FIX knockout (KO)) mouse model by demonstrating a marked reduction in Hct loss following injury (incision) to FIX KO mice.
Collapse
Affiliation(s)
- F DeRosa
- Shire Pharmaceuticals, Lexington, MA, USA
| | - B Guild
- Shire Pharmaceuticals, Lexington, MA, USA
| | - S Karve
- Shire Pharmaceuticals, Lexington, MA, USA
| | - L Smith
- Shire Pharmaceuticals, Lexington, MA, USA
| | - K Love
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J R Dorkin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - K J Kauffman
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Zhang
- Shire Pharmaceuticals, Lexington, MA, USA
| | - B Yahalom
- Biomedical Research Models, Inc., Worcester, MA, USA
| | - D G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
178
|
Yang J, Kopeček J. Design of smart HPMA copolymer-based nanomedicines. J Control Release 2016; 240:9-23. [DOI: 10.1016/j.jconrel.2015.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 01/13/2023]
|
179
|
Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MVP, Mohseni M, Lawson D, Reimer C, Blakey DC, Xiao X, Hsu J, Revenko A, Monia BP, MacLeod AR. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med 2016; 7:314ra185. [PMID: 26582900 DOI: 10.1126/scitranslmed.aac5272] [Citation(s) in RCA: 344] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Next-generation sequencing technologies have greatly expanded our understanding of cancer genetics. Antisense technology is an attractive platform with the potential to translate these advances into improved cancer therapeutics, because antisense oligonucleotide (ASO) inhibitors can be designed on the basis of gene sequence information alone. Recent human clinical data have demonstrated the potent activity of systemically administered ASOs targeted to genes expressed in the liver. We describe the preclinical activity and initial clinical evaluation of a class of ASOs containing constrained ethyl modifications for targeting the gene encoding the transcription factor STAT3, a notoriously difficult protein to inhibit therapeutically. Systemic delivery of the unformulated ASO, AZD9150, decreased STAT3 expression in a broad range of preclinical cancer models and showed antitumor activity in lymphoma and lung cancer models. AZD9150 preclinical activity translated into single-agent antitumor activity in patients with highly treatment-refractory lymphoma and non-small cell lung cancer in a phase 1 dose-escalation study.
Collapse
Affiliation(s)
- David Hong
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Razelle Kurzrock
- UC San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA.
| | - Youngsoo Kim
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Richard Woessner
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - Anas Younes
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - John Nemunaitis
- Mary Crowley Cancer Research Center, 7777 Forest Lane, Dallas, TX 75230, USA
| | - Nathan Fowler
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Tianyuan Zhou
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Joanna Schmidt
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Minji Jo
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Samantha J Lee
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Mason Yamashita
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Steven G Hughes
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Luis Fayad
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Sarina Piha-Paul
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Murali V P Nadella
- Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, Waltham, MA 02451, USA
| | - Morvarid Mohseni
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - Deborah Lawson
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - Corinne Reimer
- Cancer Bioscience Drug Discovery, AstraZeneca Pharmaceuticals, 35 Gatehouse Drive, Waltham, MA 02451, USA
| | - David C Blakey
- Oncology iMED, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield SK10 4TF, UK
| | - Xiaokun Xiao
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Jeff Hsu
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Alexey Revenko
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - Brett P Monia
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA
| | - A Robert MacLeod
- Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92008, USA.
| |
Collapse
|
180
|
Sarisozen C, Salzano G, Torchilin VP. Recent advances in siRNA delivery. Biomol Concepts 2016; 6:321-41. [PMID: 26609865 DOI: 10.1515/bmc-2015-0019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/04/2015] [Indexed: 01/05/2023] Open
Abstract
In the 1990s an unexpected gene-silencing phenomena in plants, the later called RNA interference (RNAi), perplexed scientists. Following the proof of activity in mammalian cells, small interfering RNAs (siRNAs) have quickly crept into biomedical research as a new powerful tool for the potential treatment of different human diseases based on altered gene expression. In the past decades, several promising data from ongoing clinical trials have been reported. However, despite surprising successes in many pre-clinical studies, concrete obstacles still need to be overcome to translate therapeutic siRNAs into clinical reality. Here, we provide an update on the recent advances of RNAi-based therapeutics and highlight novel synthetic platforms for the intracellular delivery of siRNAs.
Collapse
|
181
|
Functional polyesters enable selective siRNA delivery to lung cancer over matched normal cells. Proc Natl Acad Sci U S A 2016; 113:E5702-10. [PMID: 27621434 DOI: 10.1073/pnas.1606886113] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Conventional chemotherapeutics nonselectively kill all rapidly dividing cells, which produces numerous side effects. To address this challenge, we report the discovery of functional polyesters that are capable of delivering siRNA drugs selectively to lung cancer cells and not to normal lung cells. Selective polyplex nanoparticles (NPs) were identified by high-throughput library screening on a unique pair of matched cancer/normal cell lines obtained from a single patient. Selective NPs promoted rapid endocytosis into HCC4017 cancer cells, but were arrested at the membrane of HBEC30-KT normal cells during the initial transfection period. When injected into tumor xenografts in mice, cancer-selective NPs were retained in tumors for over 1 wk, whereas nonselective NPs were cleared within hours. This translated to improved siRNA-mediated cancer cell apoptosis and significant suppression of tumor growth. Selective NPs were also able to mediate gene silencing in xenograft and orthotopic tumors via i.v. injection or aerosol inhalation, respectively. Importantly, this work highlights that different cells respond differentially to the same drug carrier, an important factor that should be considered in the design and evaluation of all NP carriers. Because no targeting ligands are required, these functional polyester NPs provide an exciting alternative approach for selective drug delivery to tumor cells that may improve efficacy and reduce adverse side effects of cancer therapies.
Collapse
|
182
|
Eldredge AC, Johnson ME, Oldenhuis NJ, Guan Z. Focused Library Approach to Discover Discrete Dipeptide Bolaamphiphiles for siRNA Delivery. Biomacromolecules 2016; 17:3138-3144. [PMID: 27563833 DOI: 10.1021/acs.biomac.6b00635] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In this study, we report a new dipeptide functionalization strategy for developing new dendritic bolaamphiphile vectors for efficient siRNA transfection. A focused library of dipeptides was constructed using four amino acids: l-arginine, l-histidine, l-lysine, and l-tryptophan. The dipeptides were coupled to two dendritic bolaamphiphile scaffolds that we developed previously, allowing us to quickly access a focused library of discrete vectors with multivalent dendritic dipeptide functionalities. The resulting discrete bolaamphiphiles were screened for siRNA delivery in vitro in HEK-293 and HeLa cells. Bolaamphiphiles functionalized with dipeptides containing Lys or Arg and either His or Trp were the most effective for in vitro siRNA delivery. Necessary cationic charge to ensure efficient siRNA binding are provided by Arg and Lys residues, whereas endosomal escape is provided through pH responsive buffering of His or membrane interactions of Trp. The most effective vectors (F10 HR/RH) exhibited greater than 75% gene silencing in multiple cell lines and exhibited serum stability.
Collapse
Affiliation(s)
- Alexander C Eldredge
- Department of Chemistry, University of California , Irvine, California 92697, United States
| | - Mark E Johnson
- Department of Chemistry, University of California , Irvine, California 92697, United States
| | - Nathan J Oldenhuis
- Department of Chemistry, University of California , Irvine, California 92697, United States
| | - Zhibin Guan
- Department of Chemistry, University of California , Irvine, California 92697, United States
| |
Collapse
|
183
|
Wang J, Mi P, Lin G, Wáng YXJ, Liu G, Chen X. Imaging-guided delivery of RNAi for anticancer treatment. Adv Drug Deliv Rev 2016; 104:44-60. [PMID: 26805788 DOI: 10.1016/j.addr.2016.01.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/27/2015] [Accepted: 01/13/2016] [Indexed: 12/12/2022]
Abstract
The RNA interference (RNAi) technique is a new modality for cancer therapy, and several candidates are being tested clinically. In the development of RNAi-based therapeutics, imaging methods can provide a visible and quantitative way to investigate the therapeutic effect at anatomical, cellular, and molecular level; to noninvasively trace the distribution; to and study the biological processes in preclinical and clinical stages. Their abilities are important not only for therapeutic optimization and evaluation but also for shortening of the time of drug development to market. Typically, imaging-functionalized RNAi therapeutics delivery that combines nanovehicles and imaging techniques to study and improve their biodistribution and accumulation in tumor site has been progressively integrated into anticancer drug discovery and development processes. This review presents an overview of the current status of translating the RNAi cancer therapeutics in the clinic, a brief description of the biological barriers in drug delivery, and the roles of imaging in aspects of administration route, systemic circulation, and cellular barriers for the clinical translation of RNAi cancer therapeutics, and with partial content for discussing the safety concerns. Finally, we focus on imaging-guided delivery of RNAi therapeutics in preclinical development, including the basic principles of different imaging modalities, and their advantages and limitations for biological imaging. With growing number of RNAi therapeutics entering the clinic, various imaging methods will play an important role in facilitating the translation of RNAi cancer therapeutics from bench to bedside.
Collapse
|
184
|
Lee SH, Kang YY, Jang HE, Mok H. Current preclinical small interfering RNA (siRNA)-based conjugate systems for RNA therapeutics. Adv Drug Deliv Rev 2016; 104:78-92. [PMID: 26514375 DOI: 10.1016/j.addr.2015.10.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/01/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Recent promising clinical results of RNA therapeutics have drawn big attention of academia and industries to RNA therapeutics and their carrier systems. To improve their feasibility in clinics, systemic evaluations of currently available carrier systems under clinical trials and preclinical studies are needed. In this review, we focus on recent noticeable preclinical studies and clinical results regarding siRNA-based conjugates for clinical translations. Advantages and drawbacks of siRNA-based conjugates are discussed, compared to particle-based delivery systems. Then, representative siRNA-based conjugates with aptamers, peptides, carbohydrates, lipids, polymers, and nanostructured materials are introduced. To improve feasibility of siRNA conjugates in preclinical studies, several considerations for the rational design of siRNA conjugates in terms of cleavability, immune responses, multivalent conjugations, and mechanism of action are also presented. Lastly, we discuss lessons from previous preclinical and clinical studies related to siRNA conjugates and perspectives of their clinical applications.
Collapse
Affiliation(s)
- Soo Hyeon Lee
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Zurich, Switzerland
| | - Yoon Young Kang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyo-Eun Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea.
| |
Collapse
|
185
|
Kim HJ, Kim A, Miyata K, Kataoka K. Recent progress in development of siRNA delivery vehicles for cancer therapy. Adv Drug Deliv Rev 2016; 104:61-77. [PMID: 27352638 DOI: 10.1016/j.addr.2016.06.011] [Citation(s) in RCA: 313] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/21/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022]
Abstract
Recent progress in RNA biology has broadened the scope of therapeutic targets of RNA drugs for cancer therapy. However, RNA drugs, typically small interfering RNAs (siRNAs), are rapidly degraded by RNases and filtrated in the kidney, thereby requiring a delivery vehicle for efficient transport to the target cells. To date, various delivery formulations have been developed from cationic lipids, polymers, and/or inorganic nanoparticles for systemic delivery of siRNA to solid tumors. This review describes the current status of clinical trials related to siRNA-based cancer therapy, as well as the remaining issues that need to be overcome to establish a successful therapy. It, then introduces various promising design strategies of delivery vehicles for stable and targeted siRNA delivery, including the prospects for future design.
Collapse
|
186
|
Che Harun NF, Takemoto H, Nomoto T, Tomoda K, Matsui M, Nishiyama N. Artificial Control of Gene Silencing Activity Based on siRNA Conjugation with Polymeric Molecule Having Coil-Globule Transition Behavior. Bioconjug Chem 2016; 27:1961-4. [PMID: 27506383 DOI: 10.1021/acs.bioconjchem.6b00322] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A new strategy for controlling gene silencing activity of siRNA in the cell was developed in the present study. siRNA was linearly conjugated with PNIPAAm, where coil-globule transition of the conjugated PNIPAAm allows thermoresponsive exposure of the vicinal siRNA molecule; a coil form of PNIPAAm (T < LCST) inhibits siRNA interaction with gene silencing-related proteins due to the steric hindrance effect, while a globule form of PNIPAAm (T > LCST) allows a ready access of siRNA to gene silencing pathway. As a result, at T > LCST, PNIPAAm-siRNA elicited effective association of siRNA with a gene silencing-related protein of Ago2, while siRNA recruitment into the gene silencing pathway was significantly suppressed at T < LCST. Ultimately, gene silencing efficacy of PNIPAAm-siRNA was close to unconjugated siRNA at T > LCST (∼80%), while it was dramatically decreased to ∼20% at T < LCST, suggesting that coil-globule transition of the conjugated polymer can control the bioactivity of the vicinal siRNA molecule.
Collapse
Affiliation(s)
- Noor Faizah Che Harun
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Hiroyasu Takemoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Keishiro Tomoda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , R1-11, 4259, Nagatsuta, Midori-Ku, Yokohama, Kanagawa 226-8503, Japan
| |
Collapse
|
187
|
Gooding M, Malhotra M, Evans JC, Darcy R, O'Driscoll CM. Oligonucleotide conjugates - Candidates for gene silencing therapeutics. Eur J Pharm Biopharm 2016; 107:321-40. [PMID: 27521696 DOI: 10.1016/j.ejpb.2016.07.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 11/18/2022]
Abstract
The potential therapeutic and diagnostic applications of oligonucleotides (ONs) have attracted great attention in recent years. The capability of ONs to selectively inhibit target genes through antisense and RNA interference mechanisms, without causing un-intended sideeffects has led them to be investigated for various biomedical applications, especially for the treatment of viral diseases and cancer. In recent years, many researchers have focused on enhancing the stability and target specificity of ONs by encapsulating/complexing them with polymers or lipid chains to formulate nanoparticles/nanocomplexes/micelles. Also, chemical modification of nucleic acids has emerged as an alternative to impart stability to ONs against nucleases and other degrading enzymes and proteins found in blood. In addition to chemically modifying the nucleic acids directly, another strategy that has emerged, involves conjugating polymers/peptide/aptamers/antibodies/proteins, preferably to the sense strand (3'end) of siRNAs. Conjugation to the siRNA not only enhances the stability and targeting specificity of the siRNA, but also allows for the development of self-administering siRNA formulations, with a much smaller size than what is usually observed for nanoparticle (∼200nm). This review concentrates mainly on approaches and studies involving ON-conjugates for biomedical applications.
Collapse
Affiliation(s)
- Matt Gooding
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - James C Evans
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Raphael Darcy
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | | |
Collapse
|
188
|
Aalinkeel R, Nair B, Chen C, Mahajan SD, Reynolds JL, Zhang H, Sun H, Sykes DE, Chadha KC, Turowski SG, Bothwell KD, Seshadri M, Cheng C, Schwartz SA. Nanotherapy silencing the interleukin-8 gene produces regression of prostate cancer by inhibition of angiogenesis. Immunology 2016; 148:387-406. [PMID: 27159450 PMCID: PMC4948039 DOI: 10.1111/imm.12618] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 04/05/2016] [Accepted: 05/03/2016] [Indexed: 01/12/2023] Open
Abstract
Interleukin-8 (IL-8) is a pro-angiogenic cytokine associated with aggressive prostate cancer (CaP). We detected high levels of IL-8 in sera from patients with CaP compared with healthy controls and patients with benign prostatic hypertrophy. This study examines the role of IL-8 in the pathogenesis of metastatic prostate cancer. We developed a biocompatible, cationic polylactide (CPLA) nanocarrier to complex with and efficiently deliver IL-8 small interfering RNA (siRNA) to CaP cells in vitro and in vivo. CPLA IL-8 siRNA nanocomplexes (nanoplexes) protect siRNA from rapid degradation, are non-toxic, have a prolonged lifetime in circulation, and their net positive charge facilitates penetration of cell membranes and subsequent intracellular trafficking. Administration of CPLA IL-8 siRNA nanoplexes to immunodeficient mice bearing human CaP tumours produced significant antitumour activities with no adverse effects. Systemic (intravenous) or local intra-tumour administration of IL-8 siRNA nanoplexes resulted in significant inhibition of CaP growth. Magnetic resonance imaging and ultrasonography of experimental animals demonstrated reduction of tumour perfusion in vivo following nanoplex treatment. Staining of tumour sections for CD31 confirmed significant damage to tumour neovasculature after nanoplex therapy. These studies demonstrate the efficacy of IL-8 siRNA nanotherapy for advanced, treatment-resistant human CaP.
Collapse
Affiliation(s)
- Ravikumar Aalinkeel
- Department of MedicineDivision of Allergy, Immunology and RheumatologyUniversity at Buffalo and Kaleida HealthBuffaloNYUSA
| | - Bindukumar Nair
- Department of MedicineDivision of Allergy, Immunology and RheumatologyUniversity at Buffalo and Kaleida HealthBuffaloNYUSA
| | - Chih‐Kuang Chen
- Department of Chemical and Biological EngineeringUniversity at BuffaloBuffaloNYUSA
| | - Supriya D. Mahajan
- Department of MedicineDivision of Allergy, Immunology and RheumatologyUniversity at Buffalo and Kaleida HealthBuffaloNYUSA
| | - Jessica L. Reynolds
- Department of MedicineDivision of Allergy, Immunology and RheumatologyUniversity at Buffalo and Kaleida HealthBuffaloNYUSA
| | - Hanguang Zhang
- Department of Chemical and Biological EngineeringUniversity at BuffaloBuffaloNYUSA
| | - Haotian Sun
- Department of Chemical and Biological EngineeringUniversity at BuffaloBuffaloNYUSA
| | - Donald E. Sykes
- Department of MedicineDivision of Allergy, Immunology and RheumatologyUniversity at Buffalo and Kaleida HealthBuffaloNYUSA
| | - Kailash C. Chadha
- Department of Molecular and Cellular BiologyRoswell Park Cancer InstituteBuffaloNYUSA
| | - Steven G. Turowski
- Department of Pharmacology and TherapeuticsCancer Cell Center Roswell Park Cancer InstituteBuffaloNYUSA
| | - Katelyn D. Bothwell
- Department of Pharmacology and TherapeuticsCancer Cell Center Roswell Park Cancer InstituteBuffaloNYUSA
| | - Mukund Seshadri
- Department of Pharmacology and TherapeuticsCancer Cell Center Roswell Park Cancer InstituteBuffaloNYUSA
| | - Chong Cheng
- Department of Chemical and Biological EngineeringUniversity at BuffaloBuffaloNYUSA
| | - Stanley A. Schwartz
- Department of MedicineDivision of Allergy, Immunology and RheumatologyUniversity at Buffalo and Kaleida HealthBuffaloNYUSA
| |
Collapse
|
189
|
Beckert L, Kostka L, Kessel E, Krhac Levacic A, Kostkova H, Etrych T, Lächelt U, Wagner E. Acid-labile pHPMA modification of four-arm oligoaminoamide pDNA polyplexes balances shielding and gene transfer activity in vitro and in vivo. Eur J Pharm Biopharm 2016; 105:85-96. [DOI: 10.1016/j.ejpb.2016.05.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 11/26/2022]
|
190
|
Liu Y, Chen C. Role of nanotechnology in HIV/AIDS vaccine development. Adv Drug Deliv Rev 2016; 103:76-89. [PMID: 26952542 DOI: 10.1016/j.addr.2016.02.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 12/25/2022]
Abstract
HIV/AIDS is one of the worst crises affecting global health and influencing economic development and social stability. Preventing and treating HIV infection is a crucial task. However, there is still no effective HIV vaccine for clinical application. Nanotechnology has the potential to solve the problems associated with traditional HIV vaccines. At present, various nano-architectures and nanomaterials can function as potential HIV vaccine carriers or adjuvants, including inorganic nanomaterials, liposomes, micelles and polymer nanomaterials. In this review, we summarize the current progress in the use of nanotechnology for the development of an HIV/AIDS vaccine and discuss its potential to greatly improve the solubility, permeability, stability and pharmacokinetics of HIV vaccines. Although nanotechnology holds great promise for applications in HIV/AIDS vaccines, there are still many inadequacies that result in a variety of risks and challenges. The potential hazards to the human body and environment associated with some nano-carriers, and their underlying mechanisms require in-depth study. Non-toxic or low-toxic nanomaterials with adjuvant activity have been identified. However, studying the confluence of factors that affect the adjuvant activity of nanomaterials may be more important for the optimization of the dosage and immunization strategy and investigations into the exact mechanism of action. Moreover, there are no uniform standards for investigations of nanomaterials as potential vaccine adjuvants. These limitations make it harder to analyze and deduce rules from the existing data. Developing vaccine nano-carriers or adjuvants with high benefit-cost ratios is important to ensure their broad usage. Despite some shortcomings, nanomaterials have great potential and application prospects in the fields of AIDS treatment and prevention.
Collapse
Affiliation(s)
- Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
191
|
Kim JB, Lee YM, Ryu J, Lee E, Kim WJ, Keum G, Bang EK. Coordinative Amphiphiles as Tunable siRNA Transporters. Bioconjug Chem 2016; 27:1850-6. [DOI: 10.1021/acs.bioconjchem.6b00260] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jin Bum Kim
- Center
for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yeong Mi Lee
- Center
for Self-Assembly and Complexity, Institute for Basic Science (IBS),
and Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang 37673, Republic of Korea
| | - Jooyeon Ryu
- Graduate
School of Analytical Science and Technology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Eunji Lee
- Graduate
School of Analytical Science and Technology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Won Jong Kim
- Center
for Self-Assembly and Complexity, Institute for Basic Science (IBS),
and Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang 37673, Republic of Korea
| | - Gyochang Keum
- Center
for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Eun-Kyoung Bang
- Center
for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
192
|
Sapag A, Irrazábal T, Lobos-González L, Muñoz-Brauning CR, Quintanilla ME, Tampier L. Hairpin Ribozyme Genes Curtail Alcohol Drinking: from Rational Design to in vivo Effects in the Rat. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e335. [PMID: 27404720 PMCID: PMC5330938 DOI: 10.1038/mtna.2016.41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/12/2016] [Indexed: 01/02/2023]
Abstract
Ribozyme genes were designed to reduce voluntary alcohol drinking in a rat model of alcohol dependence. Acetaldehyde generated from alcohol in the liver is metabolized by the mitochondrial aldehyde dehydrogenase (ALDH2) such that diminishing ALDH2 activity leads to the aversive effects of blood acetaldehyde upon alcohol intake. A stepwise approach was followed to design genes encoding ribozymes targeted to the rat ALDH2 mRNA. In vitro studies of accessibility to oligonucleotides identified suitable target sites in the mRNA, one of which fulfilled hammerhead and hairpin ribozyme requirements (CGGUC). Ribozyme genes delivered in plasmid constructs were tested in rat cells in culture. While the hairpin ribozyme reduced ALDH2 activity 56% by cleavage and blockade (P < 0.0001), the hammerhead ribozyme elicited minor effects by blockade. The hairpin ribozyme was tested in vivo by adenoviral gene delivery to UChB alcohol drinker rats. Ethanol intake was curtailed 47% for 34 days (P < 0.0001), while blood acetaldehyde more than doubled upon ethanol administration and ALDH2 activity dropped 25% in liver homogenates, not affecting other ALDH isoforms. Thus, hairpin ribozymes targeted to 16 nt in the ALDH2 mRNA provide durable and specific effects in vivo, representing an improvement on previous work and encouraging development of gene therapy for alcoholism.
Collapse
Affiliation(s)
- Amalia Sapag
- Laboratory of Gene Pharmacotherapy, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Thergiory Irrazábal
- Laboratory of Gene Pharmacotherapy, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Laboratory of Gene Pharmacotherapy, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Carlos R Muñoz-Brauning
- Laboratory of Gene Pharmacotherapy, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Programme, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lutske Tampier
- Molecular and Clinical Pharmacology Programme, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
193
|
Structure, activity and uptake mechanism of siRNA-lipid nanoparticles with an asymmetric ionizable lipid. Int J Pharm 2016; 510:350-8. [PMID: 27374199 DOI: 10.1016/j.ijpharm.2016.06.124] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/09/2016] [Accepted: 06/26/2016] [Indexed: 12/31/2022]
Abstract
Lipid nanoparticles (LNPs) represent the most advanced platform for the systemic delivery of siRNA. We have previously reported the discovery of novel ionizable lipids with asymmetric lipid tails, enabling potent gene-silencing activity in hepatocytes in vivo; however, the structure and delivery mechanism had not been elucidated. Here, we report the structure, activity and uptake mechanism of LNPs with an asymmetric ionizable lipid. Zeta potential and hemolytic activity of LNPs showed that LNPs were neutral at the pH of the blood compartment but become increasingly charged and fusogenic in the acidic endosomal compartment. (31)P NMR experiments indicated that the siRNA was less mobile inside particles, presumably because of an electrostatic interaction with an ionizable lipid. The role of Apolipoprotein E (apoE) was studied using recombinant human apoE both in vitro and in vivo. A comparative study in wild-type and apoE-deficient mice revealed that apoE significantly influenced the in vivo biodistribution of LNPs and enhanced the cellular uptake. Pretreatment of mice with siRNA targeting low-density lipoprotein receptor (LDLR) impaired gene-silencing of the following siRNA treatment, demonstrating that in vivo activity of LNPs is dependent on LDLR. Our studies on the detailed mechanism should lead to the creation of more sophisticated LNP-based RNAi therapeutics.
Collapse
|
194
|
Sato Y, Nakamura T, Yamada Y, Harashima H. Development of a multifunctional envelope-type nano device and its application to nanomedicine. J Control Release 2016; 244:194-204. [PMID: 27374187 DOI: 10.1016/j.jconrel.2016.06.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
Successful nanomedicines should be based on sound drug delivery systems (DDS) the permit intracellular trafficking as well as the biodistribution of cargos to be controlled. We have been developing new types of DDS that are multifunctional envelope-type nano devices referred to as MENDs. First, we will focus the in vivo delivery of siRNA to hepatocytes using a YSK-MEND which is composed of pH-responsive cationic lipids. The YSK-MEND is capable of inducing efficient silencing activity in hepatocytes and can be used to cure mice that are infected with hepatitis C or B. The YSK-MEND can also be applied to cancer immunotherapy through the activation of immune cells by delivering different compounds such as cyclic-di-GMP, siRNA or alpha-galactosylceramide as a lipid antigen. The findings indicate that, as predicted, these compounds, when encapsulated in the YSK-MEND, can be delivered to the site of action and induced immune activation through different mechanisms. Finally, a MITO-Porter, a membrane fusion-based delivery system to mitochondria, is introduced as an organelle targeting DDS and a new strategy for cancer therapy is proposed by delivering gentamicin to mitochondria of cancer cells. These new technologies are expected to extend the therapeutic area of Nanomedicine by increasing the power of DDS, especially from the view point of controlled intracellular trafficking.
Collapse
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
195
|
Reinhard S, Wagner E. How to Tackle the Challenge of siRNA Delivery with Sequence-Defined Oligoamino Amides. Macromol Biosci 2016; 17. [PMID: 27328447 DOI: 10.1002/mabi.201600152] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/24/2016] [Indexed: 12/31/2022]
Abstract
RNA interference (RNAi) as a mechanism of gene regulation provides exciting opportunities for medical applications. Synthetic small interfering RNA (siRNA) triggers the knockdown of complementary mRNA sequences in a catalytic fashion and has to be delivered into the cytosol of the targeted cells. The design of adequate carrier systems to overcome multiple extracellular and intracellular roadblocks within the delivery process has utmost importance. Cationic polymers form polyplexes through electrostatic interaction with negatively charged nucleic acids and present a promising class of carriers. Issues of polycations regarding toxicity, heterogeneity, and polydispersity can be overcome by solid-phase-assisted synthesis of sequence-defined cationic oligomers. These medium-sized highly versatile nucleic acid carriers display low cytotoxicity and can be modified and tailored in multiple ways to meet specific requirements of nucleic acid binding, polyplex size, shielding, targeting, and intracellular release of the cargo. In this way, sequence-defined cationic oligomers can mimic the dynamic and bioresponsive behavior of viruses.
Collapse
Affiliation(s)
- Sören Reinhard
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig Maximilians University, 81377, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig Maximilians University, 81377, Munich, Germany.,Nanosystems Initiative Munich (NIM), 80799, Munich, Germany
| |
Collapse
|
196
|
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: 51] [Impact Index Per Article: 5.7] [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
|
197
|
Haussecker D. Stacking up CRISPR against RNAi for therapeutic gene inhibition. FEBS J 2016; 283:3249-60. [PMID: 27090508 DOI: 10.1111/febs.13742] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/10/2016] [Accepted: 04/18/2016] [Indexed: 12/16/2022]
Abstract
Both RNA interference (RNAi) and clustered regularly-interspaced short palindromic repeats (CRISPR) technologies allow for the sequence-specific inhibition of gene function and therefore have the potential to be used as therapeutic modalities. By judging the current public and scientific journal interest, it would seem that CRISPR, by enabling clean, durable knockouts, will dominate therapeutic gene inhibition, also at the expense of RNAi. This review aims to look behind prevailing sentiments and to more clearly define the likely scope of the therapeutic applications of the more recently developed CRISPR technology and its relative strengths and weaknesses with regards to RNAi. It is found that largely because of their broadly overlapping delivery constraints, while CRISPR presents formidable competition for DNA-directed RNAi strategies, its impact on RNAi therapeutics triggered by synthetic oligonucleotides will likely be more moderate. Instead, RNAi and genome editing, and in particular CRISPR, are poised to jointly promote a further shift toward sequence-targeted precision medicines.
Collapse
|
198
|
PEGylation of 6-amino-6-deoxy-curdlan for efficient in vivo siRNA delivery. Carbohydr Polym 2016; 141:92-8. [DOI: 10.1016/j.carbpol.2015.12.077] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/22/2015] [Accepted: 12/29/2015] [Indexed: 11/19/2022]
|
199
|
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: 591] [Impact Index Per Article: 65.7] [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
|
200
|
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
|