201
|
Using two-fluid nozzle for spray freeze drying to produce porous powder formulation of naked siRNA for inhalation. Int J Pharm 2018; 552:67-75. [PMID: 30244146 DOI: 10.1016/j.ijpharm.2018.09.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/01/2018] [Accepted: 09/18/2018] [Indexed: 01/05/2023]
Abstract
Spray freeze drying is an attractive technology to produce powder formulation for inhalation. It can be used to generate large porous particles which tend to aerosolize efficiently and do not aggregate readily. It also avoids material to be exposed to elevated temperature. In this study, we reported the use of two-fluid nozzle to produce spray freeze dried powder of small interfering RNA (siRNA). The effect of atomization gas flow rate and liquid feed rate were inspected initially using herring sperm DNA (hsDNA) as nucleic acid model. The atomization gas flow rate was found to have a major impact on the aerosol properties. The higher the atomization gas flow rate, the smaller the particle size, the higher the fine particle fraction (FPF). In contrast, the liquid feed rate had very minor effect. Subsequently, spray freeze dried siRNA powder was produced at various atomization gas flow rates. The particles produced were highly porous as examined with the scanning electron microscopy, and the structural integrity of the siRNA was demonstrated with gel electrophoresis. The gene-silencing effect of the siRNA was also successfully preserved in vitro. The best performing siRNA formulation was prepared at the highest atomization gas flow rate investigated with a moderate FPF of 30%. However, this was significantly lower than that obtained with the corresponding hsDNA counterparts (FPF ∼57%). A direct comparison between the hsDNA and siRNA formulations revealed that the former exhibited a lower density, hence a smaller aerodynamic diameter despite similar geometric size.
Collapse
|
202
|
Islam MA, Xu Y, Tao W, Ubellacker JM, Lim M, Aum D, Lee GY, Zhou K, Zope H, Yu M, Cao W, Oswald JT, Dinarvand M, Mahmoudi M, Langer R, Kantoff PW, Farokhzad OC, Zetter BR, Shi J. Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat Biomed Eng 2018; 2:850-864. [PMID: 31015614 PMCID: PMC6486184 DOI: 10.1038/s41551-018-0284-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 07/30/2018] [Indexed: 01/06/2023]
Abstract
PTEN is a well-characterized tumour-suppressor gene that is lost or mutated in about half of metastatic castration-resistant prostate cancers and in many other human cancers. The restoration of functional PTEN as a treatment for prostate cancer has however proven difficult. Here, we show that PTEN mRNA can be reintroduced into PTEN-null prostate cancer cells in vitro and in vivo via its encapsulation in polymer-lipid hybrid nanoparticles coated with a poly(ethylene glycol) shell. The nanoparticles are stable in serum, elicit low toxicity, enable high PTEN mRNA transfection in prostate cancer cells, and lead to significant inhibition of tumour growth when delivered systemically in multiple mouse models of prostate cancer. We also show that the restoration of PTEN function in PTEN-null prostate cancer cells inhibits the PI3K-AKT pathway and enhances apoptosis. Our findings provide proof-of-principle evidence of the restoration of mRNA-based tumour suppression in vivo.
Collapse
Affiliation(s)
- Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Oncology Division, Immunomic Therapeutics, Inc., Rockville, MD, USA
| | - Yingjie Xu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessalyn M Ubellacker
- Hematology Division, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael Lim
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Nanotechnology Engineering Program, University of Waterloo, Waterloo, Ontario, Canada
| | - Daniel Aum
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gha Young Lee
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kun Zhou
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Harshal Zope
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikyung Yu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wuji Cao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Nanotechnology Engineering Program, University of Waterloo, Waterloo, Ontario, Canada
| | - James Trevor Oswald
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Nanotechnology Engineering Program, University of Waterloo, Waterloo, Ontario, Canada
| | - Meshkat Dinarvand
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Morteza Mahmoudi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Bruce R Zetter
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
203
|
Zhang D, Lee H, Wang X, Rai A, Groot M, Jin Y. Exosome-Mediated Small RNA Delivery: A Novel Therapeutic Approach for Inflammatory Lung Responses. Mol Ther 2018; 26:2119-2130. [PMID: 30005869 PMCID: PMC6127502 DOI: 10.1016/j.ymthe.2018.06.007] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 02/08/2023] Open
Abstract
Exosomes (EXOs) are a type of extracellular nanovesicles released from living cells. Accumulating evidence suggests that EXOs are involved in the pathogenesis of human diseases, including lung conditions. In recent years, the potential of EXO-mediated drug delivery has gained increasing interest. In this report, we investigated whether inhaled EXOs serve as an efficient and practical delivery vehicle to activate or inhibit alveolar macrophages (AMs), subsequently modulating pulmonary immune responses. We first identified the recipient cells of the inhaled EXOs, which were labeled with PKH26. We found that only lung macrophages efficiently take up intratracheally instilled EXOs in vivo. Using modified calcium chloride-mediated transformation, we manipulated small RNA molecules in serum-derived EXOs, including siRNAs, microRNA (miRNA) mimics, and miRNA inhibitors. Via intratracheal instillation, we successfully delivered siRNA and miRNA mimics or inhibitors into lung macrophages using the serum-derived EXOs as vehicles. Furthermore, EXO siRNA or miRNA molecules are functional in modulating LPS-induced lung inflammation in vivo. Beneficially, serum-derived EXOs themselves do not trigger lung immune responses, adding more favorable features to serve as drug delivery agents. Collectively, we developed a novel protocol using serum-derived EXOs to deliver designated small RNA molecules into lung macrophages in vivo, potentially shedding light on future gene therapy of human lung diseases.
Collapse
Affiliation(s)
- Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Xiaoyun Wang
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Ashish Rai
- North Shore Medical Center, Salem Hospital, Boston, MA 01970, USA
| | - Michael Groot
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA.
| |
Collapse
|
204
|
Glycogen-nucleic acid constructs for gene silencing in multicellular tumor spheroids. Biomaterials 2018; 176:34-49. [DOI: 10.1016/j.biomaterials.2018.05.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/01/2018] [Accepted: 05/14/2018] [Indexed: 12/29/2022]
|
205
|
Chang D, Lim M, Goos JACM, Qiao R, Ng YY, Mansfeld FM, Jackson M, Davis TP, Kavallaris M. Biologically Targeted Magnetic Hyperthermia: Potential and Limitations. Front Pharmacol 2018; 9:831. [PMID: 30116191 PMCID: PMC6083434 DOI: 10.3389/fphar.2018.00831] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022] Open
Abstract
Hyperthermia, the mild elevation of temperature to 40–43°C, can induce cancer cell death and enhance the effects of radiotherapy and chemotherapy. However, achievement of its full potential as a clinically relevant treatment modality has been restricted by its inability to effectively and preferentially heat malignant cells. The limited spatial resolution may be circumvented by the intravenous administration of cancer-targeting magnetic nanoparticles that accumulate in the tumor, followed by the application of an alternating magnetic field to raise the temperature of the nanoparticles located in the tumor tissue. This targeted approach enables preferential heating of malignant cancer cells whilst sparing the surrounding normal tissue, potentially improving the effectiveness and safety of hyperthermia. Despite promising results in preclinical studies, there are numerous challenges that must be addressed before this technique can progress to the clinic. This review discusses these challenges and highlights the current understanding of targeted magnetic hyperthermia.
Collapse
Affiliation(s)
- David Chang
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.,Department of Radiation Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Sydney, NSW, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, Australia
| | - May Lim
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Jeroen A C M Goos
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ruirui Qiao
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Yun Yee Ng
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Friederike M Mansfeld
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Michael Jackson
- Department of Radiation Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Chemistry, University of Warwick, Coventry, United Kingdom
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
206
|
Zhao R, Liang X, Zhao B, Chen M, Liu R, Sun S, Yue X, Wang S. Ultrasound assisted gene and photodynamic synergistic therapy with multifunctional FOXA1-siRNA loaded porphyrin microbubbles for enhancing therapeutic efficacy for breast cancer. Biomaterials 2018; 173:58-70. [DOI: 10.1016/j.biomaterials.2018.04.054] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/20/2022]
|
207
|
Herrera VLM, Colby AH, Ruiz-Opazo N, Coleman DG, Grinstaff MW. Nucleic acid nanomedicines in Phase II/III clinical trials: translation of nucleic acid therapies for reprogramming cells. Nanomedicine (Lond) 2018; 13:2083-2098. [PMID: 30204054 PMCID: PMC6219437 DOI: 10.2217/nnm-2018-0122] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/14/2018] [Indexed: 01/01/2023] Open
Abstract
This review presents an integrated analysis of the current-state-of-the-art in nucleic acid nanotherapies and highlights the importance of nanotechnology in the delivery of nucleic acid therapies. While there is no one dominant nanodesign, the diversity of nanodesigns and delivery of different siRNAs, miRNA and DNA to inhibit more than 20 targets in seven disease states in Phase II/III clinical trials reflects the potential of nucleic acid therapies to treat intractable diseases and non-druggable targets. We provide benchmarks to aid in comparing the design, proof-of-concept studies and clinical trials. From this, we demonstrate the importance of generating a strategic framework for integrating clinical 'wish lists' for a means to treat intractable diseases with engineering 'design checklists' for nucleic acid nanotherapies.
Collapse
Affiliation(s)
- Victoria LM Herrera
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Aaron H Colby
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Nelson Ruiz-Opazo
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - David G Coleman
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mark W Grinstaff
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Department of Chemistry, Boston University, Boston, MA 02215, USA
| |
Collapse
|
208
|
Wang R, Degirmenci V, Xin H, Li Y, Wang L, Chen J, Hu X, Zhang D. PEI-Coated Fe₃O₄ Nanoparticles Enable Efficient Delivery of Therapeutic siRNA Targeting REST into Glioblastoma Cells. Int J Mol Sci 2018; 19:ijms19082230. [PMID: 30065155 PMCID: PMC6121642 DOI: 10.3390/ijms19082230] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/21/2018] [Accepted: 07/30/2018] [Indexed: 01/05/2023] Open
Abstract
Glioblastomas (GBM) are the most frequent brain tumors lacking efficient treatment. The increasingly elucidated gene targets make siRNA-based gene therapy a promising anticancer approach, while an efficient delivery system is urgently needed. Here, polyethyleneimine (PEI)-coated Fe₃O₄ nanoparticles (NPs) have been developed and applied for siRNA delivery into GBM cells to silence repressor element 1-silencing transcription factor (REST). The prepared PEI-coated Fe₃O₄ NPs were characterized as magnetic nanoparticles with a positive charge, by transmission electronic microscopy, dynamic light-scattering analysis and a magnetometer. By gel retardation assay, the nanoparticles were found to form complexes with siRNA and the interaction proportion of NP to siRNA was 2.8:1. The cellular uptake of NP/siRNA complexes was verified by prussian blue staining, fluorescence labeling and flow cytometry in U-87 and U-251 GBM cells. Furthermore, the REST silencing examined by realtime polymerase chain reaction (PCR) and Western blotting presented significant reduction of REST in transcription and translation levels. Upon the treatment of NP/siRNA targeting REST, the GBM cell viabilities were inhibited and the migration capacities were repressed remarkably, analyzed by cell counting kit-8 and transwell assay separately. In this study, we demonstrated the PEI-coated Fe₃O₄ nanoparticle as a vehicle for therapeutic siRNA delivery, at an appropriate NP/siRNA weight ratio for REST silencing in GBM cells, inhibiting cell proliferation and migration efficiently. These might represent a novel potential treatment strategy for GBM.
Collapse
Affiliation(s)
- Rui Wang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| | | | - Hongchuan Xin
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China.
| | - Ying Li
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| | - Liping Wang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| | - Jiayu Chen
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| | - Xiaoyu Hu
- College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| | - Dianbao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China.
| |
Collapse
|
209
|
Sun J, Kormakov S, Liu Y, Huang Y, Wu D, Yang Z. Recent Progress in Metal-Based Nanoparticles Mediated Photodynamic Therapy. Molecules 2018; 23:E1704. [PMID: 30002333 PMCID: PMC6099795 DOI: 10.3390/molecules23071704] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 12/18/2022] Open
Abstract
Photodynamic therapy (PDT) is able to non-invasively treat and diagnose various cancers and nonmalignant diseases by combining light, oxygen, and photosensitizers (PSs). However, the application of PDT is hindered by poor water solubility and limited light-penetration depth of the currently available photosensitizers (PSs). Water solubility of PSs is crucial for designing pharmaceutical formulation and administration routes. Wavelength of light source at visible range normally has therapeutic depth less than 1 mm. In this review, focus is on the recent research progress of metal-based nanoparticles being applied in PDT. The potential toxicity of these nanoscales and future directions are further discussed.
Collapse
Affiliation(s)
- Jingyao Sun
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | - Semen Kormakov
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ying Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing 100029, China.
| | - Yao Huang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Daming Wu
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing 100029, China.
| | - Zhaogang Yang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
210
|
Xue F, Wang Y, Zhang Q, Han S, Zhang F, Jin T, Li C, Hu H, Zhang J. Self-assembly of affinity-controlled nanoparticles via host-guest interactions for drug delivery. NANOSCALE 2018; 10:12364-12377. [PMID: 29682667 DOI: 10.1039/c8nr01518j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
There has been increasing interest in constructing affinity-based drug delivery systems via different non-covalent interactions. Herein we report a host-guest interaction-based strategy to develop effective drug delivery systems using cyclodextrin-containing copolymers. Hydrophilic copolymers with one polyethylene glycol block and another block containing either α-cyclodextrin or β-cyclodextrin were synthesized. Using poly(β-benzyl l-aspartate) and pyrene as model guest compounds, we demonstrated the nanoparticle formation by host-guest interaction-mediated self-assembly. When an antioxidant and anti-inflammatory drug Tempol was used, the formation of well-defined spherical nanoparticles and therapeutic loading can be simultaneously realized. The obtained nanotherapy showed affinity-controlled drug release. In vitro cell culture experiments suggested that the host-guest nanotherapy exhibited desirable antioxidant and anti-inflammatory effects in macrophages. In a mouse model of an inflammatory disease ulcerative colitis, the orally administered host-guest nanoparticle can be effectively accumulated in the inflamed colonic tissue. Oral treatment of mice bearing colitis with the nanotherapy led to significantly improved efficacy in comparison with free drugs. A good in vivo safety profile was also observed for the developed host-guest nanotherapy. Accordingly, these types of affinity nanoparticles based on CD-containing copolymers can function as effective nanoplatforms for targeted treatment of a plethora of diseases.
Collapse
Affiliation(s)
- Fangchao Xue
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Sahu A, Choi WI, Tae G. Recent Progress in the Design of Hypoxia-Specific Nano Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| | - Won Il Choi
- Center for Convergence Bioceramic Materials; Convergence R&D Division; Korea Institute of Ceramic Engineering and Technology; 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu Cheongju Chungbuk 28160 Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| |
Collapse
|
212
|
Qin SY, Cheng YJ, Lei Q, Zhang AQ, Zhang XZ. Combinational strategy for high-performance cancer chemotherapy. Biomaterials 2018; 171:178-197. [DOI: 10.1016/j.biomaterials.2018.04.027] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 04/10/2018] [Accepted: 04/14/2018] [Indexed: 12/21/2022]
|
213
|
Abstract
Alzheimer disease (AD) is the most common form of dementia. Pathologically, AD is characterized by amyloid plaques and neurofibrillary tangles in the brain, with associated loss of synapses and neurons, resulting in cognitive deficits and eventually dementia. Amyloid-β (Aβ) peptide and tau protein are the primary components of the plaques and tangles, respectively. In the decades since Aβ and tau were identified, development of therapies for AD has primarily focused on Aβ, but tau has received more attention in recent years, in part because of the failure of various Aβ-targeting treatments in clinical trials. In this article, we review the current status of tau-targeting therapies for AD. Initially, potential anti-tau therapies were based mainly on inhibition of kinases or tau aggregation, or on stabilization of microtubules, but most of these approaches have been discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting therapies in clinical trials are immunotherapies, which have shown promise in numerous preclinical studies. Given that tau pathology correlates better with cognitive impairments than do Aβ lesions, targeting of tau is expected to be more effective than Aβ clearance once the clinical symptoms are evident. With future improvements in diagnostics, these two hallmarks of the disease might be targeted prophylactically.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
214
|
Zheng X, Zhang F, Zhao Y, Zhang J, Dawulieti J, Pan Y, Cui L, Sun M, Shao D, Li M, He K, Zhang M, Li J, Chen L. Self-assembled dual fluorescence nanoparticles for CD44-targeted delivery of anti-miR-27a in liver cancer theranostics. Am J Cancer Res 2018; 8:3808-3823. [PMID: 30083261 PMCID: PMC6071528 DOI: 10.7150/thno.25255] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
Despite the vital role miRNA-27a plays in driving the development and progress of liver cancer, miRNA-based inhibition therapy is hampered due to its undesired degradation and off-target effects. Herein, a multifunctional nanoparticle for noninvasive tracking of targeted delivery of anti-miR-27a oligonucleotides against liver cancer was constructed. Methods: Dual-fluorescent conjugates (QD-HA-PEI) were first fabricated through crosslinking hyaluronic acid (HA), polyethyleneimine (PEI) and near-infrared (NIR) fluorescent quantum dots (QDs) via a facile one-pot approach. Antisense oligonucleotide was then encapsulated by QD-HA-PEI to form anti-miR-27a/QD-HA-PEI via electrostatic interactions. Targeting, biodistribution, bioimaging, in vitro cytotoxicity and in vivo anti-tumor effects were evaluated and the underlying mechanism was studied. Results: The NIR fluorescence of anti-miR-27a/QD-HA-PEI could be employed to monitor CD44 receptor-targeted cellular uptake and tumor accumulation. Importantly, the intrinsic fluorescence of anti-miR-27a/QD-HA-PEI remained in the “ON” state in extracellular or blood environment, but switched to the “OFF” state in the intracellular environment, indicating pH-responsive oligonucleotide release. Furthermore, anti-miR-27a/QD-HA-PEI exhibited effective and selective anti-cancer effects in vitro and in vivo with fewer side effects via the direct down-regulation of oncogenic transcription factors FOXO1 and PPAR-γ. Conclusion: Our findings validate the dual-fluorescent nanoparticles as delivery vectors of therapeutic miRNA, capable of simultaneous tumor imaging and tracking of miRNA-based modulation therapy, thereby providing an efficient and safe approach for liver cancer theranostics.
Collapse
|
215
|
Zhang QY, Ho PY, Tu MJ, Jilek JL, Chen QX, Zeng S, Yu AM. Lipidation of polyethylenimine-based polyplex increases serum stability of bioengineered RNAi agents and offers more consistent tumoral gene knockdown in vivo. Int J Pharm 2018; 547:537-544. [PMID: 29894758 DOI: 10.1016/j.ijpharm.2018.06.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/04/2018] [Accepted: 06/08/2018] [Indexed: 01/30/2023]
Abstract
Recently we have established a novel approach to produce bioengineered noncoding RNA agents (BERAs) in living cells that carry target RNAi molecules (e.g., siRNA and miRNA) and thus act as "prodrugs". Using GFP-siRNA-loaded BERA (BERA/GFP-siRNA) as a model molecule, this study was to define the in vitro and in vivo knockdown efficiency of BERAs delivered by liposome-polyethylenimine nanocomplex (lipopolyplex or LPP). Compared to in vivo-jetPEI® (IVJ-PEI) and polyplex formulations, LPP offered greater protection of BERA/GFP-siRNA against degradation by serum RNases. Particle sizes and zeta potentials of LPP nanocomplex remained stable over 28 days when stored at 4 °C. Furthermore, comparable levels of BERA/GFP-siRNA were delivered by LPP and IVJ-PEI to luciferase/GFP-expressing human SK-Hep1-Luc-GFP or A549-Luc-GFP cells, which were selectively processed into target GFP-siRNA and subsequently knocked down GFP mRNA and protein levels. In addition, LPP-carried BERA/GFP-siRNA was successfully delivered into xenograft tumors and offered more consistent knockdown of tumoral GFP mRNA level in an orthotopic hepatocellular carcinoma (HCC) SK-Hep1-Luc-GFP xenograft mouse model, while IVJ-PEI formulation showed larger variation. These findings demonstrated that lipidation of polyplexes improved serum stability of biologic RNAi molecules, which was efficiently delivered to orthotopic HCC tissues to knock down target gene expression.
Collapse
Affiliation(s)
- Qian-Yu Zhang
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Pui Yan Ho
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Mei-Juan Tu
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph L Jilek
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Qiu-Xia Chen
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ai-Ming Yu
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
216
|
Diaz-Cañestro C, Merlini M, Bonetti NR, Liberale L, Wüst P, Briand-Schumacher S, Klohs J, Costantino S, Miranda M, Schoedon-Geiser G, Kullak-Ublick GA, Akhmedov A, Paneni F, Beer JH, Lüscher TF, Camici GG. Sirtuin 5 as a novel target to blunt blood–brain barrier damage induced by cerebral ischemia/reperfusion injury. Int J Cardiol 2018; 260:148-155. [DOI: 10.1016/j.ijcard.2017.12.060] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 10/25/2022]
|
217
|
MacLeod AR, Crooke ST. RNA Therapeutics in Oncology: Advances, Challenges, and Future Directions. J Clin Pharmacol 2018; 57 Suppl 10:S43-S59. [PMID: 28921648 DOI: 10.1002/jcph.957] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
RNA-based therapeutic technologies represent a rapidly expanding class of therapeutic opportunities with the power to modulate cellular biology in ways never before possible. With RNA-targeted therapeutics, inhibitors of previously undruggable proteins, gene expression modulators, and even therapeutic proteins can be rationally designed based on sequence information alone, something that is not possible with other therapeutic modalities. The most advanced RNA therapeutic modalities are antisense oligonucleotides (ASOs) and small interfering RNAs. Particularly with ASOs, recent clinical data have demonstrated proof of mechanism and clinical benefit with these approaches across several nononcology disease areas by multiple routes of administration. In cancer, next-generation ASOs have recently demonstrated single-agent activity in patients with highly refractory cancers. Here we discuss advances in RNA therapeutics for the treatment of cancer and the challenges that remain to solidify these as mainstay therapeutic modalities to bridge the pharmacogenomic divide that remains in cancer drug discovery.
Collapse
Affiliation(s)
- A Robert MacLeod
- Vice President, Oncology Discovery, Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Stanley T Crooke
- CEO and Chairman of the Board, Ionis Pharmaceuticals, Carlsbad, CA, USA
| |
Collapse
|
218
|
Liu L, Liu Y, Xu B, Liu C, Jia Y, Liu T, Fang C, Wang W, Ren J, He Z, Men K, Liang X, Luo M, Shao B, Mao Y, Xiao H, Qian Z, Geng J, Dong B, Mi P, Jiang Y, Wei Y, Wei X. Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C + monocytes. Theranostics 2018; 8:3138-3152. [PMID: 29896308 PMCID: PMC5996362 DOI: 10.7150/thno.21693] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 04/13/2018] [Indexed: 02/05/2023] Open
Abstract
Rationale: Cationic nanocarriers present with well-known toxicities, including inflammatory toxicity, which limit their clinical application. How the cationic nanocarrier-induced inflammatory response is negatively regulated is unknown. Herein, we found that following a sublethal dose of cationic nanocarriers, the induced inflammatory response is characterized by early neutrophil infiltration and spontaneous resolution within 1 week. Methods: C57BL/6 mice were intravenously injected with a dosage of 1-100 mg/kg cationic DOTAP liposomes as well as other cationic materials. Cell necrosis was detected by flow cytometry. Release of mitochondrial DNA was quantified by qPCR via Taqman probes. Signal proteins were detected by Western blotting. PGE2 production in the supernatant was quantitated using an enzyme immunoassay (EIA). The infiltrated inflammatory cells were observed in WT mice, Ccr2-/- mice, Sting-/-mice and Tlr9-/-mice. Results: The early stage (24-48 h) inflammatory neutrophil infiltration was followed by an increasing percentage of monocytes; and, compared with WT mice, Ccr2-/- mice presented with more severe pulmonary inflammation. A previously uncharacterized population of regulatory monocytes expressing both inflammatory and immunosuppressive cytokines was identified in this model. The alteration in monocyte phenotype was directly induced by mtDNA release from cationic nanocarrier-induced necrotic cells via a STING- or TLR9-dependent pathway. Neutrophil activation was specifically inhibited by PGE2 from Ly6C+ inflammatory monocytes, and intravenous injections of dual-phenotype monocytes beneficially modified the immune response; this inhibitory effect was abolished after treatment with indomethacin. Moreover, we provide clear evidence that mitochondrial DNA activated Ly6C+ monocytes and increased PGE2 production through TLR9- or STING-mediated MAPK-NF-κB-COX2 pathways. Conclusion: Our findings suggest that Ly6C+ monocytes and mtDNA-induced Ly6C+ monocyte PGE2 production may be part of a feedback mechanism that contributes to the resolution of cationic nanocarrier-induced inflammatory toxicity and may have important implications for understanding nanoparticle biocompatibility and designing better, safer drug delivery systems.
Collapse
|
219
|
Sun W, Chen X, Xie C, Wang Y, Lin L, Zhu K, Shuai X. Co-Delivery of Doxorubicin and Anti-BCL-2 siRNA by pH-Responsive Polymeric Vector to Overcome Drug Resistance in In Vitro and In Vivo HepG2 Hepatoma Model. Biomacromolecules 2018; 19:2248-2256. [PMID: 29690766 DOI: 10.1021/acs.biomac.8b00272] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Drug resistance, developed through multiple mechanisms, is a major hindrance to successful chemotherapy of tumor. Combination therapy of chemotherapeutic drugs and siRNA represents an emerging strategy which may improve anticancer effect by synergistic actions. In this study, triblock copolymer of poly(ethylene glycol)- block-poly(l-lysine)- block-poly aspartyl ( N-( N', N'-diisopropylaminoethyl)) (PEG-PLL-PAsp(DIP)) was synthesized for the first time to enable the codelivery of BCL-2 siRNA and DOX. The system is supposed to not only bypass drug efflux but also down-regulate the antiapoptotic gene and consequently confronting against chemoresistance as well. Moreover, the pH responsive ability of the codelivery system can prevent drug leakage during circulation and guarantee swift drug release at tumors. The codelivered siRNA serves to suppress the expression of antiapoptotic BCL-2 and hence sensitize the cancer cells to anticancer drugs and produce improved therapeutic effect. Consequently, the codelivery of BCL-2 siRNA and anticancer drug DOX serves as a promising strategy against drug resistance in chemotherapy.
Collapse
Affiliation(s)
- Weitong Sun
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-Sen University , Guangzhou , 510275 , China.,Department of Biomedical Engineering, School of Engineering , Sun Yat-sen University , Guangzhou , 510006 , China.,Pharmaceutical College of Jiamusi University , Jiamusi , 154007 , China
| | - Xiaoyan Chen
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-Sen University , Guangzhou , 510275 , China
| | - Chao Xie
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-Sen University , Guangzhou , 510275 , China
| | - Yong Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-Sen University , Guangzhou , 510275 , China
| | - Liteng Lin
- Department of Minimally Invasive Interventional Radiology, and Department of Radiology , the Second Affiliated Hospital of Guangzhou Medical University , Guangzhou , 510260 , China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Department of Radiology , the Second Affiliated Hospital of Guangzhou Medical University , Guangzhou , 510260 , China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering , Sun Yat-Sen University , Guangzhou , 510275 , China
| |
Collapse
|
220
|
Fuertes A, Juanes M, Granja JR, Montenegro J. Supramolecular functional assemblies: dynamic membrane transporters and peptide nanotubular composites. Chem Commun (Camb) 2018. [PMID: 28636028 DOI: 10.1039/c7cc02997g] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The fabrication of functional molecular devices constitutes one of the most important current challenges for chemical sciences. The complex processes accomplished by living systems continuously demand the assistance of non-covalent interactions between molecular building blocks. Additionally, these building blocks (proteins, membranes, nucleotides) are also constituted by self-assembled structures. Therefore, supramolecular chemistry is the discipline required to understand the properties of the minimal self-assembled building blocks of living systems and to develop new functional smart materials. In the first part of this feature article, we highlight selected examples of the preparation of supramolecular membrane transporters with special emphasis on the application of dynamic covalent bonds. In the second section of the paper we review recent breakthroughs in the preparation of peptide nanotube hybrids with functional applications. The development of these devices constitutes an exciting process from where we can learn how to understand and manipulate supramolecular functional assemblies.
Collapse
Affiliation(s)
- Alberto Fuertes
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | | | | | | |
Collapse
|
221
|
Harrison EB, Azam SH, Pecot CV. Targeting Accessories to the Crime: Nanoparticle Nucleic Acid Delivery to the Tumor Microenvironment. Front Pharmacol 2018; 9:307. [PMID: 29670528 PMCID: PMC5893903 DOI: 10.3389/fphar.2018.00307] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/16/2018] [Indexed: 12/18/2022] Open
Abstract
Nucleic acid delivery for cancer holds extraordinary promise. Increasing expression of tumor suppressor genes or inhibition of oncogenes in cancer cells has important therapeutic potential. However, several barriers impair progress in cancer gene delivery. These include effective delivery to cancer cells and relevant intracellular compartments. Although viral gene delivery can be effective, it has the disadvantages of being immuno-stimulatory, potentially mutagenic and lacking temporal control. Various nanoparticle (NP) platforms have been developed to overcome nucleic acid delivery hurdles, but several challenges still exist. One such challenge has been the accumulation of NPs in non-cancer cells within the tumor microenvironment (TME) as well as the circulation. While uptake by these cancer-associated cells is considered to be an off-target effect in some contexts, several strategies have now emerged to utilize NP-mediated gene delivery to intentionally alter the TME. For example, the similarity of NPs in shape and size to pathogens promotes uptake by antigen presenting cells, which can be used to increase immune stimulation and promote tumor killing by T-lymphocytes. In the era of immunotherapy, boosting the ability of the immune system to eliminate cancer cells has proven to be an exciting new area in cancer nanotechnology. Given the importance of cancer-associated cells in tumor growth and metastasis, targeting these cells in the TME opens up new therapeutic applications for NPs. This review will cover evidence for non-cancer cell accumulation of NPs in animal models and patients, summarize characteristics that promote NP delivery to different cell types, and describe several therapeutic strategies for gene modification within the TME.
Collapse
Affiliation(s)
- Emily B. Harrison
- Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Salma H. Azam
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
222
|
Recent advances in siRNA delivery for cancer therapy using smart nanocarriers. Drug Discov Today 2018; 23:900-911. [DOI: 10.1016/j.drudis.2018.01.042] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/07/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022]
|
223
|
Miao T, Wang J, Zeng Y, Liu G, Chen X. Polysaccharide-Based Controlled Release Systems for Therapeutics Delivery and Tissue Engineering: From Bench to Bedside. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700513. [PMID: 29721408 PMCID: PMC5908359 DOI: 10.1002/advs.201700513] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/19/2017] [Indexed: 05/08/2023]
Abstract
Polysaccharides or polymeric carbohydrate molecules are long chains of monosaccharides that are linked by glycosidic bonds. The naturally based structural materials are widely applied in biomedical applications. This article covers four different types of polysaccharides (i.e., alginate, chitosan, hyaluronic acid, and dextran) and emphasizes their chemical modification, preparation approaches, preclinical studies, and clinical translations. Different cargo fabrication techniques are also presented in the third section. Recent progresses in preclinical applications are then discussed, including tissue engineering and treatment of diseases in both therapeutic and monitoring aspects. Finally, clinical translational studies with ongoing clinical trials are summarized and reviewed. The promise of new development in nanotechnology and polysaccharide chemistry helps clinical translation of polysaccharide-based drug delivery systems.
Collapse
Affiliation(s)
- Tianxin Miao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- Collaborative Innovation Center of Guangxi Biological Medicine and theMedical and Scientific Research CenterGuangxi Medical UniversityNanning530021China
| | - Yun Zeng
- Department of PharmacologyXiamen Medical CollegeXiamen361008China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell BiologySchool of Life SciencesXiamen UniversityXiamen361102China
- State Key Laboratory of Physical Chemistry of Solid Surfaces and The MOE Key Laboratory of Spectrochemical Analysis & InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and NanomedicineNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
224
|
Nikam RR, Gore KR. Journey of siRNA: Clinical Developments and Targeted Delivery. Nucleic Acid Ther 2018; 28:209-224. [PMID: 29584585 DOI: 10.1089/nat.2017.0715] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Since the evolutionary discovery of RNA interference and its utilization for gene knockdown in mammalian cell, a remarkable progress has been achieved in small interfering RNA (siRNA) therapeutics. siRNA is a promising tool, utilized as therapeutic agent against various diseases. Despite its significant potential benefits, safe, efficient, and target oriented delivery of siRNA is one of the major challenges in siRNA therapeutics. This review covers major achievements in clinical trials and targeted delivery of siRNAs using various targeting ligand-receptor pair. Local and systemically administered siRNA drug candidates at various phases in clinical trials are described in this review. This review also provides a deep insight in development of targeted delivery of siRNA. Various targeting ligand-siRNA pair with complexation and conjugation approaches are discussed in this review. This will help to achieve further optimization and development in targeted delivery of siRNAs to achieve higher gene silencing efficiency with lowest siRNA dose availability.
Collapse
Affiliation(s)
| | - Kiran R Gore
- Department of Chemistry, University of Mumbai , Mumbai, India
| |
Collapse
|
225
|
Shen J, Zhang W, Qi R, Mao ZW, Shen H. Engineering functional inorganic-organic hybrid systems: advances in siRNA therapeutics. Chem Soc Rev 2018; 47:1969-1995. [PMID: 29417968 PMCID: PMC5861001 DOI: 10.1039/c7cs00479f] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer treatment still faces a lot of obstacles such as tumor heterogeneity, drug resistance and systemic toxicities. Beyond the traditional treatment modalities, exploitation of RNA interference (RNAi) as an emerging approach has immense potential for the treatment of various gene-caused diseases including cancer. The last decade has witnessed enormous research and achievements focused on RNAi biotechnology. However, delivery of small interference RNA (siRNA) remains a key challenge in the development of clinical RNAi therapeutics. Indeed, functional nanomaterials play an important role in siRNA delivery, which could overcome a wide range of sequential physiological and biological obstacles. Nanomaterial-formulated siRNA systems have potential applications in protection of siRNA from degradation, improving the accumulation in the target tissues, enhancing the siRNA therapy and reducing the side effects. In this review, we explore and summarize the role of functional inorganic-organic hybrid systems involved in the siRNA therapeutic advancements. Additionally, we gather the surface engineering strategies of hybrid systems to optimize for siRNA delivery. Major progress in the field of inorganic-organic hybrid platforms including metallic/non-metallic cores modified with organic shells or further fabrication as the vectors for siRNA delivery is discussed to give credit to the interdisciplinary cooperation between chemistry, pharmacy, biology and medicine.
Collapse
Affiliation(s)
- Jianliang Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China and Wenzhou Institute of Biomaterials and Engineering, Chinese Academy of Science, Wenzhou, 325001, China and Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Wei Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Ruogu Qi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA. and Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY10065, USA
| |
Collapse
|
226
|
Ahmed K, Kren BT, Abedin MJ, Vogel RI, Shaughnessy DP, Nacusi L, Korman VL, Li Y, Dehm SM, Zimmerman CL, Niehans GA, Unger GM, Trembley JH. CK2 targeted RNAi therapeutic delivered via malignant cell-directed tenfibgen nanocapsule: dose and molecular mechanisms of response in xenograft prostate tumors. Oncotarget 2018; 7:61789-61805. [PMID: 27557516 PMCID: PMC5308691 DOI: 10.18632/oncotarget.11442] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/15/2016] [Indexed: 01/25/2023] Open
Abstract
CK2, a protein serine/threonine kinase, promotes cell proliferation and suppresses cell death. This essential-for-survival signal demonstrates elevated expression and activity in all cancers examined, and is considered an attractive target for cancer therapy. Here, we present data on the efficacy of a tenfibgen (TBG) coated nanocapsule which delivers its cargo of siRNA (siCK2) or single stranded RNA/DNA oligomers (RNAi-CK2) simultaneously targeting CK2α and α' catalytic subunits. Intravenous administration of TBG-siCK2 or TBG-RNAi-CK2 resulted in significant xenograft tumor reduction at low doses in PC3-LN4 and 22Rv1 models of prostate cancer. Malignant cell uptake and specificity in vivo was verified by FACS analysis and immunofluorescent detection of nanocapsules and PCR detection of released oligomers. Dose response was concordant with CK2αα' RNA transcript levels and the tumors demonstrated changes in CK2 protein and in markers of proliferation and cell death. Therapeutic response corresponded to expression levels for argonaute and GW proteins, which function in oligomer processing and translational repression. No toxicity was detected in non-tumor tissues or by serum chemistry. Tumor specific delivery of anti-CK2 RNAi via the TBG nanoencapsulation technology warrants further consideration of translational potential.
Collapse
Affiliation(s)
- Khalil Ahmed
- Research Service, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Urology, University of Minnesota, Minneapolis, MN, U.S.A.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, U.S.A
| | - Betsy T Kren
- Research Service, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, U.S.A
| | - Md Joynal Abedin
- Research Service, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A
| | - Rachel I Vogel
- Department of Obstretrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN, U.S.A.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, U.S.A
| | - Daniel P Shaughnessy
- Research Service, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A
| | | | | | - Yingming Li
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A
| | - Scott M Dehm
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Urology, University of Minnesota, Minneapolis, MN, U.S.A.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, U.S.A
| | - Cheryl L Zimmerman
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, U.S.A
| | - Gloria A Niehans
- Research Service, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A
| | | | - Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, University of Minnesota, Minneapolis, MN, U.S.A.,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, U.S.A.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, U.S.A
| |
Collapse
|
227
|
Wu JY, Wang ZX, Zhang G, Lu X, Qiang GH, Hu W, Ji AL, Wu JH, Jiang CP. Targeted co-delivery of Beclin 1 siRNA and FTY720 to hepatocellular carcinoma by calcium phosphate nanoparticles for enhanced anticancer efficacy. Int J Nanomedicine 2018; 13:1265-1280. [PMID: 29551896 PMCID: PMC5842779 DOI: 10.2147/ijn.s156328] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose FTY720, known as fingolimod, is a new immunosuppressive agent with effective anticancer properties. Although it was recently confirmed that FTY720 inhibits cancer cell proliferation, FTY720 can also induce protective autophagy and reduce cytotoxicity. Blocking autophagy with Beclin 1 siRNA after treatment with FTY720 promotes apoptosis. The objective of this study was to enhance the anticancer effect of FTY720 in hepatocellular carcinoma (HCC) by targeted co-delivery of FTY720 and Beclin 1 siRNA using calcium phosphate (CaP) nanoparticles (NPs). Materials and methods First, the siRNA was encapsulated within the CaP core. To form an asymmetric lipid bilayer structure, we then used an anionic lipid for the inner leaflet and a cationic lipid for the outer leaflet; after removing chloroform by rotary evaporation, these lipids were dispersed in a saline solution with FTY720. The NPs were analyzed by transmission electron microscopy, dynamic light scattering and ultraviolet–visible spectrophotometry. Cancer cell viability and cell death were analyzed by MTT assays, fluorescence-activated cell sorting analysis and Western blotting. In addition, the in vivo effects of the NPs were investigated using an athymic nude mouse subcutaneous transplantation tumor model. Results When the CaP NPs, called LCP-II NPs, were loaded with FTY720 and siRNA, they exhibited the expected size and were internalized by cells. These NPs were stable in systemic circulation. Furthermore, co-delivery of FTY720 and Beclin 1 siRNA significantly increased cytotoxicity in vitro and in vivo compared with that caused by treatment with the free drug alone. Conclusion The CaP NP system can be further developed for co-delivery of FTY720 and Beclin 1 siRNA to treat HCC, enhancing the anticancer efficacy of FTY720. Our findings provide a new insight into HCC treatment with co-delivered small molecules and siRNA, and these results can be readily translated into cancer clinical trials.
Collapse
Affiliation(s)
- Jun-Yi Wu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhong-Xia Wang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guang Zhang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xian Lu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guang-Hui Qiang
- Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - An-Lai Ji
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jun-Hua Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Chun-Ping Jiang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
228
|
The Special AT-rich Sequence Binding Protein 1 (SATB1) and its role in solid tumors. Cancer Lett 2018; 417:96-111. [DOI: 10.1016/j.canlet.2017.12.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
|
229
|
Polymer-Mediated Inhibition of Pro-invasive Nucleic Acid DAMPs and Microvesicles Limits Pancreatic Cancer Metastasis. Mol Ther 2018; 26:1020-1031. [PMID: 29550075 DOI: 10.1016/j.ymthe.2018.02.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/16/2018] [Accepted: 02/18/2018] [Indexed: 12/16/2022] Open
Abstract
Nucleic acid binding polymers (NABPs) have been extensively used as vehicles for DNA and RNA delivery. More recently, we discovered that a subset of these NABPs can also serve as anti-inflammatory agents by capturing pro-inflammatory extracellular nucleic acids and associated protein complexes that promote activation of toll-like receptors (TLRs) in diseases such as lupus erythematosus. Nucleic-acid-mediated TLR signaling also facilitates tumor progression and metastasis in several cancers, including pancreatic cancer (PC). In addition, extracellular DNA and RNA circulate on or within lipid microvesicles, such as microparticles or exosomes, which also promote metastasis by inducing pro-tumorigenic signaling in cancer cells and pre-conditioning secondary sites for metastatic establishment. Here, we explore the use of an NABP, the 3rd generation polyamidoamine dendrimer (PAMAM-G3), as an anti-metastatic agent. We show that PAMAM-G3 not only inhibits nucleic-acid-mediated activation of TLRs and invasion of PC tumor cells in vitro, but can also directly bind extracellular microvesicles to neutralize their pro-invasive effects as well. Moreover, we demonstrate that PAMAM-G3 dramatically reduces liver metastases in a syngeneic murine model of PC. Our findings identify a promising therapeutic application of NABPs for combating metastatic disease in PC and potentially other malignancies.
Collapse
|
230
|
Liu Y, Ji X, Tong WWL, Askhatova D, Yang T, Cheng H, Wang Y, Shi J. Engineering Multifunctional RNAi Nanomedicine To Concurrently Target Cancer Hallmarks for Combinatorial Therapy. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201710144] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yanlan Liu
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
- Molecular Science and Biomedicine Laboratory; State Key Laboratory of Chemo/Biosensing and Chemometrics; College of Chemistry and Chemical Engineering; Hunan University; Changsha 410082 P. R. China
| | - Xiaoyuan Ji
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Winnie W. L. Tong
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Diana Askhatova
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Tingyuan Yang
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
- State Key Laboratory of Biochemical Engineering; Institute of Process Engineering; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Hongwei Cheng
- Department of Experimental Therapeutics; British Columbia Cancer Agency; Vancouver BC V5Z 1L3 Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics; British Columbia Cancer Agency; Vancouver BC V5Z 1L3 Canada
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| |
Collapse
|
231
|
Liu Y, Ji X, Tong WWL, Askhatova D, Yang T, Cheng H, Wang Y, Shi J. Engineering Multifunctional RNAi Nanomedicine To Concurrently Target Cancer Hallmarks for Combinatorial Therapy. Angew Chem Int Ed Engl 2018; 57:1510-1513. [PMID: 29276823 DOI: 10.1002/anie.201710144] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/06/2017] [Indexed: 01/18/2023]
Abstract
Cancer hallmarks allow the complexity and heterogeneity of tumor biology to be better understood, leading to the discovery of various promising targets for cancer therapy. An amorphous iron oxide nanoparticle (NP)-based RNAi strategy is developed to co-target two cancer hallmarks. The NP technology can modulate the glycolysis pathway by silencing MCT4 to induce tumor cell acidosis, and concurrently exacerbate oxidative stress in tumor cells via the Fenton-like reaction. This strategy has the following features for systemic siRNA delivery: 1) siRNA encapsulation within NPs for improving systemic stability; 2) effective endosomal escape through osmotic pressure and/or endosomal membrane oxidation; 3) small size for enhancing tumor tissue penetration; and 4) triple functions (RNAi, Fenton-like reaction, and MRI) for combinatorial therapy and in vivo tracking.
Collapse
Affiliation(s)
- Yanlan Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Xiaoyuan Ji
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Winnie W L Tong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Diana Askhatova
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Tingyuan Yang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hongwei Cheng
- Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, V5Z 1L3, Canada
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, British Columbia Cancer Agency, Vancouver, BC, V5Z 1L3, Canada
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
232
|
Angelbello AJ, Chen JL, Childs-Disney JL, Zhang P, Wang ZF, Disney MD. Using Genome Sequence to Enable the Design of Medicines and Chemical Probes. Chem Rev 2018; 118:1599-1663. [PMID: 29322778 DOI: 10.1021/acs.chemrev.7b00504] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rapid progress in genome sequencing technology has put us firmly into a postgenomic era. A key challenge in biomedical research is harnessing genome sequence to fulfill the promise of personalized medicine. This Review describes how genome sequencing has enabled the identification of disease-causing biomolecules and how these data have been converted into chemical probes of function, preclinical lead modalities, and ultimately U.S. Food and Drug Administration (FDA)-approved drugs. In particular, we focus on the use of oligonucleotide-based modalities to target disease-causing RNAs; small molecules that target DNA, RNA, or protein; the rational repurposing of known therapeutic modalities; and the advantages of pharmacogenetics. Lastly, we discuss the remaining challenges and opportunities in the direct utilization of genome sequence to enable design of medicines.
Collapse
Affiliation(s)
- Alicia J Angelbello
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jonathan L Chen
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jessica L Childs-Disney
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Peiyuan Zhang
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Zi-Fu Wang
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Matthew D Disney
- Departments of Chemistry and Neuroscience, The Scripps Research Institute , 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
233
|
Chen C, Yang Z, Tang X. Chemical modifications of nucleic acid drugs and their delivery systems for gene-based therapy. Med Res Rev 2018; 38:829-869. [PMID: 29315675 DOI: 10.1002/med.21479] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 12/12/2022]
Abstract
Gene-based therapy is one of essential therapeutic strategies for precision medicine through targeting specific genes in specific cells of target tissues. However, there still exist many problems that need to be solved, such as safety, stability, selectivity, delivery, as well as immunity. Currently, the key challenges of gene-based therapy for clinical potential applications are the safe and effective nucleic acid drugs as well as their safe and efficient gene delivery systems. In this review, we first focus on current nucleic acid drugs and their formulation in clinical trials and on the market, including antisense oligonucleotide, siRNA, aptamer, and plasmid nucleic acid drugs. Subsequently, we summarize different chemical modifications of nucleic acid drugs as well as their delivery systems for gene-based therapeutics in vivo based on nucleic acid chemistry and nanotechnology methods.
Collapse
Affiliation(s)
- Changmai Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xinjing Tang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
234
|
Gibori H, Eliyahu S, Krivitsky A, Ben-Shushan D, Epshtein Y, Tiram G, Blau R, Ofek P, Lee JS, Ruppin E, Landsman L, Barshack I, Golan T, Merquiol E, Blum G, Satchi-Fainaro R. Amphiphilic nanocarrier-induced modulation of PLK1 and miR-34a leads to improved therapeutic response in pancreatic cancer. Nat Commun 2018; 9:16. [PMID: 29295989 PMCID: PMC5750234 DOI: 10.1038/s41467-017-02283-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022] Open
Abstract
The heterogeneity of pancreatic ductal adenocarcinoma (PDAC) suggests that successful treatment might rely on simultaneous targeting of multiple genes, which can be achieved by RNA interference-based therapeutic strategies. Here we show a potent combination of microRNA and siRNA delivered by an efficient nanocarrier to PDAC tumors. Using proteomic-microRNA profiles and survival data of PDAC patients from TCGA, we found a novel signature for prolonged survival. Accordingly, we used a microRNA-mimic to increase miR-34a together with siRNA to silence PLK1 oncogene. For in vivo dual-targeting of this combination, we developed a biodegradable amphiphilic polyglutamate amine polymeric nanocarrier (APA). APA-miRNA-siRNA polyplexes systemically administered to orthotopically inoculated PDAC-bearing mice showed no toxicity and accumulated at the tumor, resulting in an enhanced antitumor effect due to inhibition of MYC oncogene, a common target of both miR-34a and PLK1. Taken together, our findings warrant this unique combined polyplex's potential as a novel nanotherapeutic for PDAC.
Collapse
Affiliation(s)
- Hadas Gibori
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Shay Eliyahu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yana Epshtein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Rachel Blau
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Joo Sang Lee
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, 20742, USA
| | - Eytan Ruppin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, 20742, USA
- Blavatnik School of Computer Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, 52621, Israel
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Talia Golan
- Department of Pathology, Sheba Medical Center, Tel Hashomer, 52621, Israel
| | - Emmanuelle Merquiol
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Ein Kerem Campus, The Hebrew University, Jerusalem, Israel
| | - Galia Blum
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Ein Kerem Campus, The Hebrew University, Jerusalem, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
235
|
Dreaden EC, Kong YW, Quadir MA, Correa S, Suárez‐López L, Barberio AE, Hwang MK, Shi AC, Oberlton B, Gallagher PN, Shopsowitz KE, Elias KM, Yaffe MB, Hammond PT. RNA-Peptide nanoplexes drug DNA damage pathways in high-grade serous ovarian tumors. Bioeng Transl Med 2018; 3:26-36. [PMID: 29376131 PMCID: PMC5773954 DOI: 10.1002/btm2.10086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/27/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022] Open
Abstract
DNA damaging chemotherapy is a cornerstone of current front-line treatments for advanced ovarian cancer (OC). Despite the fact that a majority of these patients initially respond to therapy, most will relapse with chemo-resistant disease; therefore, adjuvant treatments that synergize with DNA-damaging chemotherapy could improve treatment outcomes and survival in patients with this deadly disease. Here, we report the development of a nanoscale peptide-nucleic acid complex that facilitates tumor-specific RNA interference therapy to chemosensitize advanced ovarian tumors to frontline platinum/taxane therapy. We found that the nanoplex-mediated silencing of the protein kinase, MK2, profoundly sensitized mouse models of high-grade serous OC to cytotoxic chemotherapy by blocking p38/MK2-dependent cell cycle checkpoint maintenance. Combined RNAi therapy improved overall survival by 37% compared with platinum/taxane chemotherapy alone and decreased metastatic spread to the lungs without observable toxic side effects. These findings suggest (a) that peptide nanoplexes can serve as safe and effective delivery vectors for siRNA and (b) that combined inhibition of MK2 could improve treatment outcomes in patients currently receiving frontline chemotherapy for advanced OC.
Collapse
Affiliation(s)
- Erik C. Dreaden
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
- Present address:
Wallace H. Coulter Dept. of Biomedical EngineeringGeorgia Institute of Technology and Emory University, Atlanta, GA 30322; Dept. of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of MedicineAtlantaGA 30322
| | - Yi Wen Kong
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of BiologyMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Mohiuddin A. Quadir
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
- Present address:
Dept. of Coatings and Polymeric MaterialsNorth Dakota State UniversityFargoND 58108
| | - Santiago Correa
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Lucia Suárez‐López
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of BiologyMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Antonio E. Barberio
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Mun Kyung Hwang
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of BiologyMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Aria C. Shi
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Benjamin Oberlton
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Paige N. Gallagher
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
| | - Kevin E. Shopsowitz
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
- Present address:
Faculty of MedicineUniversity of British ColumbiaBC V1Y 1T3Canada
| | - Kevin M. Elias
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's HospitalHarvard Medical SchoolBostonMA 02215
| | - Michael B. Yaffe
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of BiologyMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
- Div. of Acute Care Surgery, Trauma, and Surgical Critical Care, Dept. of Surgery, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA 02215
| | - Paula T. Hammond
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA 02139
- Dept. of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA 02139
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA 02139
| |
Collapse
|
236
|
Du XJ, Wang ZY, Wang YC. Redox-sensitive dendrimersomes assembled from amphiphilic Janus dendrimers for siRNA delivery. Biomater Sci 2018; 6:2122-2129. [DOI: 10.1039/c8bm00491a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A cationic redox-sensitive Janus dendrimer (ssJD) that self-assembles into redox-sensitive dendrimersomes (RSDs) to complex with siRNA can readily deliver siRNA into tumor cells, and then rapidly release siRNA in a reductive environment to down-regulate a targeted gene.
Collapse
Affiliation(s)
- Xiao-Jiao Du
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- P.R. China
| | - Ze-Yu Wang
- International Department
- The Affiliated High School of South China Normal University
- Guangzhou
- P.R. China
| | - Yu-Cai Wang
- School of Life Sciences
- University of Science and Technology of China
- Hefei
- P.R. China
| |
Collapse
|
237
|
Clusterin inhibition mediates sensitivity to chemotherapy and radiotherapy in human cancer. Anticancer Drugs 2017; 28:702-716. [PMID: 28471806 DOI: 10.1097/cad.0000000000000507] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Since its discovery in 1983, the protein clusterin (CLU) has been isolated from almost all human tissues and fluids and linked to the development of different physiopathological processes, including carcinogenesis and tumor progression. During the last few years, several studies have shown the cytoprotective role of secretory CLU in tumor cells, inhibiting their apoptosis and enhancing their resistance to conventional treatments including hormone depletion, chemotherapy, and radiotherapy. In an effort to determine the therapeutic potential that the inhibition of this protein could have on the development of new strategies for cancer treatment, numerous studies have been carried out in this field, with results, in most cases, satisfactory but sometimes contradictory. In this document, we summarize for the first time the current knowledge of the effects that CLU inhibition has on sensitizing tumor cells to conventional cancer treatments and discuss its importance in the development of new strategies against cancer.
Collapse
|
238
|
Kedves AT, Gleim S, Liang X, Bonal DM, Sigoillot F, Harbinski F, Sanghavi S, Benander C, George E, Gokhale PC, Nguyen QD, Kirschmeier PT, Distel RJ, Jenkins J, Goldberg MS, Forrester WC. Recurrent ubiquitin B silencing in gynecological cancers establishes dependence on ubiquitin C. J Clin Invest 2017; 127:4554-4568. [PMID: 29130934 DOI: 10.1172/jci92914] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 10/09/2017] [Indexed: 02/06/2023] Open
Abstract
Transcriptional repression of ubiquitin B (UBB) is a cancer-subtype-specific alteration that occurs in a substantial population of patients with cancers of the female reproductive tract. UBB is 1 of 2 genes encoding for ubiquitin as a polyprotein consisting of multiple copies of ubiquitin monomers. Silencing of UBB reduces cellular UBB levels and results in an exquisite dependence on ubiquitin C (UBC), the second polyubiquitin gene. UBB is repressed in approximately 30% of high-grade serous ovarian cancer (HGSOC) patients and is a recurrent lesion in uterine carcinosarcoma and endometrial carcinoma. We identified ovarian tumor cell lines that retain UBB in a repressed state, used these cell lines to establish orthotopic ovarian tumors, and found that inducible expression of a UBC-targeting shRNA led to tumor regression, and substantial long-term survival benefit. Thus, we describe a recurrent cancer-specific lesion at the level of ubiquitin production. Moreover, these observations reveal the prognostic value of UBB repression and establish UBC as a promising therapeutic target for ovarian cancer patients with recurrent UBB silencing.
Collapse
Affiliation(s)
- Alexia T Kedves
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Scott Gleim
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Xiaoyou Liang
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Dennis M Bonal
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Frederic Sigoillot
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Fred Harbinski
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Sneha Sanghavi
- Neurosciences, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Christina Benander
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Elizabeth George
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | | | | | | | | | - Jeremy Jenkins
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | | | - William C Forrester
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| |
Collapse
|
239
|
Targeted Delivery of siRNA Therapeutics to Malignant Tumors. JOURNAL OF DRUG DELIVERY 2017; 2017:6971297. [PMID: 29218233 PMCID: PMC5700508 DOI: 10.1155/2017/6971297] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/10/2017] [Indexed: 01/11/2023]
Abstract
Over the past 20 years, a diverse group of ligands targeting surface biomarkers or receptors has been identified with several investigated to target siRNA to tumors. Many approaches to developing tumor-homing peptides, RNA and DNA aptamers, and single-chain variable fragment antibodies by using phage display, in vitro evolution, and recombinant antibody methods could not have been imagined by researchers in the 1980s. Despite these many scientific advances, there is no reason to expect that the ligand field will not continue to evolve. From development of ligands based on novel or existing biomarkers to linking ligands to drugs and gene and antisense delivery systems, several fields have coalesced to facilitate ligand-directed siRNA therapeutics. In this review, we discuss the major categories of ligand-targeted siRNA therapeutics for tumors, as well as the different strategies to identify new ligands.
Collapse
|
240
|
Andreozzi P, Diamanti E, Py-Daniel KR, Cáceres-Vélez PR, Martinelli C, Politakos N, Escobar A, Muzi-Falconi M, Azevedo R, Moya SE. Exploring the pH Sensitivity of Poly(allylamine) Phosphate Supramolecular Nanocarriers for Intracellular siRNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:38242-38254. [PMID: 29039643 DOI: 10.1021/acsami.7b11132] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Silencing RNA (siRNA) technologies emerge as a promising therapeutic tool for the treatment of multiple diseases. An ideal nanocarrier (NC) for siRNAs should be stable at physiological pH and release siRNAs in acidic endosomal pH, fulfilling siRNA delivery only inside cells. Here, we show a novel application of polyamine phosphate NCs (PANs) based on their capacity to load negatively charged nucleic acids and their pH stability. PANs are fabricated by complexation of phosphate anions from phosphate buffer solution (PB) with the amine groups of poly(allylamine) hydrochloride as carriers for siRNAs. PANs are stable in a narrow pH interval, from 7 to 9, and disassemble at pH's higher than 9 and lower than 6. siRNAs are encapsulated by complexation with poly(allylamine) hydrochloride before or after PAN formation. PANs with encapsulated siRNAs are stable in cell media. Once internalized in cells following endocytic pathways, PANs disassemble at the low endosomal pH and release the siRNAs into the cytoplasm. Confocal laser scanning microscopy (CLSM) images of Rhodamine Green labeled PANs (RG-PANs) with encapsulated Cy3-labeled siRNA in A549 cells show that siRNAs are released from the PANs. Colocalization experiments with labeled endosomes and either labeled siRNAs prove the translocation of siRNAs into the cytosol. As a proof of concept, it is shown that PANs with encapsulated green fluorescence protein (GFP) siRNAs silence GFP in A549 cells expressing this protein. Silencing efficacy was evaluated by flow cytometry, CLSM, and Western blot assays. These results open the way for the use of poly(allylamine) phosphate nanocarriers for the intracellular delivery of genetic materials.
Collapse
Affiliation(s)
- Patrizia Andreozzi
- Soft Matter Nanotechnology Group, CIC biomaGUNE , Paseo Miramón 182 C, San Sebastián, Guipúzcoa 20014, Spain
| | - Eleftheria Diamanti
- Soft Matter Nanotechnology Group, CIC biomaGUNE , Paseo Miramón 182 C, San Sebastián, Guipúzcoa 20014, Spain
| | - Karen Rapp Py-Daniel
- Departamento de Genética e Morfologia, Universidade de Brasília, Instituto de Ciências Biológicas , Brasília, Distrito Federal 70910-900, Brazil
| | - Paolin Rocio Cáceres-Vélez
- Departamento de Genética e Morfologia, Universidade de Brasília, Instituto de Ciências Biológicas , Brasília, Distrito Federal 70910-900, Brazil
| | - Chiara Martinelli
- Department of Biosciences, University of Milan , Via Giovanni Celoria, 26, Milan 20133, Italy
| | - Nikolaos Politakos
- Soft Matter Nanotechnology Group, CIC biomaGUNE , Paseo Miramón 182 C, San Sebastián, Guipúzcoa 20014, Spain
| | - Ane Escobar
- Soft Matter Nanotechnology Group, CIC biomaGUNE , Paseo Miramón 182 C, San Sebastián, Guipúzcoa 20014, Spain
| | - Marco Muzi-Falconi
- Department of Biosciences, University of Milan , Via Giovanni Celoria, 26, Milan 20133, Italy
| | - Ricardo Azevedo
- Departamento de Genética e Morfologia, Universidade de Brasília, Instituto de Ciências Biológicas , Brasília, Distrito Federal 70910-900, Brazil
| | - Sergio E Moya
- Soft Matter Nanotechnology Group, CIC biomaGUNE , Paseo Miramón 182 C, San Sebastián, Guipúzcoa 20014, Spain
| |
Collapse
|
241
|
Abstract
The principles of engineering and physics have been applied to oncology for nearly 50 years. Engineers and physical scientists have made contributions to all aspects of cancer biology, from quantitative understanding of tumour growth and progression to improved detection and treatment of cancer. Many early efforts focused on experimental and computational modelling of drug distribution, cell cycle kinetics and tumour growth dynamics. In the past decade, we have witnessed exponential growth at the interface of engineering, physics and oncology that has been fuelled by advances in fields including materials science, microfabrication, nanomedicine, microfluidics, imaging, and catalysed by new programmes at the National Institutes of Health (NIH), including the National Institute of Biomedical Imaging and Bioengineering (NIBIB), Physical Sciences in Oncology, and the National Cancer Institute (NCI) Alliance for Nanotechnology. Here, we review the advances made at the interface of engineering and physical sciences and oncology in four important areas: the physical microenvironment of the tumour and technological advances in drug delivery; cellular and molecular imaging; and microfluidics and microfabrication. We discussthe research advances, opportunities and challenges for integrating engineering and physical sciences with oncology to develop new methods to study, detect and treat cancer, and we also describe the future outlook for these emerging areas.
Collapse
Affiliation(s)
- Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 S. 33rd Street, Philadelphia, Pennsylvania 19104, USA
- Department of Chemical Engineering, David H. Koch Institute for Integrated Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories of Tumour Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Cox 7, Boston, Massachusetts 02114, USA
| | - Robert Langer
- Department of Chemical Engineering, David H. Koch Institute for Integrated Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
242
|
Zou Y, Zheng M, Yang W, Meng F, Miyata K, Kim HJ, Kataoka K, Zhong Z. Virus-Mimicking Chimaeric Polymersomes Boost Targeted Cancer siRNA Therapy In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28961339 DOI: 10.1002/adma.201703285] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/02/2017] [Indexed: 05/16/2023]
Abstract
Small interfering RNA (siRNA) offers a highly selective and effective pharmaceutical for various life-threatening diseases, including cancers. The clinical translation of siRNA is, however, challenged by its short plasma life, poor cell uptake, and cumbersome intracellular trafficking. Here, cNGQGEQc peptide-functionalized reversibly crosslinked chimaeric polymersomes (cNGQ/RCCPs) is shown to mediate high-efficiency targeted delivery of Polo-like kinase1 specific siRNA (siPLK1) to orthotopic human lung cancer in nude mice. Strikingly, siRNA is completely and tightly loaded into the aqueous lumen of the polymersomes at an unprecedentedly low N/P ratio of 0.45. cNGQ/RCCPs loaded with firefly luciferase specific siRNA (siGL3) or siPLK1 are efficiently taken up by α3 β1 -integrin-overexpressing A549 lung cancer cells and quickly release the payloads to the cytoplasm, inducing highly potent and sequence-specific gene silencing in vitro. The in vivo studies using nude mice bearing orthotopic A549 human lung tumors reveal that siPLK1-loaded cNGQ/RCCPs boost long circulation, superb tumor accumulation and selectivity, effective suppression of tumor growth, and significantly improved survival time. These virus-mimicking chimaeric polymersomes provide a robust and potent platform for targeted cancer siRNA therapy.
Collapse
Affiliation(s)
- Yan Zou
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Meng Zheng
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Weijing Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8656, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, and Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hyun Jin Kim
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, and Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kazunori Kataoka
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8656, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, and Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
- Policy Alternative Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Innovation Center of NanoMedicine, Institute of Industry Promotion-Kawasaki, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
243
|
Mutisya D, Hardcastle T, Cheruiyot SK, Pallan PS, Kennedy SD, Egli M, Kelley ML, Smith AVB, Rozners E. Amide linkages mimic phosphates in RNA interactions with proteins and are well tolerated in the guide strand of short interfering RNAs. Nucleic Acids Res 2017; 45:8142-8155. [PMID: 28854734 PMCID: PMC5737567 DOI: 10.1093/nar/gkx558] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/18/2017] [Indexed: 12/21/2022] Open
Abstract
While the use of RNA interference (RNAi) in molecular biology and functional genomics is a well-established technology, in vivo applications of synthetic short interfering RNAs (siRNAs) require chemical modifications. We recently found that amides as non-ionic replacements for phosphodiesters may be useful modifications for optimization of siRNAs. Herein, we report a comprehensive study of systematic replacement of a single phosphate with an amide linkage throughout the guide strand of siRNAs. The results show that amides are surprisingly well tolerated in the seed and central regions of the guide strand and increase the silencing activity when placed between nucleosides 10 and 12, at the catalytic site of Argonaute. A potential explanation is provided by the first crystal structure of an amide-modified RNA-DNA with Bacillus halodurans RNase H1. The structure reveals how small changes in both RNA and protein conformation allow the amide to establish hydrogen bonding interactions with the protein. Molecular dynamics simulations suggest that these alternative binding modes may compensate for interactions lost due to the absence of a phosphodiester moiety. Our results suggest that an amide can mimic important hydrogen bonding interactions with proteins required for RNAi activity and may be a promising modification for optimization of biological properties of siRNAs.
Collapse
Affiliation(s)
- Daniel Mutisya
- Department of Chemistry, Binghamton University, The State University of New York, Binghamton, NY 13902, USA
| | | | - Samwel K Cheruiyot
- Department of Chemistry, Binghamton University, The State University of New York, Binghamton, NY 13902, USA
| | - Pradeep S Pallan
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Scott D Kennedy
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Martin Egli
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | - Eriks Rozners
- Department of Chemistry, Binghamton University, The State University of New York, Binghamton, NY 13902, USA
| |
Collapse
|
244
|
Ki67 targeted strategies for cancer therapy. Clin Transl Oncol 2017; 20:570-575. [DOI: 10.1007/s12094-017-1774-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
|
245
|
Sharma B, Crist RM, Adiseshaiah PP. Nanotechnology as a Delivery Tool for Precision Cancer Therapies. AAPS JOURNAL 2017; 19:1632-1642. [DOI: 10.1208/s12248-017-0152-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/19/2017] [Indexed: 01/20/2023]
|
246
|
Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv Drug Deliv Rev 2017; 120:89-107. [PMID: 28736304 DOI: 10.1016/j.addr.2017.07.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 12/18/2022]
Abstract
Cancer progression to metastatic stages is still unmanageable and the promise of effective anti-metastatic therapy remains largely unmet, emphasizing the need to develop novel therapeutics. The special focus here is on cancer stem cells (CSC) as the seed of tumor initiation, epithelial-mesenchymal transition, chemoresistance and, as a consequence, drivers of metastatic dissemination. We report on targeted therapies gearing towards the CSC's internal and membrane-anchored markers using agents such as antibody derivatives, nucleic therapeutics, small molecules and genetic payloads. Another emphasis lies on novel proceedings envisaged to deliver current and prospective therapies to the target sites using newest viral and non-viral vector technologies. In this review, we summarize recent progress and remaining challenges in therapeutic strategies to combat CSC.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany.
| | - Manish Solanki
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| | - Ottmar Herchenröder
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| |
Collapse
|
247
|
Liu T, Wu X, Chen T, Luo Z, Hu X. Downregulation of DNMT3A by miR-708-5p Inhibits Lung Cancer Stem Cell-like Phenotypes through Repressing Wnt/β-catenin Signaling. Clin Cancer Res 2017; 24:1748-1760. [PMID: 28972040 DOI: 10.1158/1078-0432.ccr-17-1169] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/14/2017] [Accepted: 09/21/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Lung cancer is the leading cause of cancer-related death in the world, and emerging evidences suggest that lung cancer stem cells (CSC) are associated with its poor prognosis, tumor recurrence, and therapy resistance. Here we reveal a novel role for miR-708-5p in inhibiting lung CSC-like features.Experimental Design: Phenotypic effects of miR-708-5p on the lung CSC-like properties were examined by in vitro sphere formation assay and in xenografted animal models. Immunoblotting, dual luciferase reporter, and immunocytochemistry were performed to determine the target of miR-708-5p. DNA methylation of CDH1 promoter region was tested using bisulfate sequencing. Genome-wide miRNA sequencing data of 990 patients from The Cancer Genome Atlas (TCGA) dataset and 148 patients from China cohort were analyzed to excavate the pathogenic implications of miR-708-5p.Results: Expression of miR-708-5p inhibits the CSC traits of NSCLC cells in vitro while antagonizing miR-708-5p promotes tumorigenesis in vivo miR-708-5p directly suppresses the translation of DNMT3A, which results in a substantial reduction of global DNA methylation and the upregulated expression of tumor suppressor CDH1. The upregulation of CDH1 decreased the activity of Wnt/β-catenin signaling and then impaired the stemness characteristics of NSCLC cells. Clinically, patients with high miR-708-5p expression show significantly better survival and lower recurrence. Furthermore, miR-708-5p has a promising potential to apply to differentiating histologic subtypes in NSCLC.Conclusions: Our findings support that miR-708-5p suppresses NSCLC initiation, development, and stemness through interfering DNMT3A-dependent DNA methylation. miR-708-5p may function as a novel diagnostic and prognostic biomarker in NSCLC. Clin Cancer Res; 24(7); 1748-60. ©2017 AACR.
Collapse
Affiliation(s)
- Tianchi Liu
- Population and Quantitative Genetics Laboratory, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoping Wu
- Population and Quantitative Genetics Laboratory, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Tong Chen
- Population and Quantitative Genetics Laboratory, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zewei Luo
- Population and Quantitative Genetics Laboratory, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China. .,School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Xiaohua Hu
- Population and Quantitative Genetics Laboratory, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
248
|
Pentecost AE, Witherel CE, Gogotsi Y, Spiller KL. Anti-inflammatory effects of octadecylamine-functionalized nanodiamond on primary human macrophages. Biomater Sci 2017; 5:2131-2143. [PMID: 28875995 PMCID: PMC5719499 DOI: 10.1039/c7bm00294g] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic inflammatory disorders such as rheumatoid arthritis are characterized by excessive pro-inflammatory or "M1" activation of macrophages, the primary cells of the innate immune system. Current treatments include delivery of glucocorticoids (e.g. dexamethasone - Dex), which reduce pro-inflammatory M1 behaviour in macrophages. However, these treatments have many off-target effects on cells other than macrophages, resulting in broad immunosuppression. To limit such side effects, drug-incorporated nano- and microparticles may be used to selectively target macrophages via phagocytosis, because of their roles as highly effective phagocytes in the body. In this study, surface-modified nanodiamond (ND) was explored as a platform for the delivery of dexamethasone to macrophages because of ND's rich surface chemistry, which contributes to ND's high potential as a versatile drug delivery platform. After finding that octadecylamine-functionalized nanodiamond (ND-ODA) enhanced adsorption of Dex compared to carboxylated ND, the effects of Dex, ND-ODA, and Dex-adsorbed ND-ODA on primary human macrophage gene expression were characterized. Surprisingly, even in the absence of Dex, ND-ODA had strong anti-inflammatory effects, as determined by multiplex gene expression via NanoString and by protein secretion analysis via ELISA. ND-ODA also inhibited expression of M2a markers yet increased the expression of M2c markers and phagocytic receptors. Interestingly, the adsorption of Dex to ND-ODA further increased some anti-inflammatory effects, but abrogated the effect on phagocytic receptors, compared to its individual components. Overall, the ability of ND-ODA to promote anti-inflammatory and pro-phagocytic behaviour in macrophages, even in the absence of loaded drugs, suggests its potential for use as an anti-inflammatory therapeutic to directly target macrophages through phagocytosis.
Collapse
Affiliation(s)
- A E Pentecost
- Department of Materials Science and Engineering, College of Engineering, Drexel University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
249
|
Targeted Delivery of siRNA with pH-Responsive Hybrid Gold Nanostars for Cancer Treatment. Int J Mol Sci 2017; 18:ijms18102029. [PMID: 28937584 PMCID: PMC5666711 DOI: 10.3390/ijms18102029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022] Open
Abstract
In this work, we report the engineering of gold nanostars (GNS) to deliver small interfering RNA (siRNA) into HepG2 cells. The ligand DG-PEG-Lipoic acid (LA)-Lys-9R (hydrazone) was designed to functionalize GNS, and create the nanoparticles named as 9R/DG-GNS (hydrazone). In the ligand, 2-deoxyglucose (DG) is the targeting molecule, polyethylene glycol (PEG) helps to improve the dispersity and biocompatibility, 9-poly-d-arginine (9R) is employed to provide a positive surface charge and adsorb negative siRNA, and hydrazone bonds are pH-responsive and can avoid receptor-mediated endosomal recycling. Compared to GNS alone, 9R/DG-GNS (hydrazone) showed superior transfection efficiency. The expressions of cyclooxygenase-2 (COX-2) in HepG2 and SGC7901 cells were significantly suppressed by siRNA/9R/DG-GNS (hydrazone) complex. Notably, 9R/DG-GNS (hydrazone) possessed low cytotoxicity even at high concentrations in both normal cells and tumor cells. The combination treatment of siRNA/9R/DG-GNS (hydrazone) complex inhibited the cell growth rate by more than 75%. These results verified that the pH-responsive GNS complex is a promising siRNA delivery system for cancer therapy, and it is anticipated that near-infrared absorbing GNS with good photothermal conversion efficiency can be potentially used for photothermal therapy of tumors.
Collapse
|
250
|
Fu Q, Liu P, Sun X, Huang S, Han F, Zhang L, Xu Y, Liu T. Ribonucleic acid interference knockdown of IL-6 enhances the efficacy of cisplatin in laryngeal cancer stem cells by down-regulating the IL-6/STAT3/HIF1 pathway. Cancer Cell Int 2017; 17:79. [PMID: 28878571 PMCID: PMC5584337 DOI: 10.1186/s12935-017-0448-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/29/2017] [Indexed: 12/26/2022] Open
Abstract
Background Cisplatin has been used in the treatment of many cancers, including laryngeal cancer; however, its efficacy can be reduced due to the development of drug resistance. This study aimed to investigate whether interleukin-6 (IL-6) knockdown may enhance the efficacy of cisplatin in laryngeal cancer stem cells (CSC) and the potential involvement of the signal transducer and activator of transcription 3 (STAT3) and hypoxia-inducible factor 1 (HIF1) in this effect. Methods The ALDH+ and CD44+ CSC in Hep2 human laryngeal squamous cancer cells were identified by the fluorescence-activated cell sorting technique. IL-6, STAT3 and HIF1 mRNA and protein expressions were examined with quantitative real-time polymerase chain reaction and Western blot, respectively. Cell proliferation was measured by MTT assay. Tumorigenicity was measured by a colony formation assay and invasion was determined by a cell invasion assay. Apoptotic cells were counted by flow cytometry. Immunohistochemistry was performed to detect immunoreactive IL-6, STAT3 and HIF1 cells in xenografts. Results The mRNA and protein levels of IL-6, STAT3 and HIF1 were significantly increased in Hep2-CSC as compared with those from Hep2 cells. Application of siRNA-IL-6 to knockdown IL-6 resulted in significantly decreased IL-6, STAT3 and HIF1 mRNA and protein levels. IL-6 knockdown reduced cell proliferation, tumorigenicity and invasion and increased apoptosis within CSC. Enhanced degrees of suppression in these parameters were observed when IL-6 knockdown was combined with cisplatin in these CSC. Results from the xenograft study showed that the combination of IL-6 knockdown and cisplatin further inhibited the growth of xenografts as compared with that obtained in the cisplatin-injected group alone. Immunoreactive IL-6, STAT3 and HIF1 cell numbers were markedly reduced in IL-6 knockdown tumor tissues. IL-6, STAT3 and HIF1 immunoreactive cell counts were further reduced in tissue where IL-6 knockdown was combined with cisplatin treatment as compared with tissue receiving cisplatin alone. Conclusions IL-6 knockdown can increase chemo-drug efficacy of cisplatin, inhibit tumor growth and reduce the potential for tumor recurrence and metastasis in laryngeal cancer. The IL-6/STAT3/HIF1 pathway may represent an important target for investigating therapeutic strategies for the treatment of laryngeal cancer.
Collapse
Affiliation(s)
- Qiang Fu
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003 China
| | - Pengruofeng Liu
- Department of Stomatology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Xiumei Sun
- Department of Otolaryngology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264003 China
| | - Shanshan Huang
- Department of Otolaryngology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264003 China
| | - Fengchan Han
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003 China
| | - Lili Zhang
- Department of Otolaryngology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264003 China
| | - Yannan Xu
- Department of Otolaryngology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264003 China
| | - Tingyan Liu
- Department of Otolaryngology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264003 China
| |
Collapse
|