1
|
Kazemi Shariat Panahi H, Dehhaghi M, Guillemin GJ, Peng W, Aghbashlo M, Tabatabaei M. Targeting microRNAs as a promising anti-cancer therapeutic strategy against traffic-related air pollution-mediated lung cancer. Cancer Metastasis Rev 2024; 43:657-672. [PMID: 37910296 DOI: 10.1007/s10555-023-10142-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023]
Abstract
Air pollutants are increasingly emitted into the atmosphere because of the high dependency of humans on fossil-derived fuels. Wind speed and direction assisted high dispersibility and uncontrolled nature of air pollution across geo-/demographical borders, making it one of the major global concerns. Besides climate change, air pollution has been found to be associated with various diseases, such as cancer. Lung cancer, which is the world's most common type of cancer, has been found to be associated with traffic-related air pollution. Research and political efforts have been taken to explore green/renewable energy sources. However, these efforts at the current intensity cannot cope with the increasing need for fossil fuels. More specifically, political tensions such as the Russian-Ukraine war, economic tension (e.g., China-USA economic tensions), and other issues (e.g., pandemic, higher inflation rate, and poverty) significantly hindered phasing out fossil fuels. In this context, an increasing global population will be exposed to traffic-related air pollution, which justifies the current uptrend in the number of lung cancer patients. To combat this health burden, novel treatments with higher efficiency and specificity must be designed. One of the potential "life changer" options is microRNA (miRNA)-based therapy to target the expression of oncogenic genes. That said, this review discusses the association of traffic-related air pollution with lung cancer, the changes in indigenous miRNAs in the body during lung cancer, and the current status of miRNA therapeutics for lung cancer treatment. We believe that the article will significantly appeal to a broad readership of oncologists, environmentalists, and those who work in the field of (bio)energy. It may also gain the policymakers' attention to establish better health policies and regulations about air pollution, for example, by promoting (bio)fuel exploration, production, and consumption.
Collapse
Affiliation(s)
- Hamed Kazemi Shariat Panahi
- Henan Province Engineering Research Center for Biomass Value-Added Products, School of Forestry, Henan Agricultural University, Zhengzhou, 450002, China
- Neuroinflammation Group, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Biofuel Research Team (BRTeam), Kuala Terengganu, Terengganu, Malaysia
| | - Mona Dehhaghi
- Neuroinflammation Group, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Biofuel Research Team (BRTeam), Kuala Terengganu, Terengganu, Malaysia
| | | | - Wanxi Peng
- Henan Province Engineering Research Center for Biomass Value-Added Products, School of Forestry, Henan Agricultural University, Zhengzhou, 450002, China.
| | - Mortaza Aghbashlo
- Department of Mechanical Engineering of Agricultural Machinery, Faculty of Agricultural Engineering and Technology, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | - Meisam Tabatabaei
- Henan Province Engineering Research Center for Biomass Value-Added Products, School of Forestry, Henan Agricultural University, Zhengzhou, 450002, China.
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries (AKUATROP), Universiti Malaysia Terengganu, 21030, Kuala Nerus, Terengganu, Malaysia.
- Department of Biomaterials, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, 600 077, India.
| |
Collapse
|
2
|
Zhou Y, Jia H, Hu A, Liu R, Zeng X, Wang H. Nanoparticles Targeting Delivery Antagomir-483-5p to Bone Marrow Mesenchymal Stem Cells Treat Osteoporosis by Increasing Bone Formation. Curr Stem Cell Res Ther 2023; 18:115-126. [PMID: 35473519 DOI: 10.2174/1574888x17666220426120850] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/12/2022] [Accepted: 03/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Promoting bone marrow mesenchymal stem cell (BMSC) osteoblastic differentiation is a promising therapeutic strategy for osteoporosis (OP). The present study demonstrates that miR- 483-5p inhibits the osteogenic differentiation of BMSCs. Therefore, selectively delivering the nanoparticles carrying antagomir-483-5p (miR-483-5p inhibitor) to BMSCs is expected to become an effective treatment drug for OP. METHODS Real-time PCR assays were used to analyze miR-483-5p, ALP and Bglap levels in BMSCs of ovariectomized and aged osteoporotic mice. Immunoglobulin G and poloxamer-188 encapsulated the functional small molecules, and a BMSC-targeting aptamer was employed to confirm the direction of the nanoparticles to selectively and efficiently deliver antagomir-483-5p to BMSCs in vivo. Luciferase assays were used to determine the target genes of miR-483-5p. Western blot assays and immunohistochemistry staining were used to detect the targets in vitro and in vivo. RESULTS miR-483-5p levels were increased in BMSCs of ovariectomized and aged osteoporotic mice. Inhibiting miR-483-5p levels in BMSCs by antagomir-483-5p in vitro promoted the expression of bone formation markers, such as ALP and Bglap. The FAM-BMSC-aptamer-nanoparticles carrying antagomir- 483-5p were taken up by BMSCs, resulting in stimulation of BMSC osteoblastic differentiation in vitro and osteoporosis prevention in vivo. Furthermore, our research demonstrated that mitogen-activated protein kinase 1 (MAPK1) and SMAD family member 5 (Smad5) were direct targets of miR-483-5p in regulating BMSC osteoblastic differentiation and osteoporosis pathological processes. CONCLUSIONS The important therapeutic role of FAM-BMSC-aptamer-nanoparticles carrying antagomir- 483-5p in osteoporosis was established in our study. These nanoparticles are a novel candidate for the clinical prevention and treatment of osteoporosis. The optimized, targeted drug delivery platform for small molecules will provide new ideas for treating clinical diseases.
Collapse
Affiliation(s)
- Yue Zhou
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Hainan Medical University, Haikou, 571199, China
| | - Hao Jia
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Hainan Medical University, Haikou, 571199, China
| | - Aihua Hu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Hainan Medical University, Haikou, 571199, China
| | - Rangru Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Hainan Medical University, Haikou, 571199, China.,Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Xiangzhou Zeng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Hainan Medical University, Haikou, 571199, China
| | - Hua Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Hainan Medical University, Haikou, 571199, China
| |
Collapse
|
3
|
Abstract
This Review examines the state-of-the-art in the delivery of nucleic acid therapies that are directed to the vascular endothelium. First, we review the most important homeostatic functions and properties of the vascular endothelium and summarize the nucleic acid tools that are currently available for gene therapy and nucleic acid delivery. Second, we consider the opportunities available with the endothelium as a therapeutic target and the experimental models that exist to evaluate the potential of those opportunities. Finally, we review the progress to date from investigations that are directly targeting the vascular endothelium: for vascular disease, for peri-transplant therapy, for angiogenic therapies, for pulmonary endothelial disease, and for the blood-brain barrier, ending with a summary of the future outlook in this field.
Collapse
Affiliation(s)
| | | | | | - W. Mark Saltzman
- Department of Biomedical Engineering
- Department of Chemical & Environmental Engineering
- Department of Cellular & Molecular Physiology
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06510
| |
Collapse
|
4
|
Chen CH, Lin YJ, Cheng LT, Lin CH, Ke GM. Poloxamer-188 Adjuvant Efficiently Maintains Adaptive Immunity of SARS-CoV-2 RBD Subunit Vaccination through Repressing p38MAPK Signaling. Vaccines (Basel) 2022; 10:vaccines10050715. [PMID: 35632471 PMCID: PMC9145454 DOI: 10.3390/vaccines10050715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Poloxamer-188 (P188) is a nonionic triblock linear copolymer that can be used as a pharmaceutical excipient because of its amphiphilic nature. This study investigated whether P188 can act as an adjuvant to improve the immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor binding domain (RBD) subunit vaccine. BALB/c mice were vaccinated twice with the RBD antigen alone or in combination with P188 or MF59 (a commercial adjuvant for comparison purposes). The resulting humoral and cellular immunity were assessed. Results showed that P188 helped elicit higher neutralizing activity than MF59 after vaccination. P188 induced significant humoral immune response, along with type 1 T helper (Th1) and type 2 T helper (Th2) cellular immune response when compared with MF59 due to repressing p38MAPK phosphorylation. Furthermore, P188 did not result in adverse effects such as fibrosis of liver or kidney after vaccination. In conclusion, P188 is a novel adjuvant that may be used for safe and effective immune enhancement of the SARS-CoV-2 RBD antigen.
Collapse
Affiliation(s)
- Chao-Hung Chen
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 10650, Taiwan; (C.-H.C.); (Y.-J.L.); (L.-T.C.)
- General Research Service Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Yu-Jen Lin
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 10650, Taiwan; (C.-H.C.); (Y.-J.L.); (L.-T.C.)
- Country Best Biotech Co., Ltd., Taipei 100411, Taiwan;
| | - Li-Ting Cheng
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 10650, Taiwan; (C.-H.C.); (Y.-J.L.); (L.-T.C.)
| | | | - Guan-Ming Ke
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 10650, Taiwan; (C.-H.C.); (Y.-J.L.); (L.-T.C.)
- Correspondence: ; Tel.: +886-08-7703202 (ext. 5052)
| |
Collapse
|
5
|
Gencer A, Baysal I, Nemutlu E, Yabanoglu-Ciftci S, Arica B. Efficacy of Sirna-Loaded Nanoparticles in the Treatment of k-ras Mutant Lung Cancer in vitro. J Microencapsul 2022; 39:261-275. [PMID: 35356841 DOI: 10.1080/02652048.2022.2061058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIM To design and develop K-RAS silencing small interfering RNA (siRNA)-loaded poly (D, L-lactic-co-glycolic acid) nanoparticles and evaluate their efficacy in the treatment of K-RAS mutant lung cancer. METHODS The nanoparticles prepared by the double emulsion solvent evaporation method were characterized by TEM, FTIR and XPS analyzes and evaluated in vitro by XTT, PCR, ELISA, and Western-Blot. Metabolomic analyzes were performed to evaluate the changes in metabolic profiles of the cells after nanoparticles treatment. RESULTS The nanoparticles were obtained with a particle size less than 250 nm, a polydispersity index around 0.1, a surface charge of (-12) - (+14) mV, and 80% of the siRNA encapsulation. The nanoparticles didn't affect cell viability of the cells after 72 hours. In cancer cells, KRAS expression was decreased by up to 50%, protein levels were decreased by more than 90%. CONCLUSION The formulated siRNA delivery nanoparticles can be promising treatment in lung cancer.
Collapse
Affiliation(s)
- Ayse Gencer
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | | | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
6
|
Khabazian E, Vakhshiteh F, Norouzi P, Fatahi Y, Dinarvand R, Atyabi F. Cationic Liposome Decorated with Cyclic RGD Peptide for Targeted Delivery of anti-STAT3 siRNA to Melanoma Cancer Cells. J Drug Target 2021; 30:522-533. [PMID: 34482780 DOI: 10.1080/1061186x.2021.1973481] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Gene therapy is regarded as a valuable strategy for efficient cancer treatment. However, the design of effective delivery systems that can deliver gene materials such as siRNA specifically to the tumour tissues plays a pivotal role in cancer therapy. For this reason, a targeted cationic liposome for melanoma treatment was developed. This system consists of cyclic RGD peptide conjugated to DSPE-PEG2000, cholesterol, DOTAP, and DSPC as cationic and neutral lipids, respectively. Cyclic RGD was selected based on speculation that cyclic RGD would effectively transport anti-signal transducer and activator of transcription 3 (STAT3) siRNA into melanoma cell via integrin receptors. The prepared liposomes provided excellent stability against electrolyte and serum nucleases. Targeted liposomes remarkably exhibited higher cellular internalisation in comparison with the non-targeted system in flow cytometry and confocal microscopy. Furthermore, incorporating peptide on the surface of liposomes resulted in considerably high cytotoxicity, a 2.1-times raise in apoptosis induction, and a significantly enhanced STAT3 gene suppression as compared with the corresponding non-targeted formulation on B16F10 murine melanoma cells. Whole-body imaging confirmed the more significant tumour accumulation of targeted liposomes in B16F10 melanoma xenograft tumour-bearing mice. Consequently, c-RGD peptide modified liposome suggests a promising option for specific siRNA delivery into melanoma cells.
Collapse
Affiliation(s)
- Ehsan Khabazian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran
| | - Faezeh Vakhshiteh
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Norouzi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box 1417614411, Tehran, Iran.,Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Chen K, Zhang Y, Qian L, Wang P. Emerging strategies to target RAS signaling in human cancer therapy. J Hematol Oncol 2021; 14:116. [PMID: 34301278 PMCID: PMC8299671 DOI: 10.1186/s13045-021-01127-w] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
RAS mutations (HRAS, NRAS, and KRAS) are among the most common oncogenes, and around 19% of patients with cancer harbor RAS mutations. Cells harboring RAS mutations tend to undergo malignant transformation and exhibit malignant phenotypes. The mutational status of RAS correlates with the clinicopathological features of patients, such as mucinous type and poor differentiation, as well as response to anti-EGFR therapies in certain types of human cancers. Although RAS protein had been considered as a potential target for tumors with RAS mutations, it was once referred to as a undruggable target due to the consecutive failure in the discovery of RAS protein inhibitors. However, recent studies on the structure, signaling, and function of RAS have shed light on the development of RAS-targeting drugs, especially with the approval of Lumakras (sotorasib, AMG510) in treatment of KRASG12C-mutant NSCLC patients. Therefore, here we fully review RAS mutations in human cancer and especially focus on emerging strategies that have been recently developed for RAS-targeting therapy.
Collapse
Affiliation(s)
- Kun Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yalei Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling Qian
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
8
|
Inhalable hybrid nanocarriers for respiratory disorders. TARGETING CHRONIC INFLAMMATORY LUNG DISEASES USING ADVANCED DRUG DELIVERY SYSTEMS 2020. [PMCID: PMC7499343 DOI: 10.1016/b978-0-12-820658-4.00013-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Rapid advancements in the field of drug delivery lead to increased use of inhalable formulations as they are cost effective, noninvasive, and targeted and have less systemic side effects and above all better patient compliance. Development of inhalable hybrid systems has offered manifold advantages to this area of drug delivery. Inclusion of polymer and lipid, inorganic and organic substances, and metallic nanoparticles all of them aim to achieve codelivery of drugs which are incompatible in single phase systems. The recent progress in nanotechnology has gained momentum toward delivery of siRNA and miRNA and vaccines to the targeted site. The present work is an attempt to compile all the hybrid and inhalable systems to give readers an overview toward this delivery system as much more work is needed in this field to achieve better resolution of inflammatory disorders.
Collapse
|
9
|
Sacko K, Thangavel K, Shoyele SA. Codelivery of Genistein and miRNA-29b to A549 Cells Using Aptamer-Hybrid Nanoparticle Bioconjugates. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1052. [PMID: 31340494 PMCID: PMC6669731 DOI: 10.3390/nano9071052] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 11/24/2022]
Abstract
This study aimed to evaluate the anti-cancer effect of a combination therapy of miRNA-29b and genistein loaded in mucin-1 (MUC 1)-aptamer functionalized hybrid nanoparticles in non-small cell lung cancer (NSCLC) A549 cell line. Genistein-miRNA-29b-loaded hybrid nanoparticles (GMLHN) was prepared and characterized. Particle size and zeta potential were measured using photon correlation spectroscopy (PCS). Encapsulation efficiency and loading efficiency were determined using HPLC. Preferential internalization of MUC 1-aptamer functionalized GMLHN by A549 cells was evaluated and compared to normal MRC-5 cells. The ability of GMLHN to downregulate targeted oncoproteins Phosphorylated protein kinase, strain AK, Thymoma (Phosphorylated protein kinase B) (pAKT), Phosphorylated phosphoinositide 3-kinase (p-PI3K), DNA (cytosine-5-)-methyltransferase 3 beta (DNMT3B) and Myeloid Cell Leukemia Sequence 1 (MCL 1) was evaluated using western blot, while antiproliferative effect and ability to initiate apoptosis was also assessed in A549 cells. MUC 1-aptamer functionalized GMLHN nanoparticles were prepared. These nanoparticles were preferentially internalized by A549 cells but less so, in MRC-5 cells. pAKT, p-PI3K, DNMT3B and MCL 1 were efficiently downregulated by these nanoparticles without affecting the levels of AKT and PI3K in A549 cells. GMLHN demonstrated a superior antiproliferative effect compared to individual genistein and miRNA-29b-loaded nanoparticles. Results generated were able to demonstrate that genistein-miRNA-29b-loaded hybrid nanoparticles (GMLHN) could be a potential treatment modality for NSCLC because of the ability of the payloads to attack multiple targets.
Collapse
Affiliation(s)
- Koita Sacko
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Karthik Thangavel
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sunday A Shoyele
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
10
|
Cunningham RM, Hickey AM, Wilson JW, Plakos KJI, DeRose VJ. Pt-induced crosslinks promote target enrichment and protection from serum nucleases. J Inorg Biochem 2018; 189:124-133. [PMID: 30245274 PMCID: PMC7703794 DOI: 10.1016/j.jinorgbio.2018.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/23/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022]
Abstract
Identifying the interactions of small molecules with biomolecules in complex cellular environments is a significant challenge. As one important example, despite being widely used for decades, much is still not understood regarding the cellular targets of Pt(II)-based anticancer drugs. In this study we introduce a novel method for isolation of Pt(II)-bound biomolecules using a DNA hybridization pull-down approach. Using a modified Pt reagent, click-ligation of a DNA oligonucleotide to both a Pt(II)-bound DNA hairpin and bovine serum albumin (BSA) are demonstrated. Subsequent hybridization to a biotin-labeled oligonucleotide allows for efficient isolation of Pt(II)-bound species by streptavidin pulldown. We also find that platinated bovine serum albumin readily crosslinks to DNA in the absence of click ligation, and that a fraction of BSA-bound Pt(II) can transfer to DNA over time. Interestingly, in in vitro studies, fragmented mammalian DNA that is crosslinked to BSA through Pt(II) exhibits significantly increased protection from degradation by serum nucleases.
Collapse
Affiliation(s)
- Rachael M Cunningham
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA; Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Anna M Hickey
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA
| | - Jesse W Wilson
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA; Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Kory J I Plakos
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA; Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Victoria J DeRose
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA; Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
11
|
Zarredar H, Pashapour S, Ansarin K, Khalili M, Baghban R, Farajnia S. Combination therapy with KRAS siRNA and EGFR inhibitor AZD8931 suppresses lung cancer cell growth in vitro. J Cell Physiol 2018; 234:1560-1566. [DOI: 10.1002/jcp.27021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 06/26/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center Tabriz University of Medical Science Tabriz Iran
- Students Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Shadi Pashapour
- Department of Genetic Tabriz Branch, Islamic Azad University Tabriz Iran
| | - Khalil Ansarin
- Department of Genetic Tabriz Branch, Islamic Azad University Tabriz Iran
| | - Majid Khalili
- Department of Basic Science Maragheh University of Medical Science, Maragheh Iran
| | - Roghayyeh Baghban
- Drug Applied Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Safar Farajnia
- Department of Genetic Tabriz Branch, Islamic Azad University Tabriz Iran
- Biotechnology Research Center Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
12
|
Abstract
Nanoparticle delivery systems offer advantages over free drugs, in that they increase solubility and biocompatibility. Nanoparticles can deliver a high payload of therapeutic molecules while limiting off-target side effects. Therefore, delivery of an existing drug with a nanoparticle frequently results in an increased therapeutic index. Whether of synthetic or biologic origin, nanoparticle surface coatings are often required to reduce immune clearance and thereby increase circulation times allowing the carriers to reach their target site. To this end, polyethylene glycol (PEG) has long been used, with several PEGylated products reaching clinical use. Unfortunately, the growing use of PEG in consumer products has led to an increasing prevalence of PEG-specific antibodies in the human population, which in turn has fueled the search for alternative coating strategies. This review highlights alternative bioinspired nanoparticle shielding strategies, which may be more beneficial moving forward than PEG and other synthetic polymer coatings.
Collapse
Affiliation(s)
- Neetu M. Gulati
- Department of Pharmacology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Cleveland Center for Membrane and Structural Biology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Phoebe L. Stewart
- Department of Pharmacology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Cleveland Center for Membrane and Structural Biology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Radiology, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Materials Science and Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Department of Macromolecular Science and Engineering, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Division of General Medical Sciences Oncology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
13
|
Thangavel C, Perepelyuk M, Boopathi E, Liu Y, Polischak S, Deshpande DA, Rafiq K, Dicker AP, Knudsen KE, Shoyele SA, Den RB. Improvement in Therapeutic Efficacy and Reduction in Cellular Toxicity: Introduction of a Novel Anti-PSMA-Conjugated Hybrid Antiandrogen Nanoparticle. Mol Pharm 2018; 15:1778-1790. [PMID: 29616555 DOI: 10.1021/acs.molpharmaceut.7b01024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Second generation antiandrogens have improved overall survival for men with metastatic castrate resistant prostate cancer; however, the antiandrogens result in suppression of androgen receptor (AR) activity in all tissues resulting in dose limiting toxicity. We sought to overcome this limitation through encapsulation in a prostate specific membrane antigen (PSMA)-conjugated nanoparticle. We designed and characterized a novel nanoparticle containing an antiandrogen, enzalutamide. Selectivity and enhanced efficacy was achieved through coating the particle with PSMA. The PSMA-conjugated nanoparticle was internalized selectively in AR expressing prostate cancer cells. It did not elicit an inflammatory effect. The efficacy of enzalutamide was not compromised through insertion into the nanoparticle; in fact, lower systemic drug concentrations of enzalutamide resulted in comparable clinical activity. Normal muscle cells were not impacted by the PSMA-conjugated containing antiandrogen. This approach represents a novel strategy to increase the specificity and effectiveness of antiandrogen treatment for men with castrate resistant prostate cancer. The ability to deliver higher drug concentrations in prostate cancer cells may translate into improved clinical end points including overall survival.
Collapse
|
14
|
Perepelyuk M, Sacko K, Thangavel K, Shoyele SA. Evaluation of MUC1-Aptamer Functionalized Hybrid Nanoparticles for Targeted Delivery of miRNA-29b to Nonsmall Cell Lung Cancer. Mol Pharm 2018; 15:985-993. [PMID: 29432024 DOI: 10.1021/acs.molpharmaceut.7b00900] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The objective of this study was to evaluate the therapeutic efficacy and pharmacokinetic study of mucin1-aptamer functionalized miRNA-29b-loaded hybrid nanoparticles (MAFMILHNs) in lung tumor-bearing SCID mice. MAFMILHNs were manufactured using an isoelectric point based nanotechnology. They were then fully characterized for particle size, loading capacity, zeta potential, and encapsulation efficiency. The ability of MAFMILHNs to downregulate oncoprotein DNMT3B both at the cellular level and in vivo was monitored using Western blot, while the effect of the downregulation of DNMT3B on tumor growth was assessed using bioluminescence. Results indicate that the presence of MUC1-aptamer conjugated to the surface of the nanoparticles enhanced the selective delivery of miRNA-29b to tumor cells and tissues. Further, the downregulation of DNMT3B by MAFMILHNs resulted in the inhibition of tumor growth in mouse models.
Collapse
Affiliation(s)
- Maryna Perepelyuk
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| | - Koita Sacko
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| | - Karthik Thangavel
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| | - Sunday A Shoyele
- Department of Pharmaceutical Sciences, College of Pharmacy , Thomas Jefferson University , Philadelphia , Pennsylvania 19107 , United States
| |
Collapse
|
15
|
Wen ZM, Jie J, Zhang Y, Liu H, Peng LP. A self-assembled polyjuglanin nanoparticle loaded with doxorubicin and anti-Kras siRNA for attenuating multidrug resistance in human lung cancer. Biochem Biophys Res Commun 2017; 493:1430-1437. [PMID: 28958938 DOI: 10.1016/j.bbrc.2017.09.132] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 09/23/2017] [Indexed: 01/08/2023]
Abstract
Lung cancer is a leading cause of cancer-associated mortality worldwide, which has a low survival rate. Multidrug resistance (MDR) is a major obstacle that hinders the treatment of lung cancer. Doxorubicin (DOX) is an anthracycline glycoside antibiotic, having a broad spectrum of anticancer activity against various solid tumors. Juglanin is a natural production, mainly extracted from green walnut husks of Juglans mandshurica, exhibiting various bioactivities. Here, we demonstrated that the combination of drug, gene and nanoparticle overcame MDR, inhibiting lung cancer progression. A novel nanoparticular pre-chemosensitizer was applied to develop a self-assembled nanoparticle formula of amphiphilic poly(juglanin (Jug) dithiodipropionic acid (DA))-b-poly(ethylene glycol) (PEG)-siRNA Kras with DOX in the core (DOX/PJAD-PEG-siRNA). The formed nanoparticles, appeared spherical shape, had mean particle size of 81.8 nm, and the zeta potential was -18.62 mV. The in vitro drug release results suggested that a sustained release was observed in DOX/PJAD-PEG-siRNA nanoparticles compared to the free DOX. Jug could improve the cytotoxicity of DOX to cancer cells with MDR. Oncogene, Kras, was dose-dependently reduced by treatment of DOX/PJAD-PEG-siRNA nanoparticles. Additionally, P-glycoprotein (MDR1) and c-Myc, contributing to tumor progression, were suppressed by the nanoparticles, while p53 was improved in drug-resistant cells. Colony formation analysis suggested that DOX/PJAD-PEG-siRNA nanoparticles showed the most effective role in reducing cancer cell proliferation. In vivo, DOX/PJAD-PEG-siRNA nanoparticles reduced tumor growth compared to the free DOX, accompanied with reduced KI-67 and enhanced TUNEL positive levels in drug-resistant xenografted nude mice. Thus, the findings above indicated that juglanin, as a chemosensitizer, potentiate the anti-cancer role of DOX in drug-resistant cancer cells. And the nanoparticles exhibited stronger antitumor efficiency, suggesting potential value in the treatment of lung cancer.
Collapse
Affiliation(s)
- Zhong-Mei Wen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Jing Jie
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuan Zhang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Han Liu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Li-Ping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
16
|
Gu L, Deng ZJ, Roy S, Hammond PT. A Combination RNAi-Chemotherapy Layer-by-Layer Nanoparticle for Systemic Targeting of KRAS/P53 with Cisplatin to Treat Non-Small Cell Lung Cancer. Clin Cancer Res 2017; 23:7312-7323. [PMID: 28912139 DOI: 10.1158/1078-0432.ccr-16-2186] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 06/22/2017] [Accepted: 09/07/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Mutation of the Kirsten ras sarcoma viral oncogene homolog (KRAS) and loss of p53 function are commonly seen in patients with non-small cell lung cancer (NSCLC). Combining therapeutics targeting these tumor-defensive pathways with cisplatin in a single-nanoparticle platform are rarely developed in clinic.Experimental Design: Cisplatin was encapsulated in liposomes, which multiple polyelectrolyte layers, including siKRAS and miR-34a were built on to generate multifunctional layer-by-layer nanoparticle. Structure, size, and surface charge were characterized, in addition to in vitro toxicity studies. In vivo tumor targeting and therapy was investigated in an orthotopic lung cancer model by microCT, fluorescence imaging, and immunohistochemistry.Results: The singular nanoscale formulation, incorporating oncogene siKRAS, tumor-suppressor stimulating miR-34a, and cisplatin, has shown enhanced toxicity against lung cancer cell line, KP cell. In vivo, systemic delivery of the nanoparticles indicated a preferential uptake in lung of the tumor-bearing mice. Efficacy studies indicated prolonged survival of mice from the combination treatment.Conclusions: The combination RNA-chemotherapy in an LbL formulation provides an enhanced treatment efficacy against NSCLC, indicating promising potential in clinic. Clin Cancer Res; 23(23); 7312-23. ©2017 AACR.
Collapse
Affiliation(s)
- Li Gu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Zhou J Deng
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sweta Roy
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts. .,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
17
|
Madni A, Batool A, Noreen S, Maqbool I, Rehman F, Kashif PM, Tahir N, Raza A. Novel nanoparticulate systems for lung cancer therapy: an updated review. J Drug Target 2017; 25:499-512. [PMID: 28151021 DOI: 10.1080/1061186x.2017.1289540] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world. Conventional therapy for lung cancer is associated with lack of specificity and access to the normal cells resulting in cytotoxicity, reduced cellular uptake, drug resistance and rapid drug clearance from the body. The emergence of nanotechnology has revolutionized the treatment of lung cancer. The focus of nanotechnology is to target tumor cells with improved bioavailability and reduced toxicity. In the recent years, nanoparticulate systems have extensively been exploited in order to overcome the obstacles in treatment of lung cancer. Nanoparticulate systems have shown much potential for lung cancer therapy by gaining selective access to the tumor cells due to surface modifiability and smaller size. In this review, various novel nanoparticles (NPs) based formulations have been discussed in the treatment of lung cancer. Nanotechnology is expected to grow fast in future, and it will provide new avenues for the improved treatment of lung cancer. This review article also highlights the characteristics, recent advances in the designing of NPs and therapeutic outcomes.
Collapse
Affiliation(s)
- Asadullah Madni
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Amna Batool
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Sobia Noreen
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Irsah Maqbool
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Faizza Rehman
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Prince Muhammad Kashif
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Nayab Tahir
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| | - Ahmad Raza
- a Department of Pharmacy, Faculty of Pharmacy & Alternative Medicine , The Islamia University of Bahawalpur , Bahawalpur , Pakistan
| |
Collapse
|
18
|
Perepelyuk M, Shoyele O, Birbe R, Thangavel C, Liu Y, Den RB, Snook AE, Lu B, Shoyele SA. siRNA-Encapsulated Hybrid Nanoparticles Target Mutant K-ras and Inhibit Metastatic Tumor Burden in a Mouse Model of Lung Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 6:259-268. [PMID: 28325292 PMCID: PMC5363504 DOI: 10.1016/j.omtn.2016.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 10/26/2022]
Abstract
There is an unmet need in the development of an effective therapy for mutant K-ras-expressing non-small-cell lung cancer (NSCLC). Although various small molecules have been evaluated, an effective therapy remains a dream. siRNAs have the potential to downregulate mutant K-ras both at the protein and mRNA levels. However, a safe and effective delivery of siRNAs to tumors remains a limitation to their translational application in the treatment of this highly debilitating disease. Here we developed a novel hybrid nanoparticle carrier for effective delivery of anti-mutant K-ras to NSCLC (AKSLHN). The ability of this treatment modality to regress lung tumors in mouse models was evaluated as a monotherapy or as a combination treatment with erlotinib. Further, the toxicity of this treatment modality to healthy tissues was evaluated, along with its ability to elicit immune/inflammatory reactions. The results suggest that this treatment modality is a promising prospect for the treatment of mutant K-ras-expressing NSCLC without any accompanying toxicity. However, further understanding of the cellular-level interaction between AHSLHN and erlotinib needs to be attained before this promising treatment modality can be brought to the bedside.
Collapse
Affiliation(s)
- Maryna Perepelyuk
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Olubunmi Shoyele
- Department of Pathology and Laboratory Medicine, Western Connecticut Health Network, Danbury Hospital, Danbury, CT 06810, USA
| | - Ruth Birbe
- Department of Pathology and Laboratory Medicine, Cooper University Hospital-MD Anderson Cancer Center, Camden, NJ 08103, USA
| | | | - Yi Liu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia 19107, PA, USA
| | - Bo Lu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sunday A Shoyele
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
19
|
Perepelyuk M, Maher C, Lakshmikuttyamma A, Shoyele SA. Aptamer-hybrid nanoparticle bioconjugate efficiently delivers miRNA-29b to non-small-cell lung cancer cells and inhibits growth by downregulating essential oncoproteins. Int J Nanomedicine 2016; 11:3533-44. [PMID: 27555773 PMCID: PMC4970448 DOI: 10.2147/ijn.s110488] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are potentially attractive candidates for cancer therapy. However, their therapeutic application is limited by lack of availability of an efficient delivery system to stably deliver these potent molecules intracellularly to cancer cells while avoiding healthy cells. We developed a novel aptamer-hybrid nanoparticle bioconjugate delivery system to selectively deliver miRNA-29b to MUC1-expressing cancer cells. Significant downregulation of oncoproteins DNMT3b and MCL1 was demonstrated by these MUC1 aptamer-functionalized hybrid nanoparticles in A549 cells. Furthermore, downregulation of these oncoproteins led to antiproliferative effect and induction of apoptosis in a superior version when compared with Lipofectamine 2000. This novel aptamer-hybrid nanoparticle bioconjugate delivery system could potentially serve as a platform for intracellular delivery of miRNAs to cancer cells, hence improving the therapeutic outcome of lung cancer.
Collapse
Affiliation(s)
- Maryna Perepelyuk
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christina Maher
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ashakumary Lakshmikuttyamma
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sunday A Shoyele
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
20
|
Perepelyuk M, Thangavel C, Liu Y, Den RB, Lu B, Snook AE, Shoyele SA. Biodistribution and Pharmacokinetics Study of siRNA-loaded Anti-NTSR1-mAb-functionalized Novel Hybrid Nanoparticles in a Metastatic Orthotopic Murine Lung Cancer Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e282. [PMID: 26812654 PMCID: PMC5012553 DOI: 10.1038/mtna.2015.56] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/07/2015] [Indexed: 11/09/2022]
Abstract
Small interfering RNA (siRNA) is effective in silencing critical molecular pathways in cancer. The use of this tool as a treatment modality is limited by lack of an intelligent carrier system to enhance the preferential delivery of this molecule to specific targets in vivo. In the present study, the in vivo behavior of novel anti-NTSR1-mAb-functionalized antimutant K-ras siRNA-loaded hybrid nanoparticles, delivered by i.p. injection to non-small-cell lung cancer in mice models, was investigated and compared to that of a naked siRNA formulation. The siRNA in anti-NTSR1-mAb-functionalized hybrid nanoparticles was preferentially accumulated in tumor-bearing lungs and metastasized tumor for at least 48 hours while the naked siRNA formulation showed lack of preferential accumulation in all of the organs monitored. The plasma terminal half-life of nanoparticle-delivered siRNA was 11 times higher (17-1.5 hours) than that of the naked siRNA formulation. The mean residence time and AUClast were 3.4 and 33 times higher than the corresponding naked siRNA formulation, respectively. High-performance liquid chromatography analysis showed that the hybrid nanoparticle carrier system protected the encapsulated siRNA against degradation in vivo. Our novel anti-NTSR1-mAb-functionalized hybrid nanoparticles provide a useful platform for in vivo targeting of siRNA for both experimental and clinical purposes.
Collapse
Affiliation(s)
- Maryna Perepelyuk
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Yi Liu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Bo Lu
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sunday A Shoyele
- Department of Pharmaceutical Science, College of Pharmacy, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Novel targeted siRNA-loaded hybrid nanoparticles: preparation, characterization and in vitro evaluation. J Nanobiotechnology 2015; 13:61. [PMID: 26410728 PMCID: PMC4583992 DOI: 10.1186/s12951-015-0124-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/16/2015] [Indexed: 12/24/2022] Open
Abstract
Background siRNAs have a high potential for silencing critical molecular pathways that are pathogenic. Nevertheless, their clinical application has been limited by a lack of effective and safe nanotechnology-based delivery system that allows a controlled and safe transfection to cytosol of targeted cells without the associated adverse effects. Our group recently reported a very effective and safe hybrid nanoparticle delivery system composing human IgG and poloxamer-188 for siRNA delivery to cancer cells. However, these nanoparticles need to be optimized in terms of particle size, loading capacity and encapsulation efficiency. In the present study, we explored the effects of certain production parameters on particle size, loading capacity and encapsulation efficiency. Further, to make these nanoparticles more specific in their delivery of siRNA, we conjugated anti-NTSR1-mAb to the surface of these nanoparticles to target NTSR1-overexpressing cancer cells. The mechanism of siRNA release from these antiNTSR1-mAb functionalized nanoparticles was also elucidated. Results It was demonstrated that the concentration of human IgG in the starting nanoprecipitation medium and the rotation speed of the magnetic stirrer influenced the encapsulation efficiency, loading capacity and the size of the nanoparticles produced. We also successfully transformed these nanoparticles into actively targeted nanoparticles by functionalizing with anti-NTSR1-mAb to specifically target NTSR1-overexpressing cancer cells, hence able to avoid undesired accumulation in normal cells. The mechanism of siRNA release from these nanoparticles was elucidated to be by Fickian diffusion. Using flow cytometry and fluorescence microscopy, we were able to confirm the active involvement of NTSR1 in the uptake of these anti-NTSR1-mAb functionalized hybrid nanoparticles by lung adenocarcinoma cells. Conclusions This hybrid nanoparticle delivery system can be used as a platform technology for intracellular delivery of siRNAs to NTSR1-overexpressing tumor cells.
Collapse
|