1
|
Wu X, Wang F, Yang X, Gong Y, Niu T, Chu B, Qu Y, Qian Z. Advances in Drug Delivery Systems for the Treatment of Acute Myeloid Leukemia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403409. [PMID: 38934349 DOI: 10.1002/smll.202403409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Acute myeloid leukemia (AML) is a common and catastrophic hematological neoplasm with high mortality rates. Conventional therapies, including chemotherapy, hematopoietic stem cell transplantation (HSCT), immune therapy, and targeted agents, have unsatisfactory outcomes for AML patients due to drug toxicity, off-target effects, drug resistance, drug side effects, and AML relapse and refractoriness. These intrinsic limitations of current treatments have promoted the development and application of nanomedicine for more effective and safer leukemia therapy. In this review, the classification of nanoparticles applied in AML therapy, including liposomes, polymersomes, micelles, dendrimers, and inorganic nanoparticles, is reviewed. In addition, various strategies for enhancing therapeutic targetability in nanomedicine, including the use of conjugating ligands, biomimetic-nanotechnology, and bone marrow targeting, which indicates the potential to reverse drug resistance, are discussed. The application of nanomedicine for assisting immunotherapy is also involved. Finally, the advantages and possible challenges of nanomedicine for the transition from the preclinical phase to the clinical phase are discussed.
Collapse
Affiliation(s)
- Xia Wu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Fangfang Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xijing Yang
- The Experimental Animal Center of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuping Gong
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
2
|
Alimohammadvand S, Kaveh Zenjanab M, Mashinchian M, Shayegh J, Jahanban-Esfahlan R. Recent advances in biomimetic cell membrane-camouflaged nanoparticles for cancer therapy. Biomed Pharmacother 2024; 177:116951. [PMID: 38901207 DOI: 10.1016/j.biopha.2024.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
The emerging strategy of biomimetic nanoparticles (NPs) via cellular membrane camouflage holds great promise in cancer therapy. This scholarly review explores the utilization of cellular membranes derived from diverse cellular entities; blood cells, immune cells, cancer cells, stem cells, and bacterial cells as examples of NP coatings. The camouflaging strategy endows NPs with nuanced tumor-targeting abilities such as self-recognition, homotypic targeting, and long-lasting circulation, thus also improving tumor therapy efficacy overall. The comprehensive examination encompasses a variety of cell membrane camouflaged NPs (CMCNPs), elucidating their underlying targeted therapy mechanisms and delineating diverse strategies for anti-cancer applications. Furthermore, the review systematically presents the synthesis of source materials and methodologies employed in order to construct and characterize these CMCNPs, with a specific emphasis on their use in cancer treatment.
Collapse
Affiliation(s)
- Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Mashinchian
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Shayegh
- Department of Microbiology, Faculty of Veterinary and Agriculture, Islamic Azad University, Shabestar branch, Shabestar, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Li J, Wang Q, Han Y, Jiang L, Lu S, Wang B, Qian W, Zhu M, Huang H, Qian P. Development and application of nanomaterials, nanotechnology and nanomedicine for treating hematological malignancies. J Hematol Oncol 2023; 16:65. [PMID: 37353849 PMCID: PMC10290401 DOI: 10.1186/s13045-023-01460-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/30/2023] [Indexed: 06/25/2023] Open
Abstract
Hematologic malignancies (HMs) pose a serious threat to patients' health and life, and the five-year overall survival of HMs remains low. The lack of understanding of the pathogenesis and the complex clinical symptoms brings immense challenges to the diagnosis and treatment of HMs. Traditional therapeutic strategies for HMs include radiotherapy, chemotherapy, targeted therapy and hematopoietic stem cell transplantation. Although immunotherapy and cell therapy have made considerable progress in the last decade, nearly half of patients still relapse or suffer from drug resistance. Recently, studies have emerged that nanomaterials, nanotechnology and nanomedicine show great promise in cancer therapy by enhancing drug targeting, reducing toxicity and side effects and boosting the immune response to promote durable immunological memory. In this review, we summarized the strategies of recently developed nanomaterials, nanotechnology and nanomedicines against HMs and then proposed emerging strategies for the future designment of nanomedicines to treat HMs based on urgent clinical needs and technological progress.
Collapse
Affiliation(s)
- Jinxin Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Qiwei Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Yingli Han
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Lingli Jiang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Siqi Lu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Beini Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Wenchang Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Meng Zhu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - He Huang
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
| |
Collapse
|
4
|
Mendonça MCP, Kont A, Kowalski PS, O'Driscoll CM. Design of lipid-based nanoparticles for delivery of therapeutic nucleic acids. Drug Discov Today 2023; 28:103505. [PMID: 36708760 DOI: 10.1016/j.drudis.2023.103505] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/04/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023]
Abstract
The successful development of nonviral delivery systems for nucleic acids has been reported extensively over the past number of years. Among them, lipid-based nanoparticles (LNPs) represent the most advanced platform. This review provides an overview of the state-of-the-art in LNP technology, focusing on the delivery of a range of nucleic acids. Recent advances in the development of an efficient and safe lipid-based system are critically analyzed with a particular emphasis on the rationale behind the design of LNPs and on attempts to elucidate the resulting molecular assembly and structure, their interactions with cellular proteins and biodistribution. In addition, manufacturing methods including microfluidics and their potential to influence stability and scale-up are summarized.
Collapse
Affiliation(s)
| | - Ayse Kont
- School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| | - Piotr S Kowalski
- School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| | | |
Collapse
|
5
|
Dhas N, García MC, Kudarha R, Pandey A, Nikam AN, Gopalan D, Fernandes G, Soman S, Kulkarni S, Seetharam RN, Tiwari R, Wairkar S, Pardeshi C, Mutalik S. Advancements in cell membrane camouflaged nanoparticles: A bioinspired platform for cancer therapy. J Control Release 2022; 346:71-97. [PMID: 35439581 DOI: 10.1016/j.jconrel.2022.04.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022]
Abstract
The idea of employing natural cell membranes as a coating medium for nanoparticles (NPs) endows man-made vectors with natural capabilities and benefits. In addition to retaining the physicochemical characteristics of the NPs, the biomimetic NPs also have the functionality of source cell membranes. It has emerged as a promising approach to enhancing the properties of NPs for drug delivery, immune evasion, imaging, cancer-targeting, and phototherapy sensitivity. Several studies have been reported with a multitude of approaches to reengineering the surface of NPs using biological membranes. Owing to their low immunogenicity and intriguing biomimetic properties, cell-membrane-based biohybrid delivery systems have recently gained a lot of interest as therapeutic delivery systems. This review summarises different kinds of biomimetic NPs reported so far, their fabrication aspects, and their application in the biomedical field. Finally, it briefs on the latest advances available in this biohybrid concept.
Collapse
Affiliation(s)
- Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Mónica C García
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Ciudad Universitaria, X5000HUA Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Ritu Kudarha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Gasper Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Raviraja N Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ruchi Tiwari
- Pranveer Singh Institute of Technology, Kanpur, Uttar Pradesh 209305, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, 400056, India
| | - Chandrakantsing Pardeshi
- R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| |
Collapse
|
6
|
Multifunctional liposomal nanostructure-mediated siRNA/bortezomib co-delivery for SHARP1 knockdown in MLL-AF6 acute myeloid leukemia. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 134:112663. [DOI: 10.1016/j.msec.2022.112663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/17/2022]
|
7
|
Askenase PW. Ancient Evolutionary Origin and Properties of Universally Produced Natural Exosomes Contribute to Their Therapeutic Superiority Compared to Artificial Nanoparticles. Int J Mol Sci 2021; 22:1429. [PMID: 33572657 PMCID: PMC7866973 DOI: 10.3390/ijms22031429] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs), such as exosomes, are newly recognized fundamental, universally produced natural nanoparticles of life that are seemingly involved in all biologic processes and clinical diseases. Due to their universal involvements, understanding the nature and also the potential therapeutic uses of these nanovesicles requires innovative experimental approaches in virtually every field. Of the EV group, exosome nanovesicles and larger companion micro vesicles can mediate completely new biologic and clinical processes dependent on the intercellular transfer of proteins and most importantly selected RNAs, particularly miRNAs between donor and targeted cells to elicit epigenetic alterations inducing functional cellular changes. These recipient acceptor cells are nearby (paracrine transfers) or far away after distribution via the circulation (endocrine transfers). The major properties of such vesicles seem to have been conserved over eons, suggesting that they may have ancient evolutionary origins arising perhaps even before cells in the primordial soup from which life evolved. Their potential ancient evolutionary attributes may be responsible for the ability of some modern-day exosomes to withstand unusually harsh conditions, perhaps due to unique membrane lipid compositions. This is exemplified by ability of the maternal milk exosomes to survive passing the neonatal acid/enzyme rich stomach. It is postulated that this resistance also applies to their durable presence in phagolysosomes, thus suggesting a unique intracellular release of their contained miRNAs. A major discussed issue is the generally poorly realized superiority of these naturally evolved nanovesicles for therapies when compared to human-engineered artificial nanoparticles, e.g., for the treatment of diseases like cancers.
Collapse
Affiliation(s)
- Phillip W Askenase
- Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
8
|
Qiu Y, Li A, Lee J, Lee JE, Lee EW, Cho, N, Yoo HM. Inhibition of Jurkat T Cell Proliferation by Active Components of Rumex japonicus Roots Via Induced Mitochondrial Damage and Apoptosis Promotion. J Microbiol Biotechnol 2020; 30:1885-1895. [PMID: 33144550 PMCID: PMC9728342 DOI: 10.4014/jmb.2007.07018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022]
Abstract
Rumex japonicus Houtt (RJH) is a valuable plant used in traditional medicine to treat several diseases, such as scabies and jaundice. In this study, Jurkat cell growth inhibitory extracts of R. japonicus roots were subjected to bioassay-guided fractionation, resulting in the isolation of three naphthalene derivatives (3-5) along with one anthraquinone (6) and two phenolic compounds (1 and 2). Among these compounds, 2-methoxystypandrone (5) exhibited potent anti-proliferative effects on Jurkat cells. Analysis by flow cytometry confirmed that 2-methoxystypandrone (5) could significantly reduce mitochondrial membrane potential and promote increased levels of mitochondrial reactive oxygen species (ROS), suggesting a strong mitochondrial depolarization effect. Real-time quantitative polymerase chain reaction (qPCR) analysis was also performed, and the results revealed that the accumulation of ROS was caused by reduced mRNA expression levels of heme oxygenase (HO-1), catalase (CAT), glutathione peroxidase (GPx), and superoxide dismutase (SOD). In addition, 2-methoxystypandrone (5) triggered strong apoptosis that was mediated by the arrest of the G0/G1 phase of the cell cycle. Furthermore, 2-methoxystypandrone (5) downregulated p-IκB-α, p-NF-κB p65, Bcl2, and Bcl-xl and upregulated BAX proteins. Taken together, these findings revealed that 2-methoxystypandrone (5) isolated from RJH could potentially serve as an early lead compound for leukemia treatment involving intracellular signaling by increasing mitochondrial ROS and exerting anti-proliferative effects.
Collapse
Affiliation(s)
- Yinda Qiu
- College of Pharmacy, Chonnam National University, Gwangju 686, Republic of Korea
| | - Aoding Li
- College of Pharmacy, Chonnam National University, Gwangju 686, Republic of Korea
| | - Jina Lee
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Jeong Eun Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 4141, Republic of Korea,Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 3113, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 4141, Republic of Korea
| | - Namki Cho,
- College of Pharmacy, Chonnam National University, Gwangju 686, Republic of Korea,Corresponding authors N.Cho Phone: +82-62-530-2926 E-mail:
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea,H.M.Yoo Phone: 82-42-868-5362 E-mail:
| |
Collapse
|
9
|
Dobrucka R, Romaniuk-Drapała A, Kaczmarek M. Anti-Leukemia Activity of Au/CuO/ZnO Nanoparticles Synthesized used Verbena officinalis Extract. J Inorg Organomet Polym Mater 2020. [DOI: 10.1007/s10904-020-01690-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AbstractAs biological synthesis has become an alternative to chemical and physical methods for synthesizing nanoparticles, this work describes the synthesis of Au/CuO/ZnO nanoparticles using Verbena officinalis extract. The synthesized Au/CuO/ZnO nanoparticles were characterized using Ultraviolet–Visible, Fourier Transform-Infrared, Transmission Electron Microscopy and Atomic Force Microscopy. The influence of Au/CuO/ZnO nanoparticles on cell viability was evaluated in vitro, using the established cell line – Jurkat (ATCC® TIB-152™). The Annexin V binding test confirmed the previous results of the MTT assay, which indicate that the studied complex of Au/CuO/ZnO nanoparticles has a strong cytotoxic effect on the Jurkat cell line. The type of death and the effectiveness of cell elimination depended both on the concentration of the complex and the duration of culture.
Collapse
|
10
|
Tang H, Chen J, Wang L, Li Q, Yang Y, Lv Z, Bao H, Li Y, Luan X, Li Y, Ren Z, Zhou X, Cong D, Liu Z, Jia J, Chen H, Zhao W, Meng Q, Sun F, Pei J. Co-delivery of epirubicin and paclitaxel using an estrone-targeted PEGylated liposomal nanoparticle for breast cancer. Int J Pharm 2019; 573:118806. [PMID: 31678519 DOI: 10.1016/j.ijpharm.2019.118806] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/17/2022]
Abstract
Breast cancer is one of the most frequent malignancies in the female population. Recently, the development of medical products has been advanced for this disease; however, patients still suffer from the failure of current treatments and new therapeutic strategies are urgently required. In this study, due to the overexpression of the estrogen receptor (ER) in breast cancer and the ability of ER to specifically bind to its ligand estrone (ES), an ES-targeted PEGylated epirubicin (EPI) and paclitaxel (PTX) co-loaded liposomal nanoparticle (NP) (termed as ES-SSL-EPI/PTX) was developed. Physicochemical studies demonstrated that the ES-SSL-EPI/PTX had a nanoscaled particle size (~120 nm) and a neutral zeta potential (~-5 mV) and presented favorable stability in physiological media. In vitro, the ES-SSL-EPI/PTX showed a significantly higher cellular uptake in human breast cancer MCF-7 cells mainly via the receptor-ligand mediated pathway resulting in effective cytotoxic activity. In vivo targeting study, the accumulation of targeted liposomes in tumor was significantly improved. The systemic circulation time and biodistribution in main organs of EPI and PTX delivered by ES-SSL-Liposomes were increased. Consequently, the ES-SSL-EPI/PTX significantly suppressed tumor growth in the MCF-7-derived tumor-bearing mouse model without inducing toxicity. These results suggested that the ES-SSL-EPI/PTX was a promising formulation for co-delivery of chemotherapeutics in the treatment of breast cancer.
Collapse
Affiliation(s)
- Huan Tang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Jinglin Chen
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Lin Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Qianwen Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yue Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Department of Pharmacy, Ministry of Health Service, the Chinese PLA General Hospital, Beijing 100853, China
| | - Zhe Lv
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Han Bao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yao Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xue Luan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yan Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhihui Ren
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xiaowei Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Dengli Cong
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhiyi Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Juan Jia
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hongyu Chen
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Weitao Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Qin Meng
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Fei Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Jin Pei
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
11
|
Sun L, Sun C, Sun J, Yang W. Downregulation of ENDOCAN in myeloid leukemia cells inhibits proliferation and promotes apoptosis by suppressing nuclear factor‑κB activity. Mol Med Rep 2019; 19:3247-3254. [PMID: 30816462 DOI: 10.3892/mmr.2019.9969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 01/08/2019] [Indexed: 11/05/2022] Open
Abstract
Previous studies have demonstrated that ENDOCAN is elevated in leukemia, and it has been reported to be associated with poor prognosis. However, the functional role of ENDOCAN in the development of leukemia remains to be fully elucidated. In the present study, the expression levels of ENDOCAN were detected in THP‑1, U937, HL‑60 and K562 cells, and it was found that ENDOCAN was increased in U937 and K562 cells, compared with the other two cell lines. Subsequently, ENDOCAN was knocked down in U937 and K562 cells via lentiviral infection. It was found that cell proliferation and the expression of proliferating cell nuclear antigen were inhibited in myeloid leukemia cells following the silencing of ENDOCAN. ENDOCAN knockdown induced G0/G1‑phase cell cycle arrest in myeloid leukemia cells with a decreased expression of cyclin D1. Furthermore, cell apoptosis was increased in response to ENDOCAN silencing, which was accompanied by the downregulation of B‑cell lymphoma (BCL2) and the upregulation of BCL2‑associated X protein, cleaved caspases 3 and 9, and cleaved poly (ADP‑ribose) polymerase. Furthermore, it was demonstrated that the knockdown of ENDOCAN inhibited nuclear factor‑κB (NF‑κB) activity, as evidenced by the increased expression of NF‑κB inhibitor α (IκBα), decreased expression of phosphorylated (p‑)IκBα, p‑P65 and nuclear P65, and reduced NF‑κB DNA‑binding activity. In combination, the present findings suggested that ENDOCAN may serve as a potential therapeutic target in the treatment of leukemia.
Collapse
Affiliation(s)
- Lingling Sun
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Chengyu Sun
- Anorectal Department, Shenyang Anorectal Hospital, Shenyang, Liaoning 110054, P.R. China
| | - Jiaying Sun
- Intensive Care Unit, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Wei Yang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
12
|
Luan X, Rahme K, Cong Z, Wang L, Zou Y, He Y, Yang H, Holmes JD, O'Driscoll CM, Guo J. Anisamide-targeted PEGylated gold nanoparticles designed to target prostate cancer mediate: Enhanced systemic exposure of siRNA, tumour growth suppression and a synergistic therapeutic response in combination with paclitaxel in mice. Eur J Pharm Biopharm 2019; 137:56-67. [PMID: 30779980 DOI: 10.1016/j.ejpb.2019.02.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/18/2018] [Accepted: 02/15/2019] [Indexed: 12/24/2022]
Abstract
Small interfering RNA (siRNA) has recently illustrated therapeutic potential for malignant disorders. However, the clinical application of siRNA-based therapeutics is significantly retarded by the paucity of successful delivery systems. Recently, multifunctional gold nanoparticles (AuNPs) as non-viral delivery carriers have shown promise for transporting chemotherapeutics, proteins/peptides, and genes. In this study, AuNPs capped with polyethylenimine (PEI) and PEGylated anisamide (a ligand known to target the sigma receptor) have been developed to produce a range of positively charged anisamide-targeted PEGylated AuNPs (namely Au-PEI-PEG-AA). The anisamide-targeted AuNPs effectively complexed siRNA via electrostatic interaction, and the resultant complex (Au110-PEI-PEG5000-AA.siRNA) illustrated favourable physicochemical characteristics, including particle size, surface charge, and stability. In vitro, anisamide-targeted AuNPs selectively bound to human prostate cancer PC-3 cells, inducing efficient endosomal escape of siRNA, and effective downregulation of the RelA gene. In vivo, prolonged systemic exposure of siRNA was achieved by anisamide-targeted AuNPs resulting in significant tumour growth suppression in a PC3 xenograft mouse model without an increase in toxicity. In addition, a combination of siRNA-mediated NF-κB knockdown using anisamide-targeted AuNPs with Paclitaxel produced a synergistic therapeutic response, thus providing a promising therapeutic strategy for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xue Luan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Kamil Rahme
- Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Zouk Mosbeh, Lebanon; Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
| | - Zhongcheng Cong
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Limei Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Department of Pharmacy, The General Hospital of FAW, Changchun 130011, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yan He
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hao Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Justin D Holmes
- Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER@CRANN, Trinity College Dublin, Dublin 2, Ireland
| | | | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
13
|
Lettnin AP, Wagner EF, Carrett-Dias M, Dos Santos Machado K, Werhli A, Cañedo AD, Trindade GS, de Souza Votto AP. Silencing the OCT4-PG1 pseudogene reduces OCT-4 protein levels and changes characteristics of the multidrug resistance phenotype in chronic myeloid leukemia. Mol Biol Rep 2019; 46:1873-1884. [PMID: 30721421 DOI: 10.1007/s11033-019-04639-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022]
Abstract
Cancer stem cells show epigenetic plasticity and intrinsic resistance to anti-cancer therapy, rendering capable of initiating cancer relapse and progression. Transcription factor OCT-4 regulates various pathways in stem cells, but its expression can be regulated by pseudogenes. This work evaluated how OCT4-PG1 pseudogene can affect OCT-4 expression and mechanisms related to the multidrug resistance (MDR) phenotype in FEPS cells. Considering that OCT-4 protein is a transcription factor that regulates expression of ABC transporters, level of gene expression, activity of ABC proteins and cell sensitivity to chemotherapy were evaluated after OCT4-PG1 silencing. Besides we set up a STRING network. Results showed that after OCT4-PG1 silencing, cells expressed OCT-4 gene and protein to a lesser extent than mock cells. The gene and protein expression of ABCB1, as well as its activity were reduced. On the other hand, ALOX5 and ABCC1 genes was increased even as the activity of this transporter. Moreover, the silencing cells become sensitive to two chemotherapics tested. The network structure demonstrated that OCT4-PG1 protein interacts directly with OCT-4, SOX2, and NANOG and indirectly with ABC transporters. We conclude that OCT4-PG1 pseudogene plays a key role in the regulation OCT-4 transcription factor, which alters MDR phenotype in the FEPS cell line.
Collapse
Affiliation(s)
- Aline Portantiolo Lettnin
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil.,Laboratory of Cell Culture, Institute of Biological Sciences - ICB, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Eduardo Felipe Wagner
- Laboratory of Cell Culture, Institute of Biological Sciences - ICB, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Michele Carrett-Dias
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Karina Dos Santos Machado
- Center of Computational Sciences - C3, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Adriano Werhli
- Center of Computational Sciences - C3, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Andrés Delgado Cañedo
- Federal University of Pampa - UNIPAMPA, Avenue Antônio Trilha, 1847, São Gabriel, RS, Zip Code 97300-000, Brazil
| | - Gilma Santos Trindade
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil
| | - Ana Paula de Souza Votto
- Post-Graduate Program in Physiological Sciences - PPGCF, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil. .,Laboratory of Cell Culture, Institute of Biological Sciences - ICB, Federal University of Rio Grande -FURG, Avenue Itália, Km 8, Rio Grande, RS, Zip Code 96203-900, Brazil. .,Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Avenida Itália, Km 8, s/n, Rio Grande, RS, Zip Code 96203-900, Brazil.
| |
Collapse
|
14
|
Lu J, Zhang Y, Wang S, Bi Y, Huang T, Luo X, Cai YD. Analysis of Four Types of Leukemia Using Gene Ontology Term and Kyoto Encyclopedia of Genes and Genomes Pathway Enrichment Scores. Comb Chem High Throughput Screen 2019; 23:295-303. [PMID: 30599106 DOI: 10.2174/1386207322666181231151900] [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] [Received: 06/28/2018] [Revised: 09/24/2018] [Accepted: 12/05/2018] [Indexed: 12/16/2022]
Abstract
AIM AND OBJECTIVE Leukemia is the second common blood cancer after lymphoma, and its incidence rate has an increasing trend in recent years. Leukemia can be classified into four types: acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myelogenous leukemia (CML). More than forty drugs are applicable to different types of leukemia based on the discrepant pathogenesis. Therefore, the identification of specific drug-targeted biological processes and pathways is helpful to determinate the underlying pathogenesis among such four types of leukemia. METHODS In this study, the gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways that were highly related to drugs for leukemia were investigated for the first time. The enrichment scores for associated GO terms and KEGG pathways were calculated to evaluate the drugs and leukemia. The feature selection method, minimum redundancy maximum relevance (mRMR), was used to analyze and identify important GO terms and KEGG pathways. RESULTS Twenty Go terms and two KEGG pathways with high scores have all been confirmed to effectively distinguish four types of leukemia. CONCLUSION This analysis may provide a useful tool for the discrepant pathogenesis and drug design of different types of leukemia.
Collapse
Affiliation(s)
- Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 32 Qingquan Road, Yantai 264005, China
| | - YuHang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - ShaoPeng Wang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 32 Qingquan Road, Yantai 264005, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiaomin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of MateriaMedica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| |
Collapse
|
15
|
Abstract
Here we describe a simple way to create a gold nanoparticle (AuNP)-based non-viral delivery system to deliver siRNA into prostate cancer cells. Therefore, positively charged polyethylenimine (PEI)-capped AuNPs were synthesized in water and further conjugated with the targeting ligand (folic acid) for folate receptors (AuNPs-PEI-FA). The AuNPs-PEI-FA could effectively complex small interfering RNA (siRNA) through electrostatic interaction. Flow cytometry displayed that AuNPs-PEI-FA could specifically deliver siRNA into LNCaP cells, a prostate cancer cell line overexpressing prostate-specific membrane antigen (PSMA) that exhibits a hydrolase enzymatic activity with a folate substrate. In contrast, internalization of siRNA into PC-3 cells, a prostate cancer cell line not expressing PSMA or folate receptors, was not achieved using AuNPs-PEI-FA.siRNA. Following endolysosomal escape, the AuNPs-PEI-FA-.siRNA formulation resulted in significant endogenous gene silencing when compared to the nontargeted formulation, suggesting the potential of AuNPs-PEI-FA for targeted delivery of therapeutic siRNAs in the treatment of prostate cancer.
Collapse
|
16
|
Sun P, Huang W, Kang L, Jin M, Fan B, Jin H, Wang QM, Gao Z. siRNA-loaded poly(histidine-arginine) 6-modified chitosan nanoparticle with enhanced cell-penetrating and endosomal escape capacities for suppressing breast tumor metastasis. Int J Nanomedicine 2017; 12:3221-3234. [PMID: 28458542 PMCID: PMC5402910 DOI: 10.2147/ijn.s129436] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
An ideal carrier that delivers small interfering RNA (siRNA) should be designed based on two criteria: cellular-mediated internalization and endosomal escape. Poly(histidine-arginine)6(H6R6) peptide was introduced into chitosan (CS) to create a new CS derivative for siRNA delivery, 6-polyarginine (R6) as cell-penetrating peptides facilitated nanoparticle cellular internalization has been proved in our previous research, and 6-polyhistidine (H6) mediated the nanoparticle endosome escape resulted in the siRNA rapid releasing into tumor cytoplasm. H6R6-modified CS nanoparticles showed higher transfection efficiency and better endosomal escape capacity compared to ungroomed CS nanoparticle in vitro. Noticeably, H6R6-modified CS nanoparticles effectively inhibited tumor cell growth and metastases in vivo and significantly improved survival ratio. Therefore, we concluded that H6R6-modified CS copolymer can act as an ideal carrier for siRNA delivery and as a promising candidate in breast cancer therapy.
Collapse
Affiliation(s)
- Ping Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Lin Kang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Bo Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Hongyan Jin
- Yanbian University Hospital, Jilin, People's Republic of China
| | - Qi-Ming Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| |
Collapse
|
17
|
Guo J, Russell EG, Darcy R, Cotter TG, McKenna SL, Cahill MR, O’Driscoll CM. Antibody-Targeted Cyclodextrin-Based Nanoparticles for siRNA Delivery in the Treatment of Acute Myeloid Leukemia: Physicochemical Characteristics, in Vitro Mechanistic Studies, and ex Vivo Patient Derived Therapeutic Efficacy. Mol Pharm 2017; 14:940-952. [DOI: 10.1021/acs.molpharmaceut.6b01150] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jianfeng Guo
- School
of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
- Pharmacodelivery
Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Eileen G. Russell
- Tumour
Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Raphael Darcy
- Pharmacodelivery
Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Thomas G. Cotter
- Tumour
Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | | | - Mary R. Cahill
- Department
of Haematology, Cork University Hospital, Cork, Ireland
| | | |
Collapse
|
18
|
Recent advances in the design, development, and targeting mechanisms of polymeric micelles for delivery of siRNA in cancer therapy. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
19
|
Ding ZZ, Fan ZH, Huang XW, Bai SM, Song DW, Lu Q, Kaplan DL. Bioactive Natural Protein-Hydroxyapatite Nanocarriers for Optimizing Osteogenic Differentiation of Mesenchymal Stem Cells. J Mater Chem B 2016; 4:3555-3561. [PMID: 27482381 PMCID: PMC4959278 DOI: 10.1039/c6tb00509h] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Improving the controlled release of bioactive growth factors to regulate cell behavior and tissue regeneration remains a need in tissue engineering and regenerative medicine. Inorganic and polymeric nanoparticles have been extensively fabricated as bioactive biomaterials with enhanced biocompatibility and effective carriers of therapeutic agents, however, challenges remain such as the achievement of high loading capacity and sustained release, and the bioactivity preservation of growth factors. Here, a multilayered, silk coated hydroxyapatite (HA) nanocarrier with drug loading-release capacity superior to pure silk or HA nanoparticles was developed. Bone morphogenetic protein-2 (BMP-2) was bound to the silk coatings with a high binding efficiency of 99.6%, significantly higher than that in silk or the HA nanoparticles alone. The release of BMP-2 was sustained in vitro over a period of 21 days without burst release. Compared with BMP-2 loaded silk or HA particles, bone mesenchymal stem cells (BMSCs) showed improved proliferation and osteogenesis when cultured with the BMP-2 loaded composite nanocarriers. Therefore, these silk-HA composite nanoparticles present a useful approach to designing bioactive nanocarrier systems with enhanced functions for bone tissue regeneration needs.
Collapse
Affiliation(s)
- Z. Z. Ding
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, People’s Republic of China
| | - Z. H. Fan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215000, People’s Republic of China
| | - X. W. Huang
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - S. M. Bai
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - D. W. Song
- Tai’an City Central Hospital, Taian 271000, People’s Republic of China Address
| | - Q. Lu
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
| | - D. L. Kaplan
- National Engineering Laboratory for Modern Silk and Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People’s Republic of China
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, United States
| |
Collapse
|
20
|
Platelet-derived growth factor receptor/platelet-derived growth factor (PDGFR/PDGF) system is a prognostic and treatment response biomarker with multifarious therapeutic targets in cancers. Tumour Biol 2016; 37:10053-66. [PMID: 27193823 DOI: 10.1007/s13277-016-5069-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/05/2016] [Indexed: 02/06/2023] Open
Abstract
Progress in cancer biology has led to an increasing discovery of oncogenic alterations of the platelet-derived growth factor receptors (PDGFRs) in cancers. In addition, their overexpression in numerous cancers invariably makes PDGFRs and platelet-derived growth factors (PDGFs) prognostic and treatment markers in some cancers. The oncologic alterations of the PDGFR/PDGF system affect the extracellular, transmembrane and tyrosine kinase domains as well as the juxtamembrane segment of the receptor. The receptor is also involved in fusions with intracellular proteins and receptor tyrosine kinase. These discoveries undoubtedly make the system an attractive oncologic therapeutic target. This review covers elementary biology of PDGFR/PDGF system and its role as a prognostic and treatment marker in cancers. In addition, the multifarious therapeutic targets of PDGFR/PDGF system are discussed. Great potential exists in the role of PDGFR/PDGF system as a prognostic and treatment marker and for further exploration of its multifarious therapeutic targets in safe and efficacious management of cancer treatments.
Collapse
|
21
|
Guo J, O'Driscoll CM, Holmes JD, Rahme K. Bioconjugated gold nanoparticles enhance cellular uptake: A proof of concept study for siRNA delivery in prostate cancer cells. Int J Pharm 2016; 509:16-27. [PMID: 27188645 DOI: 10.1016/j.ijpharm.2016.05.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/19/2022]
Abstract
The chemistry of gold nanoparticles (AuNPs) facilitates surface modifications and thus these bioengineered NPs have been investigated as a means of delivering a variety of therapeutic cargos to treat cancer. In this study we have developed AuNPs conjugated with targeting ligands to enhance cell-specific uptake in prostate cancer cells, with a purpose of providing efficient non-viral gene delivery systems in the treatment of prostate cancer. As a consequence, two novel AuNPs were synthesised namely AuNPs-PEG-Tf (negatively charged AuNPs with the transferrin targeting ligands) and AuNPs-PEI-FA (positively charged AuNPs with the folate-receptor targeting ligands). Both bioconjugated AuNPs demonstrated low cytotoxicity in prostate cancer cells. The attachment of the targeting ligand Tf to AuNPs successfully achieved receptor-mediated cellular uptake in PC-3 cells, a prostate cancer cell line highly expressing Tf receptors. The AuNPs-PEI-FA effectively complexed small interfering RNA (siRNA) through electrostatic interaction. At the cellular level the AuNPs-PEI-FA specifically delivered siRNA into LNCaP cells, a prostate cancer cell line overexpressing prostate specific membrane antigen (PSMA, exhibits a hydrolase enzymic activity with a folate substrate). Following endolysosomal escape the AuNPs-PEI-FA.siRNA formulation produced enhanced endogenous gene silencing compared to the non-targeted formulation. Our results suggest both formulations have potential as non-viral gene delivery vectors in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jianfeng Guo
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland.
| | | | - Justin D Holmes
- Materials Chemistry and Analysis Group, Department of Chemistry and The Tyndall National Institute, University College Cork, Cork, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin 2, Ireland
| | - Kamil Rahme
- Materials Chemistry and Analysis Group, Department of Chemistry and The Tyndall National Institute, University College Cork, Cork, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin 2, Ireland; Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Zouk Mosbeh, Lebanon.
| |
Collapse
|
22
|
Xiang J, Zhang Z, Mu Y, Xu X, Guo S, Liu Y, Russo DP, Zhu H, Yan B, Bai X. Discovery of Novel Tricyclic Thiazepine Derivatives as Anti-Drug-Resistant Cancer Agents by Combining Diversity-Oriented Synthesis and Converging Screening Approach. ACS COMBINATORIAL SCIENCE 2016; 18:230-5. [PMID: 27082930 DOI: 10.1021/acscombsci.6b00010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
An efficient discovery strategy by combining diversity-oriented synthesis and converging cellular screening is described. By a three-round screening process, we identified novel tricyclic pyrido[2,3-b][1,4]benzothiazepines showing potent inhibitory activity against paclitaxel-resistant cell line H460TaxR (EC50 < 1.0 μM), which exhibits much less toxicity toward normal cells (EC50 > 100 μM against normal human fibroblasts). The most active hits also exhibited drug-like properties suitable for further preclinical research. This redeployment of antidepressing compounds for anticancer applications provides promising future prospects for treating drug-resistant tumors with fewer side effects.
Collapse
Affiliation(s)
- Jinbao Xiang
- The
Center for Combinatorial Chemistry and Drug Discovery, The School
of Pharmaceutical Sciences, and The College of Chemistry, Jilin University, Changchun, Jilin 130021, P. R. China
| | - Zhuoqi Zhang
- The
Center for Combinatorial Chemistry and Drug Discovery, The School
of Pharmaceutical Sciences, and The College of Chemistry, Jilin University, Changchun, Jilin 130021, P. R. China
| | - Yan Mu
- School
of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Xianxiu Xu
- The
Center for Combinatorial Chemistry and Drug Discovery, The School
of Pharmaceutical Sciences, and The College of Chemistry, Jilin University, Changchun, Jilin 130021, P. R. China
| | - Sigen Guo
- The
Center for Combinatorial Chemistry and Drug Discovery, The School
of Pharmaceutical Sciences, and The College of Chemistry, Jilin University, Changchun, Jilin 130021, P. R. China
| | - Yongjin Liu
- School
of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Daniel P. Russo
- The Rutgers Center for Computational and Integrative Biology, Camden, New Jersey 08102, United States
| | - Hao Zhu
- The Rutgers Center for Computational and Integrative Biology, Camden, New Jersey 08102, United States
- Department
of Chemistry, Rutgers University, Camden, New Jersey 08102, United States
| | - Bing Yan
- School
of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Xu Bai
- The
Center for Combinatorial Chemistry and Drug Discovery, The School
of Pharmaceutical Sciences, and The College of Chemistry, Jilin University, Changchun, Jilin 130021, P. R. China
| |
Collapse
|
23
|
Wang Y, Appiah-Kubi K, Wu M, Yao X, Qian H, Wu Y, Chen Y. The platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) are major players in oncogenesis, drug resistance, and attractive oncologic targets in cancer. Growth Factors 2016; 34:64-71. [PMID: 27170215 DOI: 10.1080/08977194.2016.1180293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) play a key role in signaling pathways in oncogenesis. The overexpression of PDGFs and PDGFRs and the oncogenic alterations of these receptors have been implicated in human cancers and correlated significantly with poor outcomes. This review discusses the biology of the PDGF isoforms and receptors briefly, and their role in oncogenesis. Also, the attractiveness of targeting PDGFs and PDGFRs, based on a wide display of oncologic alterations in cancers, diverse therapeutic strategies, their roles in resistance to cancer treatments with prospects of overcoming drug resistance, and the extent to which validated biomarkers have been developed for effective PDGFs and PDGFRs-based cancer management are discussed.
Collapse
Affiliation(s)
- Ying Wang
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Kwaku Appiah-Kubi
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
- b Department of Applied Biology , University for Development Studies , Navrongo , Ghana , and
| | - Min Wu
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Xiaoyuan Yao
- c Basic Medical Department, Changchun Medical College , Jilin , People's Republic of China
| | - Hai Qian
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Yan Wu
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Yongchang Chen
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| |
Collapse
|
24
|
Abstract
Abstract
The past 70 years have seen childhood acute lymphoblastic leukemia move from a fatal disease with a survival of barely 4 months to a curable disease in >85% of patients. It has become clear that as treatment has intensified, more children are cured but at the expense of increased toxicity which for some can cause significant long-term morbidity and even mortality. The drive in more recent years has been to identify sensitive markers of disease and response to treatment to allow a reduction in therapy in those who do not require it and more intensive treatment in those who do. Clinical characteristics have been used to stratify patients into different risk groups and this, coupled with following response at a molecular level, has done much to tailor treatment to the patient. Considerable research has been focused on the molecular characteristics of the leukemia itself to elucidate the biologic mechanisms underlying both the disease and the comparative or absolute resistance of some types of leukemia. These molecular markers can also act as targets for novel therapies, which require newer trial methodologies to prove their utility. There has been less focus on the biology of the patient but it is clear that some patients are more susceptible to adverse events and toxicities than others. Through the use of pharmacogenomics, modification to therapy may be appropriate in certain patients based on their genetic profile. As novel therapies become available, suitable controlled trials in children are essential for their safe use in this population and will ensure that children are not denied timely access to advances in treatment.
Collapse
|
25
|
7-formyl-10-methylisoellipticine, a novel ellipticine derivative, induces mitochondrial reactive oxygen species (ROS) and shows anti-leukaemic activity in mice. Invest New Drugs 2015; 34:15-23. [DOI: 10.1007/s10637-015-0302-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/01/2015] [Indexed: 01/05/2023]
|
26
|
Rahme K, Guo J, Holmes JD, O'Driscoll CM. Evaluation of the physicochemical properties and the biocompatibility of polyethylene glycol-conjugated gold nanoparticles: A formulation strategy for siRNA delivery. Colloids Surf B Biointerfaces 2015; 135:604-612. [PMID: 26322474 DOI: 10.1016/j.colsurfb.2015.08.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The potential of RNA interference (RNAi)-based therapeutics for cancer has received much attention; however, delivery of RNAi effectors, such as small interfering RNA (siRNA), remains an obstacle to clinical translation. Non-viral delivery vectors have been used extensively to enhance siRNA delivery. Recently, the potential of gold nanoparticles (AuNPs) for transporting drugs, proteins and genetic materials has been demonstrated. Previously, our laboratory synthesised positively charged, surfactant-free AuNPs in water by the reduction of gold (III) chloride (AuCl3) using hydroxylamine hydrochloride (NH2OH·HCl) in the presence of L-cysteine methyl ester hydrochloride (HSCH2CH(NH2)COOCH3·HCl) as a capping agent. These AuNPs, which achieve higher cell viability in comparison to cetyl trimethyl ammonium bromide (CTAB, a surfactant)-capped counterparts, have demonstrated potential for siRNA delivery. However, it is well known that systemic administration of cationic delivery systems without biological stablising moieties causes non-specific binding with negatively charged serum proteins, resulting in particle aggregation and opsonisation. Consequently, highly stable AuNPs capped with l-cysteine methyl ester hydrochloride conjugated to poly(ethylene glycol) (PEG) were synthesised in this study. PEGylation enhanced the biocompatibility of the AuNPs by reducing toxicity in a range of cell types, by inhibiting interaction with serum proteins thus avoiding aggregation, and, by providing protection against degradation by nucleases. Moreover, these PEGylated AuNPs formed nanoparticles (NPs) with siRNA (which was first compacted with protamine), and had a diameter within the nanoscale range (∼ 250 nm) and a near neutral surface charge (∼ 10 mV). In the future a bifunctional PEG chain on the AuNPs (i.e., SH-PEG-NH2, SH-PEG-COOH) will be used to facilitate conjugation of a targeting ligand to enhance cell specific uptake.
Collapse
Affiliation(s)
- Kamil Rahme
- Materials Chemistry and Analysis Group, Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER (Advanced Materials and Biological Engineering Research Centre), CRANN (Centre for Research on Adaptive Nanostructures and Nanodevices), Trinity College Dublin, Dublin, Ireland; Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Lebanon
| | - Jianfeng Guo
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Ireland
| | - Justin D Holmes
- Materials Chemistry and Analysis Group, Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER (Advanced Materials and Biological Engineering Research Centre), CRANN (Centre for Research on Adaptive Nanostructures and Nanodevices), Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
27
|
Guo J, McKenna SL, O’Dwyer ME, Cahill MR, O’Driscoll CM. RNA interference for multiple myeloma therapy: targeting signal transduction pathways. Expert Opin Ther Targets 2015; 20:107-21. [DOI: 10.1517/14728222.2015.1071355] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
28
|
Cho SK, Dang C, Wang X, Ragan R, Kwon YJ. Mixing-sequence-dependent nucleic acid complexation and gene transfer efficiency by polyethylenimine. Biomater Sci 2015. [PMID: 26221945 DOI: 10.1039/c5bm00041f] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polyplexes, complexed nucleic acids by cationic polymers, are the most common forms of nonviral gene delivery vectors. In contrast to a great deal of efforts in synthesizing novel cationic polymers and exploring their extracellular and intracellular delivery pathways, polyplex preparation methods of mixing nucleic acids and cationic polymers are often overlooked. In this study, the mixing sequence, that is adding nucleic acids to polymers or vice versa, was found to greatly affect complexation of both plasmid DNA and siRNA, polyplexes' size, and polyplexes' surface charge, which all collaboratively affected the transfection efficiency and cytotoxicity. Adding polyethylenimine (PEI), the most conventionally used standard in nonviral gene delivery, to plasmid DNA and siRNA resulted in larger polyplexes, higher gene expression and silencing, but higher cytotoxicity than polyplexes prepared in the reverse order. Based on the experimental results, the authors developed a model that gradual addition of cationic polymers (e.g., PEI) to nucleic acids (e.g., plasmid DNA and siRNA) incorporates more copies of nucleic acids in larger polyplexes in a smaller number, results in higher gene expression and silencing levels in transfected cells, and generates higher cytotoxicity by leaving more free polymers upon complete mixing than the other mixing sequence. The proposed model can be explored using a broad range of cationic polymers and nucleic acids, and provide insightful information about how to prepare polyplexed nonviral vectors for efficient and safe gene delivery.
Collapse
Affiliation(s)
- S K Cho
- Department of Chemistry, Dong-A University, Busan, South Korea
| | | | | | | | | |
Collapse
|