1
|
Zhang SS, Yu JH, Jiang SS, Wang L, Chen J, Long J, Gu SX, Li H. T7 peptide-mediated co-delivery platform overcoming multidrug-resistant breast cancer: In vitro and in vivo evaluation. Eur J Pharm Biopharm 2024; 200:114327. [PMID: 38759900 DOI: 10.1016/j.ejpb.2024.114327] [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: 12/14/2023] [Revised: 03/30/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
P-glycoprotein (P-gp) overexpressed mutidrug resistance (MDR) is currently a key factor limiting the effectiveness of breast cancer chemotherapy. Systemic administration based on P-gp-associated mechanism leads to severe toxic side effects. Here, we designed a T7 peptide-modified mixed liposome (T7-MLP@DTX/SchB) that, by active targeting co-delivering chemotherapeutic agents and P-gp inhibitors, harnessed synergistic effects to improve the treatment of MDR breast cancer. This study established drug-resistant cell models and animal models. Subsequently, comprehensive evaluations involving cell uptake, cell apoptosis, cellular toxicity assays, in vivo tumor-targeting capability, and anti-tumor activity assays were conducted to assess the drug resistance reversal effects of T7-MLP@DTX/SchB. Additionally, a systematic assessment of the biosafety profile of T7-MLP@DTX/SchB was executed, including blood profiles, biochemical markers, and histopathological examination. It was found that this co-delivery strategy successfully exerted the synergistic effects, since there was a significant tumor growth inhibitory effect on multidrug-resistant breast cancer. Targeted modification with T7 peptide enhanced the therapeutic efficacy remarkably, while vastly ameliorating the biocompatibility compared to free drugs. The intriguing results supported the promising potential use of T7-MLP@DTX/SchB in overcoming MDR breast cancer treatment.
Collapse
Affiliation(s)
- Shuang-Shuang Zhang
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, Hubei, China; Pharmaceutical Research Institute, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Jia-Hui Yu
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, Hubei, China
| | - Si-Si Jiang
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, Hubei, China
| | - Lun Wang
- Huazhong Pharmaceutical Company Limited, Xiangyang 441021, China
| | - Jiong Chen
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, Hubei, China
| | - Jiao Long
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, Hubei, China
| | - Shuang-Xi Gu
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, Hubei, China; Pharmaceutical Research Institute, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Hui Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
2
|
Zong R, Ruan H, Liu C, Fan S, Li J. Bacteria and Bacterial Components as Natural Bio-Nanocarriers for Drug and Gene Delivery Systems in Cancer Therapy. Pharmaceutics 2023; 15:2490. [PMID: 37896250 PMCID: PMC10610331 DOI: 10.3390/pharmaceutics15102490] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteria and bacterial components possess multifunctional properties, making them attractive natural bio-nanocarriers for cancer diagnosis and targeted treatment. The inherent tropic and motile nature of bacteria allows them to grow and colonize in hypoxic tumor microenvironments more readily than conventional therapeutic agents and other nanomedicines. However, concerns over biosafety, limited antitumor efficiency, and unclear tumor-targeting mechanisms have restricted the clinical translation and application of natural bio-nanocarriers based on bacteria and bacterial components. Fortunately, bacterial therapies combined with engineering strategies and nanotechnology may be able to reverse a number of challenges for bacterial/bacterial component-based cancer biotherapies. Meanwhile, the combined strategies tend to enhance the versatility of bionanoplasmic nanoplatforms to improve biosafety and inhibit tumorigenesis and metastasis. This review summarizes the advantages and challenges of bacteria and bacterial components in cancer therapy, outlines combinatorial strategies for nanocarriers and bacterial/bacterial components, and discusses their clinical applications.
Collapse
Affiliation(s)
| | | | | | - Shaohua Fan
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| | - Jun Li
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
3
|
Kesharwani P, Chadar R, Shukla R, Jain GK, Aggarwal G, Abourehab MAS, Sahebkar A. Recent advances in multifunctional dendrimer-based nanoprobes for breast cancer theranostics. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2433-2471. [PMID: 35848467 DOI: 10.1080/09205063.2022.2103627] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Breast cancer (BC) undoubtedly is one of the most common type of cancers amongst women, which causes about 5 million deaths annually. The treatments and diagnostic therapy choices currently available for Breast Cancer is very much limited . Advancements in novel nanocarrier could be a promising strategy for diagnosis and treatments of this deadly disease. Dendrimer nanoformulation could be functionalized and explored for efficient targeting of overexpressed receptors on Breast Cancer cells to achieve targeted drug delivery, for diagnostics and to overcome the resistance of the cells towards particular chemotherapeutic. Additionally, the dendrimer have shown promising potential in the improvement of therapeutic value for Breast Cancer therapy by achieving synergistic co-delivery of chemotherapeutics and genetic materials for multidirectional treatment. In this review, we have highlighted the application of dendrimer as novel multifunctional nanoplatforms for the treatment and diagnosis of Breast Cancer.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India
| | - Gaurav K Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Yang C, Lin ZI, Chen JA, Xu Z, Gu J, Law WC, Yang JHC, Chen CK. Organic/Inorganic Self-Assembled Hybrid Nano-Architectures for Cancer Therapy Applications. Macromol Biosci 2021; 22:e2100349. [PMID: 34735739 DOI: 10.1002/mabi.202100349] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/25/2021] [Indexed: 12/20/2022]
Abstract
Since the conceptualization of nanomedicine, numerous nanostructure-mediated drug formulations have progressed into clinical trials for treating cancer. However, recent clinical trial results indicate such kind of drug formulations has a limited improvement on the antitumor efficacy. This is due to the biological barriers associated with those formulations, for example, circulation stability, extravasation efficiency in tumor, tumor penetration ability, and developed multi-drug resistance. When employing for nanomedicine formulations, pristine organic-based and inorganic-based nanostructures have their own limitations. Accordingly, organic/inorganic (O/I) hybrids have been developed to integrate the merits of both, and to minimize their intrinsic drawbacks. In this context, the recent development in O/I hybrids resulting from a self-assembly strategy will be introduced. Through such a strategy, organic and inorganic building blocks can be self-assembled via either chemical covalent bonds or physical interactions. Based on the self-assemble procedure, the hybridization of four organic building blocks including liposomes, micelles, dendrimers, and polymeric nanocapsules with five functional inorganic nanoparticles comprising gold nanostructures, magnetic nanoparticles, carbon-based materials, quantum dots, and silica nanoparticles will be highlighted. The recent progress of these O/I hybrids in advanced modalities for combating cancer, such as, therapeutic agent delivery, photothermal therapy, photodynamic therapy, and immunotherapy will be systematically reviewed.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Jian-An Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jiayu Gu
- Department of Pharmacy, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020, China
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Jason Hsiao Chun Yang
- Department of Fiber and Composite Materials, Feng Chia University, Taichung, 40724, Taiwan
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| |
Collapse
|
5
|
Surekha B, Kommana NS, Dubey SK, Kumar AP, Shukla R, Kesharwani P. PAMAM dendrimer as a talented multifunctional biomimetic nanocarrier for cancer diagnosis and therapy. Colloids Surf B Biointerfaces 2021; 204:111837. [DOI: 10.1016/j.colsurfb.2021.111837] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022]
|
6
|
Tarach P, Janaszewska A. Recent Advances in Preclinical Research Using PAMAM Dendrimers for Cancer Gene Therapy. Int J Mol Sci 2021; 22:2912. [PMID: 33805602 PMCID: PMC7999260 DOI: 10.3390/ijms22062912] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022] Open
Abstract
Carriers of genetic material are divided into vectors of viral and non-viral origin. Viral carriers are already successfully used in experimental gene therapies, but despite advantages such as their high transfection efficiency and the wide knowledge of their practical potential, the remaining disadvantages, namely, their low capacity and complex manufacturing process, based on biological systems, are major limitations prior to their broad implementation in the clinical setting. The application of non-viral carriers in gene therapy is one of the available approaches. Poly(amidoamine) (PAMAM) dendrimers are repetitively branched, three-dimensional molecules, made of amide and amine subunits, possessing unique physiochemical properties. Surface and internal modifications improve their physicochemical properties, enabling the increase in cellular specificity and transfection efficiency and a reduction in cytotoxicity toward healthy cells. During the last 10 years of research on PAMAM dendrimers, three modification strategies have commonly been used: (1) surface modification with functional groups; (2) hybrid vector formation; (3) creation of supramolecular self-assemblies. This review describes and summarizes recent studies exploring the development of PAMAM dendrimers in anticancer gene therapies, evaluating the advantages and disadvantages of the modification approaches and the nanomedicine regulatory issues preventing their translation into the clinical setting, and highlighting important areas for further development and possible steps that seem promising in terms of development of PAMAM as a carrier of genetic material.
Collapse
MESH Headings
- Biocompatible Materials/administration & dosage
- Biocompatible Materials/chemical synthesis
- Dendrimers/administration & dosage
- Dendrimers/chemical synthesis
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Genetic Therapy/methods
- Government Regulation
- Humans
- MicroRNAs/administration & dosage
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Nanomedicine/legislation & jurisprudence
- Nanomedicine/methods
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Neoplasms/therapy
- Oligonucleotides, Antisense/administration & dosage
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/metabolism
- Plasmids/administration & dosage
- Plasmids/chemistry
- Plasmids/metabolism
- RNA, Messenger/administration & dosage
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Surface Properties
Collapse
Affiliation(s)
- Piotr Tarach
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | | |
Collapse
|
7
|
Padayachee J, Daniels A, Balgobind A, Ariatti M, Singh M. HER-2/neu and MYC gene silencing in breast cancer: therapeutic potential and advancement in nonviral nanocarrier systems. Nanomedicine (Lond) 2020; 15:1437-1452. [PMID: 32515263 DOI: 10.2217/nnm-2019-0459] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Globally, breast cancer is the second leading cause of cancer-related mortality among women, with approximately 1.4 million new cases diagnosed annually. Associated genetic perturbations are emerging in the face of intense scientific enquiry, facilitating its classification, prognostication and treatment. RNAi, utilizing siRNA, is a powerful treatment strategy to silence disease-causing genes. However, therapeutic siRNA instability and poor cellular uptake have limited its clinical application, necessitating the use of nanocarriers. In this review, we highlight the RNAi mechanism, HER-2/neu and MYC as breast cancer gene targets, and nonviral nanocarriers as potentially safe and efficient delivery systems.
Collapse
Affiliation(s)
- Jananee Padayachee
- Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| | - Aliscia Daniels
- Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| | - Adhika Balgobind
- Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| | - Mario Ariatti
- Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| | - Moganavelli Singh
- Nano-Gene & Drug Delivery Laboratory, Discipline of Biochemistry, School of Life Sciences, College of Agriculture, Engineering & Science, University of KwaZulu-Natal (Westville Campus), Private Bag X54001, Durban 4000, Kwa-Zulu Natal, South Africa
| |
Collapse
|
8
|
Choudhury H, Pandey M, Yin TH, Kaur T, Jia GW, Tan SQL, Weijie H, Yang EKS, Keat CG, Bhattamishra SK, Kesharwani P, Md S, Molugulu N, Pichika MR, Gorain B. Rising horizon in circumventing multidrug resistance in chemotherapy with nanotechnology. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:596-613. [PMID: 31029353 DOI: 10.1016/j.msec.2019.04.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/24/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is one of the key barriers in chemotherapy, leading to the generation of insensitive cancer cells towards administered therapy. Genetic and epigenetic alterations of the cells are the consequences of MDR, resulted in drug resistivity, which reflects in impaired delivery of cytotoxic agents to the cancer site. Nanotechnology-based nanocarriers have shown immense shreds of evidence in overcoming these problems, where these promising tools handle desired dosage load of hydrophobic chemotherapeutics to facilitate designing of safe, controlled and effective delivery to specifically at tumor microenvironment. Therefore, encapsulating drugs within the nano-architecture have shown to enhance solubility, bioavailability, drug targeting, where co-administered P-gp inhibitors have additionally combat against developed MDR. Moreover, recent advancement in the stimuli-sensitive delivery of nanocarriers facilitates a tumor-targeted release of the chemotherapeutics to reduce the associated toxicities of chemotherapeutic agents in normal cells. The present article is focused on MDR development strategies in the cancer cell and different nanocarrier-based approaches in circumventing this hurdle to establish an effective therapy against deadliest cancer disease.
Collapse
Affiliation(s)
- Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000, Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia.
| | - Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000, Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Tan Hui Yin
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Taasjir Kaur
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Gan Wei Jia
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - S Q Lawrence Tan
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - How Weijie
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Eric Koh Sze Yang
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Chin Guan Keat
- Bachelor of Pharmacy student, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Subrat Kumar Bhattamishra
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nagasekhara Molugulu
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, 57000, Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mallikarjuna Rao Pichika
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia; Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor 47500, Malaysia.
| |
Collapse
|
9
|
Herma R, Wrobel D, Liegertová M, Müllerová M, Strašák T, Maly M, Semerádtová A, Štofik M, Appelhans D, Maly J. Carbosilane dendrimers with phosphonium terminal groups are low toxic non-viral transfection vectors for siRNA cell delivery. Int J Pharm 2019; 562:51-65. [PMID: 30877030 DOI: 10.1016/j.ijpharm.2019.03.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 02/07/2023]
Abstract
Non-viral gene delivery vectors studied in the gene therapy applications are often designed with the cationic nitrogen containing groups necessary for binding and cell release of nucleic acids. Disadvantage is a relatively high toxicity which restricts the in vivo use of such nanoparticles. Here we show, that the 3rd generation carbosilane dendrimers possessing (trimethyl)phosphonium (PMe3) groups on their periphery were able to effectively deliver the functional siRNA into the cells (B14, Cricetulus griseus), release it into the cytosol and finally to achieve up to 40% gene silencing of targeted gene (glyceraldehyde-3-phosphate dehydrogenase (GAPDH)) with the comparable or, in some cases, even better effectivity as their ammonium counterparts. Moreover, such cationic dendrimers show relatively low in vivo toxicity as compared to their ammonium analogues when analyzed by standard fish embryo test (FET) on Danio rerio in vivo model, with LD50 = 6.26 µM after 48 h of incubation. This is more than 10-fold improvement as compared to published values for various other types of cationic dendrimers. We discuss the potential of further increase of the transfection efficiency, endosomal escape and decrease of toxicity of such non-viral vectors, based on the systematic screening of different types of substituents on central phosphonium atom.
Collapse
Affiliation(s)
- Regina Herma
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic
| | - Dominika Wrobel
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic
| | - Michaela Liegertová
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic
| | - Monika Müllerová
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic; Institute of Chemical Process Fundamentals of the CAS, v.v.i., Prague, Czech Republic
| | - Tomáš Strašák
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic; Institute of Chemical Process Fundamentals of the CAS, v.v.i., Prague, Czech Republic
| | - Marek Maly
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic
| | - Alena Semerádtová
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic
| | - Marcel Štofik
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic
| | - Dietmar Appelhans
- Leibniz Institute of Polymer Research Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Jan Maly
- Faculty of Science, J.E. Purkyně University in Ústí nad Labem, 40096 Ústí nad Labem, Czech Republic.
| |
Collapse
|
10
|
Dong M, Chen J, Zhang J, Liang X, Yang J, Li D, Li Q. A chemoenzymatically synthesized cholesterol-g-poly(amine-co-ester)-mediated p53 gene delivery for achieving antitumor efficacy in prostate cancer. Int J Nanomedicine 2019; 14:1149-1161. [PMID: 30863051 PMCID: PMC6391147 DOI: 10.2147/ijn.s191905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND An amphiphilic cationic copolymer cholesterol-g-poly(amine-co-ester), namely Chol-g-PMSC-PPDL synthesized in a chemoenzymatic route has been utilized as a carrier for p53 gene delivery to check its antitumor efficacy, using human prostate cancer cell line PC-3 (p53 null) as a model. MATERIALS AND METHODS The transfection efficiency was measured by quantitative PCR and Western blotting assay. The anti-proliferative effect was detected using MTT method, colony formation assay and Live/Dead staining. The anti-migration effect was evaluated through wound healing and Transwell migration assays. RESULTS The transfection efficiency assay indicated that the carrier-mediated p53 gene transfection could dramatically enhance the intracellular p53 expression level. Through p53 gene delivery, obvious anti-proliferative effect could be detected which was elucidated to be associated with the simultaneous activation of mitochondrial-dependent apoptosis pathway and cell cycle arrest at G1 phase. Meanwhile, the anti-migration effect could be obtained after p53 gene transfection. CONCLUSION Chol-g-PMSC-PPDL-mediated p53 gene transfection could potentially be employed as a promising strategy for achieving effective anti-tumor response.
Collapse
Affiliation(s)
- Mengmeng Dong
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, People's Republic of China,
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China,
| | - Jiawen Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China,
| | - Jiayuan Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China,
| | - Xiao Liang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China,
| | - Jiebing Yang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China,
| | - Dan Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun 130021, People's Republic of China,
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China,
| |
Collapse
|
11
|
Zhou Q, Song C, Liu X, Qin H, Miao L, Zhang X. Peptidylarginine deiminase 4 overexpression resensitizes MCF-7/ADR breast cancer cells to adriamycin via GSK3β/p53 activation. Cancer Manag Res 2019; 11:625-636. [PMID: 30666159 PMCID: PMC6331075 DOI: 10.2147/cmar.s191353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Adriamycin (ADR) is widely used in the clinical chemotherapy against breast cancer. But its efficacy is strongly limited due to the acquisition of multidrug resistance (MDR). Therefore, acquisition of the resistance to ADR is still a major cause of chemotherapy failure in breast cancer patients. Peptidylarginine deiminase IV (PAD4) is reported to target non-histone proteins for citrullination, regulate their substrate activities, and thereby play critical roles in maintaining cell phenotype in breast cancer cells. However, whether PAD4 is involved in the development of MDR in breast cancer is poorly understood. Materials and methods We examined the expression of PAD family members, including PAD4 in ADR-resistant MCF-7 cells compared with the parental control cells by real-time PCR and Western blotting analyses. Rescue of PAD4 expression in MCF-7/ADR cells was performed to assess whether PAD4 could restore the sensitivity of MCF-7/ADR cells to ADR treatment with cell counting kit-8, flow cytometry, TUNEL, nuclear and cytoplasmic extract preparations, and immunofluorescence staining analyses. Results Both PAD2 and PAD4 were significantly decreased in ADR-resistant cells. However, only PAD4 overexpression can increase the sensitivity of MCF-7/ADR cells to ADR treatment and decrease MDR1 gene expression. Overexpression of PAD4 in MCF-7/ADR cells inhibited cell proliferation by inducing cell apoptosis. Under ADR treatment, overexpression of PAD4 promoted nuclear accumulation of glycogen synthase kinase-3β and p53, which further activated proapoptotic gene expression and downregulated MDR1 expression. Moreover, PAD4 activity was required for activating proapoptotic gene transcripts. Conclusion We demonstrate the previously unappreciated role of PAD4 in reversing ADR resistance in MCF-7/ADR cells and help establish PAD4 as a candidate biomarker of prognosis and chemotherapy target for MDR in breast cancers.
Collapse
Affiliation(s)
- Qianqian Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China,
| | - Chao Song
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China,
| | - Xiaoqiu Liu
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, China.,Department of Microbiology, Nanjing Medical University, Nanjing, China
| | - Hao Qin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China,
| | - Lixia Miao
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China,
| | - Xuesen Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China,
| |
Collapse
|
12
|
Hu L, Li M, Zhang Z, Shen Y, Guo S. Self-assembly of biotinylated poly(ethylene glycol)-poly(curcumin) for paclitaxel delivery. Int J Pharm 2018; 553:510-521. [DOI: 10.1016/j.ijpharm.2018.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/26/2018] [Accepted: 10/07/2018] [Indexed: 01/07/2023]
|
13
|
Tian W, Hao S, Gao B, Jiang Y, Zhang X, Zhang S, Guo L, Zhao J, Zhang G, Chen Y, Li Z, Luo D. Lobaplatin inhibits breast cancer progression, cell proliferation while it induces cell apoptosis by downregulating MTDH expression. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3563-3571. [PMID: 30464390 PMCID: PMC6211578 DOI: 10.2147/dddt.s163157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective Lobaplatin shows antitumor activity against a wide range of tumors, including metastatic breast cancer (BCa). The overexpression of metadherin (MTDH) is associated with poor prognosis of BCa patients. This study was designed to investigate the effect of lobaplatin on MCF-7 cell proliferation and its association with MTDH expression. Patients and methods Clinical treatment for BCa using lobaplatin, in combination with other general chemotherapy drugs, was administered to 32 BCa patients. The safety, effectiveness, and prognosis in lobaplatin-treated BCa patients were compared with those in controls (n=32). In vitro experiments were performed in MCF-7 cells to investigate the effect of lobaplatin on cell proliferation, apoptosis, and MTDH expression. Results We found the intraoperative local chemotherapy using lobaplatin was safe and effective for BCa treatment, in comparison with the patients administered general chemotherapy drugs. Treatment of MCF-7 cell cultures with lobaplatin significantly reduced cell proliferation and increased cell apoptotic percentage. The expression of MTDH and Bcl-2 was inhibited by lobaplatin and that of Bax was increased by lobaplatin. Moreover, we observed the inhibition of MTDH by shRNA reduced cell proliferation and enhanced cell apoptosis. Conclusion Lobaplatin was a safe and effective adjuvant chemotherapy for BCa. The effect of lobaplatin on inhibiting MCF-7 cell proliferation and inducing cell apoptosis might be, as least in part, mediated by suppressing the expression of oncogene MTDH.
Collapse
Affiliation(s)
- Wuguo Tian
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Shuai Hao
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Bo Gao
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Yan Jiang
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Xiaohua Zhang
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Shu Zhang
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Lingji Guo
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Jianjie Zhao
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Gang Zhang
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Yi Chen
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Zhirong Li
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| | - Donglin Luo
- Department of Breast, Thyroid Surgery, Research Institute of Surgery, Daping Hospital, Army Military Medical University, Chongqing 400042, China,
| |
Collapse
|
14
|
Meng F, Zou L, Zhang T, Jiang L, Ding Y, Yu P, Peng J. Using LC-MS/MS-based targeted proteomics to monitor the pattern of ABC transporters expression in the development of drug resistance. Cancer Manag Res 2018; 10:2859-2870. [PMID: 30197538 PMCID: PMC6112789 DOI: 10.2147/cmar.s164766] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose The overexpression of ATP-binding cassette transporters (ABC transporters), mainly including permeability glycoproteins (P-gp), multidrug resistance (MDR)-related protein 1 (MRP1), and breast cancer resistance proteins (BCRP), is one of the main reasons for the development of MDR which directly leads to chemotherapy failure. However, most of the currently used detection methods in MDR-related studies are qualitative or semiquantitative, but not quantitative. As a result, the measurement criteria of different experiments are not unified. Moreover, there are many contradictory results of the studies of the induction effect of drugs on ABC transporters. So, it is necessary to establish a quantitative assay for the quantification of P-gp, MRP1, and BCRP to study the mechanism of drug resistance. Methods In this paper, a novel and advanced liquid chromatography/mass spectrometry (MS)/MS-based targeted proteomics method for the quantification of P-gp, MRP1, and BCRP was developed and validated. Then, the cell lines MCF-7, HepG-2, and SMMC-7721 were, respectively, induced by different concentrations of doxorubicin (adriamycin [ADM]), mitoxantrone (MX), and methotrexate (MTX), to establish resistance cell lines. The method established was used to quantify the expression of P-gp, MRP1, and BCRP. Results The result showed that the induction effects of drugs on protein were relatively stable and selective. ADM, MX, and MTX could induce overexpression of P-gp, MRP1, and BCRP. And, the induction effect of different drugs on proteins was selective. The pattern of overexpression of ABC transporters in the three types of resistance cell lines was different. Conclusion During the development of drug resistance, the cell type and patch, but not drug type, were the most important determinant factors of the overexpression level of ABC transporters in resistance cell lines. This study provides a good foundation for understanding the development of drug resistance in cell lines and can be used to explain the contradictory results in other published studies as described above.
Collapse
Affiliation(s)
- Fanqi Meng
- Department of Drug Analysis, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, China,
| | - Le Zou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Tengyu Zhang
- Department of Pharmacy, University of Copenhagen, København Ø, Denmark
| | - Lei Jiang
- Department of Drug Analysis, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, China,
| | - Yao Ding
- Department of Analyses and Testing, Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410013, Hunan Province, China
| | - Peng Yu
- Department of Drug Analysis, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, China,
| | - Jie Peng
- Department of Pharmacy, Jiangxi Provincial People's Hospital, Nanchang 330006, Jiangxi Province, China,
| |
Collapse
|
15
|
Kalaydina RV, Bajwa K, Qorri B, Decarlo A, Szewczuk MR. Recent advances in "smart" delivery systems for extended drug release in cancer therapy. Int J Nanomedicine 2018; 13:4727-4745. [PMID: 30154657 PMCID: PMC6108334 DOI: 10.2147/ijn.s168053] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advances in nanomedicine have become indispensable for targeted drug delivery, early detection, and increasingly personalized approaches to cancer treatment. Nanoparticle-based drug-delivery systems have overcome some of the limitations associated with traditional cancer-therapy administration, such as reduced drug solubility, chemoresistance, systemic toxicity, narrow therapeutic indices, and poor oral bioavailability. Advances in the field of nanomedicine include “smart” drug delivery, or multiple levels of targeting, and extended-release drug-delivery systems that provide additional methods of overcoming these limitations. More recently, the idea of combining smart drug delivery with extended-release has emerged in hopes of developing highly efficient nanoparticles with improved delivery, bioavailability, and safety profiles. Although functionalized and extended-release drug-delivery systems have been studied extensively, there remain gaps in the literature concerning their application in cancer treatment. We aim to provide an overview of smart and extended-release drug-delivery systems for the delivery of cancer therapies, as well as to introduce innovative advancements in nanoparticle design incorporating these principles. With the growing need for increasingly personalized medicine in cancer treatment, smart extended-release nanoparticles have the potential to enhance chemotherapy delivery, patient adherence, and treatment outcomes in cancer patients.
Collapse
Affiliation(s)
| | - Komal Bajwa
- Postgraduate Medical Education, Graduate Diploma and Professional Master in Medical Sciences, School of Medicine, Queen's University
| | - Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen's University,
| | | | - Myron R Szewczuk
- Department of Biomedical and Molecular Sciences, Queen's University,
| |
Collapse
|
16
|
Xu PY, Kankala RK, Pan YJ, Yuan H, Wang SB, Chen AZ. Overcoming multidrug resistance through inhalable siRNA nanoparticles-decorated porous microparticles based on supercritical fluid technology. Int J Nanomedicine 2018; 13:4685-4698. [PMID: 30154654 PMCID: PMC6103603 DOI: 10.2147/ijn.s169399] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In recent times, the co-delivery therapeutics have garnered enormous interest from researchers in the treatment of cancers with multidrug resistance (MDR) due to their efficient delivery of multiple agents, which result in synergistic effects and capable of overcoming all the obstacles of MDR in cancer. However, an efficient delivery platform is required for the conveyance of diverse agents that can successfully devastate MDR in cancer. METHODS Initially, short-interfering RNA-loaded chitosan (siRNA-CS) nanoparticles were synthesized using the ionic gelation method. Further, the siRNA-CS nanoparticles and doxorubicin hydrochloride (DOX) were co-loaded in poly-L-lactide porous microparticles (PLLA PMs) (nano-embedded porous microparticles, [NEPMs]) by the supercritical anti-solvent (SAS) process. RESULTS AND DISCUSSION The NEPM formulation exhibited an excellent aerodynamic performance and sustained release of DOX, which displayed higher anticancer efficacy in drug-resistant cells (human small cell lung cancer, H69AR cell line) than those treated with either free DOX and DOX-PLLA PMs due to the siRNA from CS nanoparticles silenced the MDR gene to DOX therapy. CONCLUSION This eco-friendly process provides a convenient way to fabricate such innovative NEPMs co-loaded with a chemotherapeutic agent and a gene, which can devastate MDR in cancer through the co-delivery system.
Collapse
Affiliation(s)
- Pei-Yao Xu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China,
| | - Ranjith Kumar Kankala
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China,
| | - Yu-Jing Pan
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China,
| | - Hui Yuan
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China,
| | - Shi-Bin Wang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China,
| | - Ai-Zheng Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China,
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China,
| |
Collapse
|
17
|
Haghiralsadat F, Amoabediny G, Naderinezhad S, Zandieh-Doulabi B, Forouzanfar T, Helder MN. Codelivery of doxorubicin and JIP1 siRNA with novel EphA2-targeted PEGylated cationic nanoliposomes to overcome osteosarcoma multidrug resistance. Int J Nanomedicine 2018; 13:3853-3866. [PMID: 30013340 PMCID: PMC6039070 DOI: 10.2147/ijn.s150017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose Osteosarcoma (OS) mostly affects children and young adults, and has only a 20%–30% 5-year survival rate when metastasized. We aimed to create dual-targeted (extracellular against EphA2 and intracellular against JNK-interacting protein 1 [JIP1]), doxorubicin (DOX)-loaded liposomes to treat OS metastatic disease. Materials and methods Cationic liposomes contained N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate (DOTAP), cholesterol, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), and distearoyl-phosphatidylethanolamine–methyl-poly(ethylene glycol) (DSPE–mPEG) conjugate. EphA2 targeting was accomplished by conjugating YSA peptide to DSPE–mPEG. Vesicles were subsequently loaded with DOX and JIP1 siRNA. Results Characteristics assessment showed that 1) size of the bilayered particles was 109 nm; 2) DOX loading efficiency was 87%; 3) siRNA could be successfully loaded at a liposome:siRNA ratio of >24:1; and 4) the zeta potential was 18.47 mV. Tumor-mimicking pH conditions exhibited 80% siRNA and 50.7% DOX sustained release from the particles. Stability studies ensured the protection of siRNA against degradation in serum. OS cell lines showed increased and more pericellular/nuclear localizations when using targeted vesicles. Nontargeted and targeted codelivery caused 70.5% and 78.6% cytotoxicity in OS cells, respectively (free DOX: 50%). Targeted codelivery resulted in 42% reduction in the siRNA target, JIP1 mRNA, and 46% decrease in JIP1 levels. Conclusion Our dual-targeted, DOX-loaded liposomes enhance toxicity toward OS cells and may be effective for the treatment of metastatic OS.
Collapse
Affiliation(s)
- Fateme Haghiralsadat
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.,Department of Orthopaedic Surgery, VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, the Netherlands.,Department of Nano Biotechnology, Research Center for New Technologies in Life Science Engineering,
| | - Ghasem Amoabediny
- Department of Nano Biotechnology, Research Center for New Technologies in Life Science Engineering, .,Department of Biotechnology and Pharmaceutical Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran, .,Department of Oral and Maxillofacial Surgery, VU University Medical Center, MOVE Research Institute Amsterdam,
| | - Samira Naderinezhad
- Department of Biotechnology and Pharmaceutical Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran,
| | - Behrouz Zandieh-Doulabi
- Department of Oral Cell Biology and Functional Anatomy, Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam and University of Amsterdam, MOVE Research Institute, Amsterdam, the Netherlands
| | - Tymour Forouzanfar
- Department of Oral and Maxillofacial Surgery, VU University Medical Center, MOVE Research Institute Amsterdam,
| | - Marco N Helder
- Department of Oral and Maxillofacial Surgery, VU University Medical Center, MOVE Research Institute Amsterdam,
| |
Collapse
|
18
|
Li J, Liang H, Liu J, Wang Z. Poly (amidoamine) (PAMAM) dendrimer mediated delivery of drug and pDNA/siRNA for cancer therapy. Int J Pharm 2018; 546:215-225. [PMID: 29787895 DOI: 10.1016/j.ijpharm.2018.05.045] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022]
Abstract
Poly (amidoamine) (PAMAM) dendrimers are well-defined, highly branched macromolecules with numerous active amine groups on the surface. Because of their unique properties, PAMAM dendrimers have steadily grown in popularity in drug delivery, gene therapy, medical imaging and diagnostic application. This review focuses on the recent developments on the application in PAMAM dendrimers as effective carriers for drug and gene (pDNA, siRNA) delivery in cancer therapy, including: a) PAMAM for anticancer drug delivery; b) PAMAM and gene therapy; c) PAMAM used in overcoming tumor multidrug resistance; d) PAMAM used for hybrid nanoparticles; and e) PAMAM linked or loaded in other nanoparticles.
Collapse
Affiliation(s)
- Jun Li
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| | - Huamin Liang
- Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230088, Anhui, China
| | - Jing Liu
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing 100084, China
| | - Ziyuan Wang
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
19
|
Liu L, Yang J, Men K, He Z, Luo M, Qian Z, Wei X, Wei Y. Current Status of Nonviral Vectors for Gene Therapy in China. Hum Gene Ther 2018; 29:110-120. [PMID: 29320893 DOI: 10.1089/hum.2017.226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Li Liu
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Jingyun Yang
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Ke Men
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Zhiyao He
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Min Luo
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Zhiyong Qian
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Xiawei Wei
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| | - Yuquan Wei
- Laboratory for Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, P.R. China
| |
Collapse
|
20
|
Ayoub NM, Al-Shami KM, Alqudah MA, Mhaidat NM. Crizotinib, a MET inhibitor, inhibits growth, migration, and invasion of breast cancer cells in vitro and synergizes with chemotherapeutic agents. Onco Targets Ther 2017; 10:4869-4883. [PMID: 29042798 PMCID: PMC5634371 DOI: 10.2147/ott.s148604] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
MET is a receptor tyrosine kinase known for its pleiotropic effects in tumorigenesis. Dysregulations of MET expression and/or signaling have been reported and determined to be associated with inferior outcomes in breast cancer patients rendering MET a versatile candidate for targeted therapeutic intervention. Crizotinib is a multi-targeted small-molecule kinase inhibitor for MET, ALK, and ROS1 kinases. This study evaluated the anti-proliferative, cytotoxic, anti-migratory, and anti-invasive effects of crizotinib in breast cancer cells in vitro. Cell viability was assessed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) colorimetric assay. In vitro wound-healing assay was used to examine the effect of crizotinib on breast cancer cell migration. The expressions of Ki-67, MET, and phospho-MET receptors were characterized using immunofluorescence staining. Results showed that crizotinib has significant anti-proliferative activity on all mammary tumor cells with IC50 values of 5.16, 1.5, and 3.85 µM in MDA-MB-231, MCF-7, and SK-BR-3 cells, respectively. Crizotinib induced cytotoxic effects in all breast cancer cells examined. Combined treatment of small dose of crizotinib with paclitaxel or doxorubicin exhibited a highly synergistic inhibition of growth of MDA-MB-231 and MCF-7 cells with combination index values <1 while no significant effect was observed in SK-BR-3 cells compared with individual compounds. Treatment with crizotinib demonstrated a remarkable reduction in the expression of Ki-67 protein in all 3 tested cell lines. Crizotinib inhibited migration and invasion of MDA-MB-231 cells in a dose-dependent fashion. Crizotinib reduced MET receptor activation in MDA-MB-231 cells when treated at effective concentrations. In conclusion, crizotinib suppressed proliferation, migration, and invasion of breast cancer cells in vitro. The results of this study demonstrated that combined treatment of crizotinib with chemotherapeutic agents resulted in a synergistic growth inhibition of specific breast cancer cell lines.
Collapse
Affiliation(s)
- Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Kamal M Al-Shami
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad A Alqudah
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Nizar M Mhaidat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
21
|
Chen X, Zhang Y, Tang C, Tian C, Sun Q, Su Z, Xue L, Yin Y, Ju C, Zhang C. Co-delivery of paclitaxel and anti-survivin siRNA via redox-sensitive oligopeptide liposomes for the synergistic treatment of breast cancer and metastasis. Int J Pharm 2017. [PMID: 28642204 DOI: 10.1016/j.ijpharm.2017.06.071] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The overexpression of survivin in breast cancer cells is an important factor of paclitaxel (PTX) resistance in breast cancer. To overcome PTX resistance and improve the antitumor effect of PTX, we developed a novel liposome-based nanosystem (PTX/siRNA/SS-L), composed of a redox-sensitive cationic oligopeptide lipid (LHSSG2C14) with a proton sponge effect, natural soybean phosphatidylcholine (SPC), and cholesterol for co-delivery of PTX and anti-survivin siRNA, which could specifically downregulate survivin overexpression. PTX/siRNA/SS-L exhibited high encapsulation efficiency and rapid redox-responsive release of both PTX and siRNA. Moreover, in vitro studies on the 4T1 breast cancer cells revealed that PTX/siRNA/SS-L offered significant advantages over other experimental groups, such as higher cellular uptake, successful endolysosomal escape, reduced survivin expression, the lowest cell viability and wound healing rate, as well as the highest apoptosis rate. In particular, in vivo evaluation of 4T1 tumor-bearing mice showed that PTX/siRNA/SS-L had lower toxicity and induced a synergistic inhibitory effect on tumor growth and pulmonary metastasis. Collectively, the collaboration of anti-survivin siRNA and PTX via redox-sensitive oligopeptide liposomes provides a promising strategy for the treatment of breast cancer and metastasis.
Collapse
Affiliation(s)
- Xinyan Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China; Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yidi Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Chunming Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Chunli Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Qiong Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Yin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, China.
| |
Collapse
|
22
|
Novel PEI/Poly-γ-Gutamic Acid Nanoparticles for High Efficient siRNA and Plasmid DNA Co-Delivery. Molecules 2017; 22:molecules22010086. [PMID: 28054985 PMCID: PMC6155750 DOI: 10.3390/molecules22010086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/21/2016] [Accepted: 12/29/2016] [Indexed: 11/25/2022] Open
Abstract
The efficient delivery of sufficient amounts of nucleic acids into target cells is critical for successful gene therapy and gene knockdown. The DNA/siRNA co-delivery system has been considered a promising approach for cancer therapy to simultaneously express and inhibit tumor suppressor genes and overexpressed oncogenes, respectively, triggering synergistic anti-cancer effects. Polyethylenimine (PEI) has been identified as an efficient non-viral vector for transgene expression. In this study, we created a very high efficient DNA/siRNA co-delivery system by incorporating a negatively-charged poly-γ-glutamic acid (γ-PGA) into PEI/nucleic acid complexes. Spherical nanoparticles with about 200 nm diameter were formed by mixing PEI/plasmid DNA/siRNA/γ-PGA (dual delivery nanoparticles; DDNPs) with specific ratio (N/P/C ratio) and the particles present positive surface charge under all manufacturing conditions. The gel retardation assay shows both nucleic acids were effectively condensed by PEI, even at low N/P ratios. The PEI-based DDNPs reveal excellent DNA/siRNA transfection efficiency in the human hepatoma cell line (Hep 3B) by simultaneously providing high transgene expression efficiency and high siRNA silencing effect. The results indicated that DDNP can be an effective tool for gene therapy against hepatoma.
Collapse
|
23
|
Yin M, Bao Y, Gao X, Wu Y, Sun Y, Zhao X, Xu H, Zhang Z, Tan S. Redox/pH dual-sensitive hybrid micelles for targeting delivery and overcoming multidrug resistance of cancer. J Mater Chem B 2017; 5:2964-2978. [DOI: 10.1039/c6tb03282f] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A three-step approach was used to enhance anticancer activity via targeted delivery, intracellular drug burst release, and depressed drug efflux.
Collapse
Affiliation(s)
- Mingxing Yin
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
| | - Yuling Bao
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
- Department of Pharmacy
| | - Xueqin Gao
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
| | - Yupei Wu
- Department of Pharmacy
- Hebei General Hospital
- Shijiazhuang 050051
- China
| | - Yu Sun
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
| | - Xinyuan Zhao
- Department of Pharmacy
- Tongji Hospital
- Tongji Medical School
- Huazhong University of Science and Technology
- Wuhan 430030
| | - Hongzhe Xu
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
| | - Zhiping Zhang
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
- National Engineering Research Centre for Nanomedicine
| | - Songwei Tan
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan 430030
- China
- National Engineering Research Centre for Nanomedicine
| |
Collapse
|