1
|
Paolino D, d'Avanzo N, Canato E, Ciriolo L, Grigoletto A, Cristiano MC, Mancuso A, Celia C, Pasut G, Fresta M. Improved anti-breast cancer activity by doxorubicin-loaded super stealth liposomes. Biomater Sci 2024; 12:3933-3946. [PMID: 38940612 DOI: 10.1039/d4bm00478g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
PEGylation is currently used for the synthesis of stealth liposomes and to enhance the pharmacokinetic and biopharmaceutical properties of payloads. PEGylated dendron phospholipids can decrease the detachment of polyethylene glycol (PEG) from the liposomal surface owing to an increased hydrophobic anchoring effect on the phospholipid bilayer of liposomes and thus generating super stealth liposomes that are suitable for the systemic delivery of anticancer drugs. Herein, doxorubicin hydrochloride-loaded super stealth liposomes were studied for the treatment of breast cancer lung metastasis in an animal model. The results demonstrated that the super stealth liposomes had suitable physicochemical properties for in vivo administration and could significantly increase the efficacy of doxorubicin in breast cancer lung metastasis tumor-bearing mice compared to the free drug. The super stealth liposomes also increased doxorubicin accumulation inside the tumor tissue. The permanence of PEG on the surface of the super stealth liposomes favored the formation of a depot of therapeutic nanocarriers inside the tumor tissue by improving their permanence after stopping treatment. The doxorubicin-loaded super stealth liposomes increased the survival of the mouse tumor model. These promising results demonstrate that the doxorubicin-loaded super stealth liposomes could be an effective nanomedicine to treat metastatic breast cancer.
Collapse
Affiliation(s)
- Donatella Paolino
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
- Research Center "ProHealth Translational Hub", Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-88100 Catanzaro, Italy
| | - Nicola d'Avanzo
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
- Research Center "ProHealth Translational Hub", Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-88100 Catanzaro, Italy
| | - Elena Canato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, I-35131 Padua, Italy.
| | - Luigi Ciriolo
- Department of Health Science, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, I-35131 Padua, Italy.
| | - Maria Chiara Cristiano
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-Catanzaro, Italy
| | - Antonia Mancuso
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
- Research Center "ProHealth Translational Hub", Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Campus Universitario "S. Venuta"-Building of BioSciences, Viale S. Venuta, I-88100 Catanzaro, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100, Chieti, Italy.
- Lithuanian University of Health Sciences, Laboratory of Drug Targets Histopathology, Institute of Cardiology, A. Mickeviciaus g. 9, LT-44307 Kaunas, Lithuania
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, I-35131 Padua, Italy.
| | - Massimo Fresta
- Department of Health Science, University of Catanzaro "Magna Græcia", V.le "S. Venuta", Catanzaro, I-88100, Italy
| |
Collapse
|
2
|
Wang J, Zhang W, Xie Z, Wang X, Sun J, Ran F, Jiang W, Liu Y, Wang Z, Ran H, Guo D. NIR-responsive copper nanoliposome composites for cascaded ferrotherapy via ferroptosis actived ICD and IFN-γ released. Biomaterials 2024; 308:122570. [PMID: 38636133 DOI: 10.1016/j.biomaterials.2024.122570] [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: 12/17/2023] [Revised: 03/14/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
Metallic biomaterials activate tumor ferroptosis by increasing oxidative stress, but their efficacy is severely limited in tumor microenvironment. Although interferon gamma (IFN-γ) can promote tumor ferroptosis sensitivity by inhibiting the antioxidant system and promoting lipid accumulation, this effect limited by the lack of IFN-γ accumulation in tumors. Herein, we report a near-infrared (NIR)-responsive HCuS nanocomposite (HCuS-PE@TSL-tlyp-1) that can stimulate immunogenic cell death (ICD)-mediated IFN-γ secretion through exogenous oxidative stress, thereby achieving cascaded ferrotherapy by mutually reinforcing ferroptosis and systemic immunity. Upon laser irradiation, the dissolution of the thermal coating, and the introduction of Cu ions and piperazine-erastin (PE) simultaneously induce oxidative stress by reactive oxygen species (ROS)/lipid peroxide (LPO) accumulation and deplete cystine-glutamate transporter (xCT)/GSH. The onset of oxidative stress-mediated ferroptosis is thus achieved, and ICD is triggered, significantly promoting cytotoxic T-cell (CTL) infiltration for IFN-γ secretion. Furthermore, IFN-γ induces immunogenic tumor ferroptosis by inhibiting xCT-antioxidant pathways and enhancing the ACSL4-fatty acid recruitment pathway, which further promotes sensitivity to ferroptosis in cells. These HCuS nanocomposites combined with aPD-L1 effectively in inhibiting tumor metastasis and recurrence. Importantly, these cascade ferrotherapy results broadens the application of HCuS biomaterials.
Collapse
Affiliation(s)
- Junrui Wang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Wenli Zhang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Zhuoyan Xie
- Department of Ultrasound, Chongqing General Hospital, Chongqing, 400014, PR China
| | - Xingyue Wang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science. Xiangyang, Hubei, 441053, PR China
| | - Jiangshan Sun
- Chongqing Medical and Health School, Chongqing, 408000, PR China
| | - Fei Ran
- Department of Dentistry, Chongqing University Fuling Hospital, Chongqing, 408000, PR China
| | - Weixi Jiang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
3
|
Liu H, Wen Z, Liu Z, Yang Y, Wang H, Xia X, Ye J, Liu Y. Unlocking the potential of amorphous calcium carbonate: A star ascending in the realm of biomedical application. Acta Pharm Sin B 2024; 14:602-622. [PMID: 38322345 PMCID: PMC10840486 DOI: 10.1016/j.apsb.2023.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 02/08/2024] Open
Abstract
Calcium-based biomaterials have been intensively studied in the field of drug delivery owing to their excellent biocompatibility and biodegradability. Calcium-based materials can also deliver contrast agents, which can enhance real-time imaging and exert a Ca2+-interfering therapeutic effect. Based on these characteristics, amorphous calcium carbonate (ACC), as a brunch of calcium-based biomaterials, has the potential to become a widely used biomaterial. Highly functional ACC can be either discovered in natural organisms or obtained by chemical synthesis However, the standalone presence of ACC is unstable in vivo. Additives are required to be used as stabilizers or core-shell structures formed by permeable layers or lipids with modified molecules constructed to maintain the stability of ACC until the ACC carrier reaches its destination. ACC has high chemical instability and can produce biocompatible products when exposed to an acidic condition in vivo, such as Ca2+ with an immune-regulating ability and CO2 with an imaging-enhancing ability. Owing to these characteristics, ACC has been studied for self-sacrificing templates of carrier construction, targeted delivery of oncology drugs, immunomodulation, tumor imaging, tissue engineering, and calcium supplementation. Emphasis in this paper has been placed on the origin, structural features, and multiple applications of ACC. Meanwhile, ACC faces many challenges in clinical translation, and long-term basic research is required to overcome these challenges. We hope that this study will contribute to future innovative research on ACC.
Collapse
Affiliation(s)
- Han Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhiyang Wen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zihan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xuejun Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
4
|
Cardoso BD, Fernandes DEM, Amorim CO, Amaral VS, Coutinho PJG, Rodrigues ARO, Castanheira EMS. Magnetoliposomes with Calcium-Doped Magnesium Ferrites Anchored in the Lipid Surface for Enhanced DOX Release. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2597. [PMID: 37764626 PMCID: PMC10535675 DOI: 10.3390/nano13182597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Nanotechnology has provided a new insight into cancer treatment by enabling the development of nanocarriers for the encapsulation, transport, and controlled release of antitumor drugs at the target site. Among these nanocarriers, magnetic nanosystems have gained prominence. This work presents the design, development, and characterization of magnetoliposomes (MLs), wherein superparamagnetic nanoparticles are coupled to the lipid surface. For this purpose, dimercaptosuccinic acid (DMSA)-functionalized Ca0.25Mg0.75Fe2O4 superparamagnetic nanoparticles were prepared for the first time. The magnetic nanoparticles demonstrated a cubic shape with an average size of 13.36 nm. Furthermore, their potential for photothermal hyperthermia was evaluated using 4 mg/mL, 2 mg/mL, and 1 mg/mL concentrations of NPs@DMSA, which demonstrated a maximum temperature variation of 20.4 °C, 11.4 °C, and 7.3 °C, respectively, during a 30 min NIR-laser irradiation. Subsequently, these nanoparticles were coupled to the lipid surface of DPPC/DSPC/CHEMS and DPPC/DSPC/CHEMS/DSPE-PEG-based MLs using a new synthesis methodology, exhibiting average sizes of 153 ± 8 nm and 136 ± 2 nm, respectively. Doxorubicin (DOX) was encapsulated with high efficiency, achieving 96% ± 2% encapsulation in non-PEGylated MLs and 98.0% ± 0.6% in stealth MLs. Finally, drug release assays of the DOX-loaded DPPC/DSPC/CHEMS MLs were performed under different conditions of temperature (37 °C and 42 °C) and pH (5.5 and 7.4), simulating physiological and therapeutic conditions. The results revealed a higher release rate at 42 °C and acidic pH. Release rates significantly increased when introducing the stimulus of laser-induced photothermal hyperthermia at 808 nm (1 W/cm2) for 5 min. After 48 h of testing, at pH 5.5, 67.5% ± 0.5% of DOX was released, while at pH 7.4, only a modest release of 27.0% ± 0.1% was achieved. The results demonstrate the potential of the MLs developed in this work to the controlled release of DOX under NIR-laser stimulation and acidic environments and to maintain a sustained and reduced release profile in physiological environments with pH 7.4.
Collapse
Affiliation(s)
- Beatriz D. Cardoso
- Physics Centre of Minho and Porto Universities (CF-UM-UP), Campus de Gualtar, 4710-057 Braga, Portugal (D.E.M.F.)
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, Universidade do Minho, 4710-057 Braga, Portugal
- CMEMS—UMinho, Universidade do Minho, DEI, 4800-058 Guimarães, Portugal
- LABBELS—Associate Laboratory, 4800-058 Guimarães, Portugal
| | - Diana E. M. Fernandes
- Physics Centre of Minho and Porto Universities (CF-UM-UP), Campus de Gualtar, 4710-057 Braga, Portugal (D.E.M.F.)
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, Universidade do Minho, 4710-057 Braga, Portugal
| | - Carlos O. Amorim
- Physics Department and CICECO, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Vítor S. Amaral
- Physics Department and CICECO, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Paulo J. G. Coutinho
- Physics Centre of Minho and Porto Universities (CF-UM-UP), Campus de Gualtar, 4710-057 Braga, Portugal (D.E.M.F.)
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, Universidade do Minho, 4710-057 Braga, Portugal
| | - Ana Rita O. Rodrigues
- Physics Centre of Minho and Porto Universities (CF-UM-UP), Campus de Gualtar, 4710-057 Braga, Portugal (D.E.M.F.)
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, Universidade do Minho, 4710-057 Braga, Portugal
| | - Elisabete M. S. Castanheira
- Physics Centre of Minho and Porto Universities (CF-UM-UP), Campus de Gualtar, 4710-057 Braga, Portugal (D.E.M.F.)
- LaPMET—Laboratory of Physics for Materials and Emergent Technologies, Universidade do Minho, 4710-057 Braga, Portugal
| |
Collapse
|
5
|
Pardhi E, Yadav R, Chaurasiya A, Madan J, Guru SK, Singh SB, Mehra NK. Multifunctional targetable liposomal drug delivery system in the management of leukemia: Potential, opportunities, and emerging strategies. Life Sci 2023; 325:121771. [PMID: 37182551 DOI: 10.1016/j.lfs.2023.121771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 05/16/2023]
Abstract
The concern impeding the success of chemotherapy in leukemia treatment is descending efficacy of drugs because of multiple drug resistance (MDR). The previous failure of traditional treatment methods is primarily responsible for the present era of innovative agents to treat leukemia effectively. The treatment option is a chemotherapeutic agent in most available treatment strategies, which unfortunately leads to high unavoidable toxicities. As a result of the recent surge in marketed products, theranostic nanoparticles, i.e., multifunctional targetable liposomes (MFTL), have been approved for improved and more successful leukemia treatment that blends therapeutic and diagnostic characteristics. Since they broadly offer the required characteristics to get past the traditional/previous limitations, such as the absence of site-specific anti-cancer therapeutic delivery and ongoing real-time surveillance of the leukemia target sites while administering therapeutic activities. To prepare MFTL, suitable targeting ligands or tumor-specific antibodies are required to attach to the surface of the liposomes. This review exhaustively covered and summarized the liposomal-based formulation in leukemia treatment, emphasizing leukemia types; regulatory considerations, patents, and clinical portfolios to overcome clinical translation hurdles have all been explored.
Collapse
Affiliation(s)
- Ekta Pardhi
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Rati Yadav
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Akash Chaurasiya
- Department of Pharmaceutics, BITS-Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, District. RR, Hyderabad, India
| | - Jitender Madan
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India.
| |
Collapse
|
6
|
Soosani Z, Rezaei B, Heydari-Bafrooei E, Ensafi AA. Chemical Sensors Based on Molecularly Imprinted Polymers Can Determine Drug Release Kinetics from Nanocarriers without Filtration, Centrifugation, and Dialysis Steps. ACS Sens 2023; 8:1891-1900. [PMID: 36877535 DOI: 10.1021/acssensors.2c02436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
With the development of drug delivery systems, the use of nanomaterials for slow, targeted, and effective drug release has grown significantly. To ensure the quality of performance, it is essential to obtain drug release profiles from therapeutic nanoparticles prior to in vivo testing. Typically, the methods of monitoring the drug release profile from nanoparticle drug delivery systems include one or more filtration, separation, and sampling steps, with or without membrane, which cause several systematic errors and make the process time-consuming. Here, the release rate of doxorubicin as a model drug from liposome as a nanocarrier was determined via highly selective binding of released doxorubicin to the doxorubicin-imprinted electropolymerized polypyrrole as a molecularly imprinted polymer (MIP). Incubation of the MIP-modified substrate with imprinted cavities complementary to doxorubicin molecules in the releasing medium leads to the binding of released doxorubicin molecules to cavities. The drug trapped in the cavities is determined by one of the analytical methods depending on its signaling properties. In this work, due to the favorable electrochemical properties of doxorubicin, the voltammetry method was used for quantitative analysis of released doxorubicin. The voltammetric oxidation peak current intensity of doxorubicin on the surface of the electrode was enhanced by increasing the release time. This membranelle platform allows fast, reliable, and simple monitoring of drug release profiles without any sample preparation, filtration, and centrifugation in buffer and blood serum samples.
Collapse
Affiliation(s)
- Zeynab Soosani
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Behzad Rezaei
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | | | - Ali A Ensafi
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Iran.,Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
7
|
Chen Q, Guo C, Liu Z, Cao M, Wang W, Zhang D, Geng H, Diao N, Chen D. Multifunctional nanoparticles with anti-inflammatory effect for improving metabolic syndromes. J Drug Target 2023; 31:286-295. [PMID: 36315421 DOI: 10.1080/1061186x.2022.2142595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metabolic syndromes are a group of metabolic disorders for which the molecular mechanisms are still unclear. An increasing number of studies have implicated metabolic syndrome in the association with inflammation. Currently, lipsomes is known to improve nanoparticle hydrophobicity. Meanwhile, in drug delivery systems the application of cholesterol, which is commonly used to stabilise liposomal structures, has essentially no pharmacological effect on liposomes. Herein, we developed an 'anti-inflammatory liposome' (Phy-Lip) to effectively handle these challenges via employing Phytosterol instead of cholesterol. Different with the conventional liposomes, Phy-Lip is a much more brilliant nanoparticle with anti-inflammatory functions. In Phy-Lip, cholesterol was substituted by Phy, which works as membrane stabiliser, anti-inflammatory adjuvant at the same time. The experimental results show that Phy-Lip has a strong anti-inflammatory effect, and improves Metabolic syndromes. This study aims to provide a way to solve the challenge.
Collapse
Affiliation(s)
- Qiang Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Chunjing Guo
- College of Marine Life Science, Ocean University of China, Qingdao, PR China
| | - Zhongxin Liu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Min Cao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Wenxin Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Dandan Zhang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Hongxu Geng
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Ningning Diao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| | - Daquan Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai, PR China
| |
Collapse
|
8
|
Xie Z, Wang J, Luo Y, Qiao B, Jiang W, Zhu L, Ran H, Wang Z, Zhu W, Ren J, Zhou Z. Tumor-penetrating nanoplatform with ultrasound "unlocking" for cascade synergistic therapy and visual feedback under hypoxia. J Nanobiotechnology 2023; 21:30. [PMID: 36698190 PMCID: PMC9878980 DOI: 10.1186/s12951-023-01765-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/03/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Combined therapy based on the effects of cascade reactions of nanoplatforms to combat specific solid tumor microenvironments is considered a cancer treatment strategy with transformative clinical value. Unfortunately, an insufficient O2 supply and the lack of a visual indication hinder further applications of most nanoplatforms for solid tumor therapy. RESULTS A visualizable nanoplatform of liposome nanoparticles loaded with GOD, H(Gd), and PFP and grafted with the peptide tLyP-1, named tLyP-1H(Gd)-GOD@PFP, was constructed. The double-domain peptide tLyP-1 was used to specifically target and penetrate the tumor cells; then, US imaging, starvation therapy and sonodynamic therapy (SDT) were then achieved by the ultrasound (US)-activated cavitation effect under the guidance of MR/PA imaging. GOD not only deprived the glucose for starvation therapy but also produced H2O2, which in coordination with 1O2 produced by H(Gd), enable the effects of SDT to achieve a synergistic therapeutic effect. Moreover, the synergistic therapy was enhanced by O2 from PFP and low-intensity focused ultrasound (LIFU)-accelerated redox effects of the GOD. The present study demonstrated that the nanoplatform could generate a 3.3-fold increase in ROS, produce a 1.5-fold increase in the maximum rate of redox reactions and a 2.3-fold increase in the O2 supply in vitro, and achieve significant tumor inhibition in vivo. CONCLUSION We present a visualizable nanoplatform with tumor-penetrating ability that can be unlocked by US to overcome the current treatment problems by improving the controllability of the O2 supply, which ultimately synergistically enhanced cascade therapy.
Collapse
Affiliation(s)
- Zhuoyan Xie
- Department of Ultrasound, Chongqing General Hospital, Chongqing, 401147 China ,grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Junrui Wang
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China ,grid.412461.40000 0004 9334 6536Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Yuanli Luo
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Bin Qiao
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Weixi Jiang
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Leilei Zhu
- Department of Ultrasound, Chongqing General Hospital, Chongqing, 401147 China ,grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Haitao Ran
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Zhigang Wang
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Wei Zhu
- grid.440771.10000 0000 8820 2504Depatment of Medical College, Hubei University for Nationalities, Enshi, 445000 Hubei China
| | - Jianli Ren
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Zhiyi Zhou
- grid.412461.40000 0004 9334 6536Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China ,Depatment of General Practice, Chongqing General Hospital, Chongqing, 401147 China
| |
Collapse
|
9
|
Biabangard A, Asoodeh A, Jaafari MR, Mashreghi M. Study of FA12 peptide-modified PEGylated liposomal doxorubicin (PLD) as an effective ligand to target Muc1 in mice bearing C26 colon carcinoma: in silico, in vitro, and in vivo study. Expert Opin Drug Deliv 2022; 19:1710-1724. [PMID: 36373415 DOI: 10.1080/17425247.2022.2147505] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVES This study tried to achieve active targeting of Muc1 in cancer; the surface of PEGylated liposomal doxorubicin (PLD/Doxil®) was decorated with FA12 peptide. METHODS According to docking results, FA12 was selected for this study, among four different peptides. MD simulation was also conducted as an additional confirmation of the binding interaction between FA12 and Muc1. Liposomal formulations were prepared; 1HNMR and HPLC techniques were used to verify peptide conjugation to DSPE-PEG2000-COOH. Afterward, DSPE-PEG2000-FA12 was post-inserted into the PLD at 50, 100, 200, and 400 peptides per liposome. The size, zeta potential, release profile, cytotoxicity (IC50), and cell uptake (using fluorescence microscopy and flow cytometry) were evaluated. In vivo biodistribution and antitumor activities were studied on mice bearing C-26 colon carcinoma. RESULTS Cell uptake and cytotoxicity results revealed that PLD-100 (targeted PLD with 100 FA12 per liposome) could significantly enhance cellular binding. Furthermore, PLD-100 demonstrated higher antitumor efficacy, indicating more remarkable survival compared to PLD and other targeted PLDs. PLD-100 exhibited higher doxorubicin tumor accumulation compared to PLD. CONCLUSIONS FA12 peptide is a promising targeting ligand for PLD to treat cancers with a high level of Muc1 expression and merits further investigations.
Collapse
Affiliation(s)
- Atefeh Biabangard
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Shi Z, Liu J, Tian L, Li J, Gao Y, Xing Y, Yan W, Hua C, Xie X, Liu C, Liang C. Insights into stimuli-responsive diselenide bonds utilized in drug delivery systems for cancer therapy. Biomed Pharmacother 2022; 155:113707. [PMID: 36122520 DOI: 10.1016/j.biopha.2022.113707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Due to the complexity and particularity of cancer cell microenvironments, redox responsive drug delivery systems (DDSs) for cancer therapy have been extensively explored. Compared with widely reported cancer treatment systems based on disulfide bonds, diselenide bonds have better redox properties and greater anticancer efficiency. In this review, the significance and application of diselenide bonds in DDSs are summarized, and the stimulation sensitivity of diselenide bonds is comprehensively reported. The potential and prospects for the application of diselenide bonds in next-generation anticancer drug treatment systems are extensively discussed.
Collapse
Affiliation(s)
- Zhenfeng Shi
- Department of Urology Surgery Center, The People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830002, PR China.
| | - Jifang Liu
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China; College of Life Science, Northwest University, Xi'an 710069, PR China.
| | - Lei Tian
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China; College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Jingyi Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Yue Gao
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Yue Xing
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Wenjing Yan
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Chenyu Hua
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Xiaolin Xie
- Shaanxi Panlong Pharmaceutical Group Co., Ltd. Xi'an 710025, PR China.
| | - Chang Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Zhuhai 519030, PR China.
| | - Chengyuan Liang
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| |
Collapse
|
11
|
Li J, Gao Y, Liu S, Cai J, Zhang Q, Li K, Liu Z, Shi M, Wang J, Cui H. Aptamer-functionalized Quercetin Thermosensitive Liposomes for Targeting Drug Delivery and Antitumor Therapy. Biomed Mater 2022; 17. [PMID: 36001994 DOI: 10.1088/1748-605x/ac8c75] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022]
Abstract
Chemo-thermotherapy, as a promising cancer combination therapy strategy, has attracted widespread attention. In this study, a novel aptamer functionalized thermosensitive liposome encapsulating hydrophobic drug quercetin was fabricated as an efficient drug delivery system. This aptamer-functionalized quercetin thermosensitive liposomes (AQTSL) combined the merits of high-loading yield, sustained drug release, long-term circulation in the body of PEGylated liposomes, passive targeting provided by 100-200 nm nanoparticles, active targeting and improved internalization effects offered by AS1411 aptamer, and temperature-responsive of quercetin release. In addition, AQTSL tail vein injection combined with 42℃ water bath heating on tumor site (AQTSL+42℃)treatment inhibited the tumor growth significantly compared with the normal saline administration (p<0.01), and the inhibition rate reached 75%. Furthermore, AQTSL+42℃ treatment also slowed down the tumor growth significantly compared with QTSL combined with 42℃ administration (p<0.05), confirming that AS1411 decoration on QTSL increased the active targeting and internalization effects of the drug delivery system, and AS1411 aptamer itself might also contribute to the tumor inhibition. These data indicate that AQTSL is a potential carrier candidate for different hydrophobic drugs and tumor targeting delivery, and this kind of targeted drug delivery system combined with temperature responsive drug release mode is expected to achieve an ideal tumor therapy effect.
Collapse
Affiliation(s)
- Jian Li
- Yanshan University, No.438,Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Yanting Gao
- Yanshan University, No.438, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Shihe Liu
- Yanshan University, No.438,Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Jiahui Cai
- Yanshan University, No.438, Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Qing Zhang
- Yanshan University, No.438, Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Kun Li
- Yanshan University, No. 438, Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Zhiwei Liu
- Yanshan University, No. 438, West Section of Hebei Street, Qinhuangdao, Hebei, 066004, CHINA
| | - Ming Shi
- Yanshan University, No.438, Hebei Street, Qinhuangdao, Hebei Province, 066004, CHINA
| | - Jidong Wang
- Yanshan University, No. 438, Hebei Street, Qinhuangdao, 066000, CHINA
| | - Hongxia Cui
- Yanshan University, No. 438, Hebei Street, Qinhuangdao, Hebei Province, 066004, CHINA
| |
Collapse
|
12
|
Lou J, Qualls ML, Hudson MM, McBee DP, Baccile JA, Best MD. Reactive Oxygen Species (ROS) Activated Liposomal Cell Delivery using a Boronate-Caged Guanidine Lipid. Chemistry 2022; 28:e202201057. [PMID: 35639353 PMCID: PMC9388614 DOI: 10.1002/chem.202201057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Indexed: 08/18/2023]
Abstract
We report boronate-caged guanidine-lipid 1 that activates liposomes for cellular delivery only upon uncaging of this compound by reactive oxygen species (ROS) to produce cationic lipid products. These liposomes are designed to mimic the exceptional cell delivery properties of cell-penetrating peptides (CPPs), while the inclusion of the boronate cage is designed to enhance selectivity such that cell entry will only be activated in the presence of ROS. Boronate uncaging by hydrogen peroxide was verified by mass spectrometry and zeta potential (ZP) measurements. A microplate-based fluorescence assay was developed to study the ROS-mediated vesicle interactions between 1-liposomes and anionic membranes, which were further elucidated via dynamic light scattering (DLS) analysis. Cellular delivery studies utilizing fluorescence microscopy demonstrated significant enhancements in cellular delivery only when 1-liposomes were incubated with hydrogen peroxide. Our results showcase that lipid 1 exhibits strong potential as an ROS-responsive liposomal platform for targeted drug delivery applications.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Megan L Qualls
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Macy M Hudson
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Dillon P McBee
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Joshua A Baccile
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| |
Collapse
|
13
|
Solid Magnetoliposomes as Multi-Stimuli-Responsive Systems for Controlled Release of Doxorubicin: Assessment of Lipid Formulations. Biomedicines 2022; 10:biomedicines10051207. [PMID: 35625942 PMCID: PMC9138220 DOI: 10.3390/biomedicines10051207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
Stimuli-responsive liposomes are a class of nanocarriers whose drug release occurs, preferentially, when exposed to a specific biological environment, to an external stimulus, or both. This work is focused on the design of solid magnetoliposomes (SMLs) as lipid-based nanosystems aiming to obtain multi-stimuli-responsive vesicles for doxorubicin (DOX) controlled release in pathological areas under the action of thermal, magnetic, and pH stimuli. The effect of lipid combinations on structural, colloidal stability, and thermodynamic parameters were evaluated. The results confirmed the reproducibility for SMLs synthesis based on nine lipid formulations (combining DPPC, DSPC, CHEMS, DOPE and/or DSPE-PEG), with structural and colloidal properties suitable for biological applications. A loss of stability and thermosensitivity was observed for formulations containing dioleoylphosphatidylethanolamine (DOPE) lipid. SMLs PEGylation is an essential step to enhance both their long-term storage stability and stealth properties. DOX encapsulation (encapsulation efficiency ranging between 87% and 96%) in the bilayers lowered its pKa, which favors the displacement of DOX from the acyl chains to the surface when changing from alkaline to acidic pH. The release profiles demonstrated a preferential release at acidic pH, more pronounced under mimetic mild-hyperthermia conditions (42 °C). Release kinetics varied with the lipid formulation, generally demonstrating hyperthermia temperatures and acidic pH as determining factors in DOX release; PEGylation was shown to act as a diffusion barrier on the SMLs surface. The integrated assessment and characterization of SMLs allows tuning lipid formulations that best respond to the needs for specific controlled release profiles of stimuli-responsive nanosystems as a multi-functional approach to cancer targeting and therapy.
Collapse
|
14
|
Razavi MS, Ebrahimnejad P, Fatahi Y, D’Emanuele A, Dinarvand R. Recent Developments of Nanostructures for the Ocular Delivery of Natural Compounds. Front Chem 2022; 10:850757. [PMID: 35494641 PMCID: PMC9043530 DOI: 10.3389/fchem.2022.850757] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
Ocular disorders comprising various diseases of the anterior and posterior segments are considered as the main reasons for blindness. Natural products have been identified as potential treatments for ocular diseases due to their anti-oxidative, antiangiogenic, and anti-inflammatory effects. Unfortunately, most of these beneficial compounds are characterised by low solubility which results in low bioavailability and rapid systemic clearance thus requiring frequent administration or requiring high doses, which hinders their therapeutic applications. Additionally, the therapeutic efficiency of ocular drug delivery as a popular route of drug administration for the treatment of ocular diseases is restricted by various anatomical and physiological barriers. Recently, nanotechnology-based strategies including polymeric nanoparticles, micelles, nanofibers, dendrimers, lipid nanoparticles, liposomes, and niosomes have emerged as promising approaches to overcome limitations and enhance ocular drug bioavailability by effective delivery to the target sites. This review provides an overview of nano-drug delivery systems of natural compounds such as thymoquinone, catechin, epigallocatechin gallate, curcumin, berberine, pilocarpine, genistein, resveratrol, quercetin, naringenin, lutein, kaempferol, baicalin, and tetrandrine for ocular applications. This approach involves increasing drug concentration in the carriers to enhance drug movement into and through the ocular barriers.
Collapse
Affiliation(s)
- Malihe Sadat Razavi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Pharmaceutical Science Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Antony D’Emanuele
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom
| | - Rassoul Dinarvand
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom
| |
Collapse
|
15
|
Mazloum-Ravasan S, Mohammadi M, Hiagh EM, Ebrahimi A, Hong JH, Hamishehkar H, Kim KH. Nano-liposomal zein hydrolysate for improved apoptotic activity and therapeutic index in lung cancer treatment. Drug Deliv 2022; 29:1049-1059. [PMID: 35363101 PMCID: PMC8979517 DOI: 10.1080/10717544.2022.2057618] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Lung cancer is one of the most common cancers in the world with a high mortality rate. Zein is a protein compound whose protein isolate is not useful and whose protein hydrolysis produces biological activity. By encapsulating this bioactive compound inside the nanoparticles (NPs), it causes itself to reach the tumor site and destroy it rapidly. In this study, the effects of zein hydrolysate (ZH) and nano-liposomal ZH (N-ZH) were investigated on the human A549 cell line. Western blotting and cell cycle analyses showed that ZH and N-ZH caused cytotoxicity. They induced apoptosis via cell cycle arrest at the G0 phase, as well as significant increases in pro-apoptotic genes, such as Bax, caspase-3, -8, -9, and p53, accompanied with significant decreases in the anti-apoptotic marker Bcl-2. Based on the results, the cytotoxic and anticancer effects of N-ZH were higher than those of free ZH. In conclusion, liposomes improved the performance of ZH and dramatically reduced the IC50 value of ZH. These findings provided the experimental evidence that N-ZH with favorable anticancer activity can be used as a therapeutic agent and strategy for lung cancer treatment in future clinical trials.
Collapse
Affiliation(s)
| | - Maryam Mohammadi
- Department of Food Science and Engineering, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran.,Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Madadi Hiagh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,Pediatrics III, University Hospital Essen, Essen, Germany
| | - Alireza Ebrahimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Joo-Hyun Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
16
|
Qualls ML, Sagar R, Lou J, Best MD. Demolish and Rebuild: Controlling Lipid Self-Assembly toward Triggered Release and Artificial Cells. J Phys Chem B 2021; 125:12918-12933. [PMID: 34792362 DOI: 10.1021/acs.jpcb.1c07406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The ability to modulate the structures of lipid membranes, predicated on our nuanced understanding of the properties that drive and alter lipid self-assembly, has opened up many exciting biological applications. In this Perspective, we focus on two endeavors in which the same principles are invoked to achieve completely opposite results. On one hand, controlled liposome decomposition enables triggered release of encapsulated cargo through the development of synthetic lipid switches that perturb lipid packing in the presence of disease-associated stimuli. In particular, recent approaches have utilized artificial lipid switches designed to undergo major conformational changes in response to a range of target conditions. On the other end of the spectrum, the ability to drive the in situ formation of lipid bilayer membranes from soluble precursors is an important component in the establishment of artificial cells. This work has culminated in chemoenzymatic strategies that enable lipid manufacturing from simple components. Herein, we describe recent advancements in these two unique undertakings that are linked by their reliance on common principles of lipid self-assembly.
Collapse
Affiliation(s)
- Megan L Qualls
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Ruhani Sagar
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| |
Collapse
|
17
|
Tereshkina YA, Torkhovskaya TI, Tikhonova EG, Kostryukova LV, Sanzhakov MA, Korotkevich EI, Khudoklinova YY, Orlova NA, Kolesanova EF. Nanoliposomes as drug delivery systems: safety concerns. J Drug Target 2021; 30:313-325. [PMID: 34668814 DOI: 10.1080/1061186x.2021.1992630] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The review highlights the safety issues of drug delivery systems based on liposomes. Due to their small sizes (about 80-120 nm, sometimes even smaller), phospholipid nanoparticles interact intensively with living systems during parenteral administration. This interaction significantly affects both their transport role and safety; therefore, special attention is paid to these issues. The review summarises the data on the basic factors affecting the safety of nanoliposomes: composition, size, surface charge, stability, the release of an incorporated drug, penetration into tissues, interaction with the complement system. Attention is paid to the authors' own research of unique phospholipid nanoparticles with a diameter of 20-30 nm. The influence of technological processes of nanoliposome production on their properties is considered. The article also discusses the modern safety assessment criteria contained in the preliminary regulatory documents of the manufacturing countries for new nanoliposome-based drugs being developed or used in the clinic.
Collapse
Affiliation(s)
- Yu A Tereshkina
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - T I Torkhovskaya
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - E G Tikhonova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - L V Kostryukova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - M A Sanzhakov
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - E I Korotkevich
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - Yu Yu Khudoklinova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - N A Orlova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - E F Kolesanova
- Laboratory of Peptide Engineering, Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
18
|
Zhai B, Wu Q, Wang W, Zhang M, Han X, Li Q, Chen P, Chen X, Huang X, Li G, Zhang Q, Zhang R, Xiang Y, Liu S, Duan T, Lou J, Xie T, Sui X. Preparation, characterization, pharmacokinetics and anticancer effects of PEGylated β-elemene liposomes. Cancer Biol Med 2021; 17:60-75. [PMID: 32296587 PMCID: PMC7142831 DOI: 10.20892/j.issn.2095-3941.2019.0156] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022] Open
Abstract
Objective: This study aimed to develop a new polyethylene glycol (PEG)ylated β-elemene liposome (PEG-Lipo-β-E) and evaluate its characterization, pharmacokinetics, antitumor effects and safety in vitro and in vivo. Methods: The liposomes were prepared by ethanol injection and high-pressure micro-jet homogenization. Characterization of the liposomes was conducted, and drug content, entrapment efficiency (EE), in vitro release and stability were studied by ultra-fast liquid chromatography (UFLC) and a liquid surface method. Blood was drawn from rats to establish the pharmacokinetic parameters. The anticancer effect was evaluated in a KU-19-19 bladder cancer xenograft model. Histological analyses were performed to evaluate safety. Results: The PEG-Lipo-β-E showed good stability and was characterized as 83.31 ± 0.181 nm in size, 0.279 ± 0.004 in polydispersity index (PDI), −21.4 ± 1.06 mV in zeta potential, 6.65 ± 0.02 in pH, 5.024 ± 0.107 mg/mL in β-elemene (β-E) content, and 95.53 ± 1.712% in average EE. The Fourier transform infrared spectroscopy (FTIR) and differential scanning calorimetry (DSC) indicated the formation of PEG-Lipo-β-E. Compared to elemene injection, PEG-Lipo-β-E demonstrated a 1.75-fold decrease in clearance, a 1.62-fold increase in half-life, and a 1.76-fold increase in area under the concentration-time curves (AUCs) from 0 hour to 1.5 hours (P < 0.05). PEG-Lipo-β-E also showed an enhanced anticancer effect in vivo. Histological analyses showed that there was no evidence of toxicity to the heart, kidney, liver, lung or spleen. Conclusions: The present study demonstrates PEG-Lipo-β-E as a new formulation with ease of preparation, high EE, good stability, improved bioavailability and antitumor effects.
Collapse
Affiliation(s)
- Bingtao Zhai
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 519020, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 519020, China
| | - Wengang Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Mingming Zhang
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Xuemeng Han
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Qiujie Li
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Peng Chen
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Xiaying Chen
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Xingxing Huang
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China
| | - Guohua Li
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Qin Zhang
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Ruonan Zhang
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Yu Xiang
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Shuiping Liu
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Ting Duan
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Jianshu Lou
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Tian Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.,Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| | - Xinbing Sui
- Department of Holistic Integrative Pharmacy Institutes and Comprehensive Cancer Diagnosis and Treatment Center, the Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou 310018, China.,Key Laboratory of Elemene Class Anticancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou 310018, China
| |
Collapse
|
19
|
Zhao X, Ye Y, Ge S, Sun P, Yu P. Cellular and Molecular Targeted Drug Delivery in Central Nervous System Cancers: Advances in Targeting Strategies. Curr Top Med Chem 2021; 20:2762-2776. [PMID: 32851962 DOI: 10.2174/1568026620666200826122402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Central nervous system (CNS) cancers are among the most common and treatment-resistant diseases. The main reason for the low treatment efficiency of the disorders is the barriers against targeted delivery of anticancer agents to the site of interest, including the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB). BBB is a strong biological barrier separating circulating blood from brain extracellular fluid that selectively and actively prevents cytotoxic agents and majority of anticancer drugs from entering the brain. BBB and BBTB are the major impediments against targeted drug delivery into CNS tumors. Nanotechnology and its allied modalities offer interesting and effective delivery strategies to transport drugs across BBB to reach brain tissue. Integrating anticancer drugs into different nanocarriers improves the delivery performance of the resultant compounds across BBB. Surface engineering of nanovehicles using specific ligands, antibodies and proteins enhances the BBB crossing efficacy as well as selective and specific targeting to the target cancerous tissues in CNS tumors. Multifunctional nanoparticles (NPs) have brought revolutionary advances in targeted drug delivery to brain tumors. This study reviews the main anatomical, physiological and biological features of BBB and BBTB in drug delivery and the recent advances in targeting strategies in NPs-based drug delivery for CNS tumors. Moreover, we discuss advances in using specific ligands, antibodies, and surface proteins for designing and engineering of nanocarriers for targeted delivery of anticancer drugs to CNS tumors. Finally, the current clinical applications and the perspectives in the targeted delivery of therapeutic molecules and genes to CNS tumors are discussed.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacy, Beilun People's Hospital, Ningbo 315800, Zhejiang Province, China
| | - Yun Ye
- Department of Pharmacy, Beilun People's Hospital, Ningbo 315800, Zhejiang Province, China
| | - Shuyu Ge
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Pingping Sun
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Ping Yu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| |
Collapse
|
20
|
Sheng D, Deng L, Li P, Wang Z, Zhang Q. Perfluorocarbon Nanodroplets with Deep Tumor Penetration and Controlled Drug Delivery for Ultrasound/Fluorescence Imaging Guided Breast Cancer Therapy. ACS Biomater Sci Eng 2021; 7:605-616. [PMID: 33464814 DOI: 10.1021/acsbiomaterials.0c01333] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Danli Sheng
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Department of Ultrasound, Fudan University, Shanghai Cancer Center, Shanghai 200032, China
| | - Liming Deng
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Pan Li
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Qunxia Zhang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| |
Collapse
|
21
|
Ruiz A, Ma G, Seitsonen J, Pereira SGT, Ruokolainen J, Al-Jamal WT. Encapsulated doxorubicin crystals influence lysolipid temperature-sensitive liposomes release and therapeutic efficacy in vitro and in vivo. J Control Release 2020; 328:665-678. [PMID: 32961247 DOI: 10.1016/j.jconrel.2020.09.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Doxorubicin (DOX)-loaded lysolipid temperature-sensitive liposomes (LTSLs) are a promising stimuli-responsive drug delivery system that rapidly releases DOX in response to mild hyperthermia (HT). This study investigates the influence of loaded DOX crystals on the thermosensitivity of LTSLs and their therapeutic efficacy in vitro and in vivo. The properties of DOX crystals were manipulated using different remote loading methods (namely (NH4)2SO4, NH4-EDTA and MnSO4) and varying the lipid:DOX weight ratio during the loading step. Our results demonstrated that (NH4)2SO4 or NH4-EDTA remote loading methods had a comparable encapsulation efficiency (EE%) into LTSLs in contrast to the low DOX EE% obtained using the metal complexation method. Cryogenic transmission electron microscopy (cryo-TEM) revealed key differences in the nature of DOX crystals formed inside LTSLs based on the loading buffer or/and the lipid:DOX ratio used, resulting in different DOX release profiles in response to mild HT. The in vitro assessment of DOX release/uptake in CT26 and PC-3 cells revealed that the use of a high lipid:DOX ratio exhibited a fast and controlled release profile in combination with mild HT, which correlated well with their cytotoxicity studies. Similarly, in vivo DOX release, tumour growth inhibition and mice survival rates were influenced by the physicochemical properties of LTSLs payload. This study demonstrates, for the first time, that the characteristics of DOX crystals loaded into LTSLs, and their conformational rearrangement during HT, are important factors that impact the TSLs performance in vivo.
Collapse
Affiliation(s)
- Amalia Ruiz
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Guanglong Ma
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Jani Seitsonen
- Department of Applied Physics, Aalto University School of Science, P.O. Box 15100, FI-00076 Aalto, Finland
| | - Sara G T Pereira
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Janne Ruokolainen
- Department of Applied Physics, Aalto University School of Science, P.O. Box 15100, FI-00076 Aalto, Finland
| | - Wafa T Al-Jamal
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, United Kingdom.
| |
Collapse
|
22
|
Lou J, Best MD. Reactive Oxygen Species-Responsive Liposomes via Boronate-Caged Phosphatidylethanolamine. Bioconjug Chem 2020; 31:2220-2230. [PMID: 32808755 DOI: 10.1021/acs.bioconjchem.0c00397] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Liposomes have proven to be effective nanocarriers due to their ability to encapsulate and deliver a wide variety of therapeutic cargo. A key goal of liposome research is to enhance control over content release at diseased sites. Though a number of stimuli have been explored for triggering liposomal release, reactive oxygen species (ROS), which have received significantly less attention, provide excellent targets due to their key roles in biology and overabundance in diseased cells. Here, we report a ROS-responsive liposome platform through the inclusion of lipid 1 bearing a boronate ester headgroup and a quinone-methide (QM) generating self-immolative linker attached onto a dioleoylphosphatidylethanolamine (DOPE) lipid scaffold. Fluorescence-based dye release assays validated that this system enables release of both hydrophobic and hydrophilic contents upon hydrogen peroxide (H2O2) addition. Details of the release process were carefully studied, and data showed that oxidative removal of the boronate headgroup is sufficient to result in hydrophobic content release, while production of DOPE is needed for hydrophilic cargo leakage. These results showcase that lipid 1 can serve as a promising ROS-responsive liposomal delivery platform for controlled release.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| |
Collapse
|
23
|
Hong C, Liang J, Xia J, Zhu Y, Guo Y, Wang A, Lu C, Ren H, Chen C, Li S, Wang D, Zhan H, Wang J. One Stone Four Birds: A Novel Liposomal Delivery System Multi-functionalized with Ginsenoside Rh2 for Tumor Targeting Therapy. NANO-MICRO LETTERS 2020; 12:129. [PMID: 34138128 PMCID: PMC7770862 DOI: 10.1007/s40820-020-00472-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/19/2020] [Indexed: 05/08/2023]
Abstract
Liposomes hold great potential in anti-cancer drug delivery and the targeting treatment of tumors. However, the clinical therapeutic efficacy of liposomes is still limited by the complexity of tumor microenvironment (TME) and the insufficient accumulation in tumor sites. Meanwhile, the application of cholesterol and polyethylene glycol (PEG), which are usually used to prolong the blood circulation and stabilize the structure of liposomes respectively, has been questioned due to various disadvantages. Herein, we developed a ginsenoside Rh2-based multifunctional liposome system (Rh2-lipo) to effectively address these challenges once for all. Different with the conventional 'wooden' liposomes, Rh2-lipo is a much more brilliant carrier with multiple functions. In Rh2-lipo, both cholesterol and PEG were substituted by Rh2, which works as membrane stabilizer, long-circulating stealther, active targeting ligand, and chemotherapy adjuvant at the same time. Firstly, Rh2 could keep the stability of liposomes and avoid the shortcomings caused by cholesterol. Secondly, Rh2-lipo showed a specifically prolonged circulation behavior in the blood. Thirdly, the accumulation of the liposomes in the tumor was significantly enhanced by the interaction of glucose transporter of tumor cells with Rh2. Fourth, Rh2-lipo could remodel the structure and reverse the immunosuppressive environment in TME. When tested in a 4T1 breast carcinoma xenograft model, the paclitaxel-loaded Rh2-lipo realized high efficient tumor growth suppression. Therefore, Rh2-lipo not only innovatively challenges the position of cholesterol as a liposome component, but also provides another innovative potential system with multiple functions for anti-cancer drug delivery.
Collapse
Affiliation(s)
- Chao Hong
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Jianming Liang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Ying Zhu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Yizhen Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Anni Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Chunyi Lu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Hongwei Ren
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Chen Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Shiyi Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Dan Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
- Shanghai Ginposome Pharmatech Co., Ltd, Shanghai, 201600, People's Republic of China
| | - Huaxing Zhan
- Shanghai Ginposome Pharmatech Co., Ltd, Shanghai, 201600, People's Republic of China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China.
- Institute of Integrated Chinese and Western Medicine, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
24
|
Zhang J, Li X, Huang L. Anticancer activities of phytoconstituents and their liposomal targeting strategies against tumor cells and the microenvironment. Adv Drug Deliv Rev 2020; 154-155:245-273. [PMID: 32473991 PMCID: PMC7704676 DOI: 10.1016/j.addr.2020.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Various bioactive ingredients have been extracted from Chinese herbal medicines (CHMs) that affect tumor progression and metastasis. To further understand the mechanisms of CHMs in cancer therapy, this article summarizes the effects of five categories of CHMs and their active ingredients on tumor cells and the tumor microenvironment. Despite their treatment potential, the undesirable physicochemical properties (poor permeability, instability, high hydrophilicity or hydrophobicity, toxicity) and unwanted pharmacokinetic profiles (short half-life in blood and low bioavailability) restrict clinical studies of CHMs. Therefore, development of liposomes through relevant surface modifying techniques to achieve targeted CHM delivery for cancer cells, i.e., extracellular and intracellular targets and targets in tumor microenvironment or vasculature, have been reviewed. Current challenges of liposomal targeting of these phytoconstituents and future perspective of CHM applications are discussed to provide an informative reference for interested readers.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Xiang Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
25
|
Tan M, Chen Y, Guo Y, Yang C, Liu M, Guo D, Wang Z, Cao Y, Ran H. A low-intensity focused ultrasound-assisted nanocomposite for advanced triple cancer therapy: local chemotherapy, therapeutic extracellular vesicles and combined immunotherapy. Biomater Sci 2020; 8:6703-6717. [PMID: 33108411 DOI: 10.1039/d0bm00804d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
CCs-SF/DOX nanocomposite based triple therapies of “local chemotherapy-therapeutic EVs-synergistic immunotherapy” irradiated by LIFU.
Collapse
Affiliation(s)
- Mixiao Tan
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Yuli Chen
- Chongqing Dazu District People's Hospital
- Chongqing
- China
| | - Yuan Guo
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Chao Yang
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Mingzhu Liu
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Dan Guo
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Zhigang Wang
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Yang Cao
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| | - Haitao Ran
- The Second Affiliated Hospital of Chongqing Medical University & Chongqing Key Laboratory of Ultrasound Molecular Imaging
- Chongqing
- China
| |
Collapse
|
26
|
Li H, Li Q, Hou W, Zhang J, Yu C, Zeng D, Liu G, Li F. Enzyme-Catalytic Self-Triggered Release of Drugs from a Nanosystem for Efficient Delivery to Nuclei of Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:43581-43587. [PMID: 31664812 DOI: 10.1021/acsami.9b15460] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Stimulus-responsive drug delivery nanosystems (DDSs) are of great significance in improving cancer therapy for intelligent control over drug release. However, among them, many DDSs are unable to realize rapid and sufficient drug release because most internal stimulants might be consumed during the release process. To address the plight, an abundant supply of stimulants is highly desirable. Herein, a core crosslinked pullulan-di-(4,1-hydroxybenzylene)diselenide nanosystem, which could generate abundant exogenous-stimulant reactive oxygen species (ROS) via tumor-specific NAD(P)H:quinone oxidoreductase-1 (NQO1) catalysis, was constructed by the encapsulation of β-lapachone. The enzyme-catalytic-generated ROS induced self-triggered cascade amplification release of loaded doxorubicin (DOX) in the tumor cells, thus achieving efficient delivery of DOX to the nuclei of tumor cells by breaking the diselenide bond of the nanosystem. As a result, the antitumor effect of this nanosystem was significantly improved in the HepG2 xenograft model. In general, this study offers a new paradigm for utilizing the interaction between the loaded agent and carrier in the tumor cells to obtain self-triggered drug release in the design of DDSs for enhanced cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , Fujian , P. R. China
| | | |
Collapse
|
27
|
Bi H, Xue J, Jiang H, Gao S, Yang D, Fang Y, Shi K. Current developments in drug delivery with thermosensitive liposomes. Asian J Pharm Sci 2019; 14:365-379. [PMID: 32104466 PMCID: PMC7032122 DOI: 10.1016/j.ajps.2018.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/26/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022] Open
Abstract
Thermosensitive liposomes (TSLs) have been an important research area in the field of tumor targeted chemotherapy. Since the first TSLs appeared that using 1,2-dipalmitoyl-sn-glyce-ro-3-phosphocholine (DPPC) as the primary liposomal lipid, many studies have been done using this type of liposome from basic and practical aspects. While TSLs composed of DPPC enhance the cargo release near the phase transition temperature, it has been shown that many factors affect their temperature sensitivity. Thus numerous attempts have been undertaken to develop new TSLs for improving their thermal response performance. The main objective of this review is to introduce the development and recent update of innovative TSLs formulations, including combination of radiofrequency ablation (RFA), high-intensity focused ultrasound (HIFU), magnetic resonance imaging (MRI) and alternating magnetic field (AMF). In addition, various factors affecting the design of TSLs, such as lipid composition, surfactant, size and serum components are also discussed.
Collapse
Key Words
- (DPPC), 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine
- (DPPGOG), 1,2-dipalmitoyl-sn-glycero-3-phosphoglyceroglycerol
- (DSPC), 1,2-distearoyl-sn-glycero-3-phosphocholine
- (DSPE-mPEG2000), 1,2-distearoyl-sn-glycero-3-phosphatiylethanol-amine-N-[methoxy(polyethyleneglycol)-2000]
- (LTSLs), lyso-lipid temperature sensitive liposomes
- (MPPC), 1-myristoyl-2-palmitoyl-sn-glycero-3-phosphatidylcholine
- (MSPC), 1-stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine
- (P-lyso-PC), lysophosphatidylcholine
- (P188), 1-palmitoyl-2-stearoyl-sn-glycero-3-phosphatidylcholinex
- (P188), HO-(C2H4O)a-(C3H6O)b-(C2H4O)c-H, a=80, b=27, c=80
- Content release rate
- Drug delivery
- Hyperthermia
- Smart liposomes
- Thermosensitive liposomes
- Tumor chemotherapy
- fTSLs, fast release TSLs
- sTSLs, slow release TSLs
Collapse
Affiliation(s)
- Hongshu Bi
- Institute of New Drug Development, Liaoning Yaolian Pharmaceutical Co., Ltd., Benxi, Liaoning 117004, China
| | - Jianxiu Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Hong Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Shan Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Dongjuan Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Yan Fang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Kai Shi
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| |
Collapse
|
28
|
Wang C, Han M, Liu X, Chen S, Hu F, Sun J, Yuan H. Mitoxantrone-preloaded water-responsive phospholipid-amorphous calcium carbonate hybrid nanoparticles for targeted and effective cancer therapy. Int J Nanomedicine 2019; 14:1503-1517. [PMID: 30880961 PMCID: PMC6396884 DOI: 10.2147/ijn.s193976] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The application of mitoxantrone (MIT) in cancer therapy has been severely limited by its inherent drawbacks. In addition, effective cancer therapy calls for drug release systems capable of enforcing drug release within cancer cells in response to infinite stimulant with enhanced drug penetration capability. METHODS MIT-preloaded phospholipid-amorphous calcium carbonate hybrid nanoparticles (PL/ACC-MIT) that surface modified with PL shell (containing shielding polymer polyethylene glycol and targeting moiety folic acid) were prepared by a facile solvent-diffusion method. RESULTS It has been proven that the resulting PL/ACC-MIT nanoparticles demonstrated satisfactory stability against various aqueous environments with minimal drug leakage and exerted strong targeting capability but selective preference to the folate receptor-overexpressing cell line. In contrast, once exposed to the enzyme-abundant and acidic environments of cancer cells, the PL/ACC-MIT nanoparticles can readily decompose to facilitate quick drug release and enhanced drug penetration to yield preferable antitumor effect both in vitro and in vivo. CONCLUSION In this study, MIT-preloaded water-responsive hybrid nanoparticles with increased stability, targetability, controlled drug release, and enhanced drug penetration were successfully developed, which might be a candidate for targeted and effective cancer therapy.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
| | - Min Han
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
- Hangzhou Zhongmei Huadong Pharmaceutical Co, Ltd, Hangzhou 310011, China
| | - Xuerong Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
| | - Shaoqing Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
| | - Fuqiang Hu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China,
| | - Hong Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China,
| |
Collapse
|
29
|
Lopez A, Liu J. DNA Oligonucleotide-Functionalized Liposomes: Bioconjugate Chemistry, Biointerfaces, and Applications. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:15000-15013. [PMID: 29936848 DOI: 10.1021/acs.langmuir.8b01368] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Interfacing DNA with liposomes has produced a diverse range of programmable soft materials, devices, and drug delivery vehicles. By simply controlling liposomal composition, bilayer fluidity, lipid domain formation, and surface charge can be systematically varied. Recent development in DNA research has produced not only sophisticated nanostructures but also new functions including ligand binding and catalysis. For noncationic liposomes, a DNA is typically covalently linked to a hydrophobic or lipid moiety that can be inserted into lipid membranes. In this article, we discuss fundamental biointerfaces formed between DNA and noncationic liposomes. The methods to prepare such conjugates and the interactions at the membrane interfaces are also discussed. The effect of DNA lateral diffusion on fluid bilayer membranes and the effect of membrane on DNA assembly are emphasized. DNA hybridization can be programmed to promote fusion of lipid membranes. Representative applications of this conjugate for drug delivery, biosensor development, and directed assembly of materials are briefly described toward the end. Some future research directions are also proposed to further understand this biointerface.
Collapse
Affiliation(s)
- Anand Lopez
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| |
Collapse
|
30
|
Zhai B, Zeng Y, Zeng Z, Zhang N, Li C, Zeng Y, You Y, Wang S, Chen X, Sui X, Xie T. Drug delivery systems for elemene, its main active ingredient β-elemene, and its derivatives in cancer therapy. Int J Nanomedicine 2018; 13:6279-6296. [PMID: 30349250 PMCID: PMC6186893 DOI: 10.2147/ijn.s174527] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
β-elemene is a noncytotoxic Class II antitumor drug extracted from the traditional Chinese medicine Curcuma wenyujin Y. H. Chen et C. Ling. β-elemene exerts its effects by inhibiting cell proliferation, arresting the cell cycle, inducing cell apoptosis, exerting antiangiogenesis and antimetastasis effects, reversing multiple-drug resistance (MDR), and enhancing the immune system. Elemene injection and oral emulsion have been used to treat various tumors, including cancer of the lung, liver, brain, breast, ovary, gastric, prostate, and other tissues, for >20 years. The safety of both elemene injection and oral emulsion in the clinic has been discussed. Recently, the secondary development of β-elemene has attracted the attention of researchers and made great progress. On the one hand, studies have been carried out on liposome-based systems (including solid lipid nanoparticles [SLNs], nanostructured lipid carriers [NLCs], long-circulating liposomes, active targeting liposomes, and multidrug-loaded liposomes) and emulsion systems (including microemulsions, self-emulsion drug delivery systems [SEDDSs], and active targeting microemulsion) to solve the issues of poor solubility in water, low bioavailability, and severe phlebitis, as well as to improve antitumor efficacy. The pharmacokinetics of different drug delivery systems of β-elemene are also summarized. On the other hand, a number of highly active anticancer β-elemene derivatives have been obtained through modification of the structure of β-elemene. This review focuses on the two drug delivery systems and derivatives of β-elemene for cancer therapy.
Collapse
Affiliation(s)
- Bingtao Zhai
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yiying Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
- College of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaowu Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Nana Zhang
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Chenxi Li
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Yijun Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Yu You
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuling Wang
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Xiabin Chen
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Xinbing Sui
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| | - Tian Xie
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, Zhejiang, China, ;
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China, ;
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou, Zhejiang, China, ;
| |
Collapse
|
31
|
de Matos MBC, Beztsinna N, Heyder C, Fens MHAM, Mastrobattista E, Schiffelers RM, Leneweit G, Kok RJ. Thermosensitive liposomes for triggered release of cytotoxic proteins. Eur J Pharm Biopharm 2018; 132:211-221. [PMID: 30223028 DOI: 10.1016/j.ejpb.2018.09.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/22/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
Abstract
Lysolipid-containing thermosensitive liposomes (LTSL) are clinically-relevant drug nanocarriers which have been used to deliver small molecule cytostatics to tumors in combination with local hyperthermia (42 °C) to trigger local drug release. The objective of this study was to investigate the feasibility of LTSL for encapsulation and triggered release of macromolecular drugs such as plant-derived cytotoxins. As therapeutic protein we used Mistletoe lectin-1 (ML1) - a ribosome-inactivating protein with potent cytotoxic activity in tumor cells. Model macromolecules (dextrans, albumin) and ML1 were encapsulated in small unilamellar LTSL with varying lipid compositions by the thin film hydration method and extrusion. LTSLs showed molecular weight dependent heat-triggered release of the loaded cargo. The most promising composition, ML1 formulated in LTSL composed of 86:10:4 %mol DPPC:MSPC:DSPE-PEG2000, was further studied for bioactivity against murine CT26 colon carcinoma cells. Confocal live-cell imaging showed uptake of released ML1 after mild hyperthermia at 42 °C, subsequently leading to potent cytotoxicity by LTSL-ML1. Our study shows that LTSL in combination with localized hyperthermia hold promise as local tumor delivery strategy for macromolecular cytotoxins.
Collapse
Affiliation(s)
- Maria B C de Matos
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Nataliia Beztsinna
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | | | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- Laboratory Clinical Chemistry & Haematology, University Medical Center Utrecht, the Netherlands
| | | | - Robbert J Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
32
|
Hua S, de Matos MBC, Metselaar JM, Storm G. Current Trends and Challenges in the Clinical Translation of Nanoparticulate Nanomedicines: Pathways for Translational Development and Commercialization. Front Pharmacol 2018; 9:790. [PMID: 30065653 PMCID: PMC6056679 DOI: 10.3389/fphar.2018.00790] [Citation(s) in RCA: 547] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023] Open
Abstract
The use of nanotechnology in medicine has the potential to have a major impact on human health for the prevention, diagnosis, and treatment of diseases. One particular aspect of the nanomedicine field which has received a great deal of attention is the design and development of nanoparticulate nanomedicines (NNMs) for drug delivery (i.e., drug-containing nanoparticles). NNMs are intended to deliver drugs via various mechanisms: solubilization, passive targeting, active targeting, and triggered release. The NNM approach aims to increase therapeutic efficacy, decrease the therapeutically effective dose, and/or reduce the risk of systemic side effects. In order to move a NNM from the bench to the bedside, several experimental challenges need to be addressed. This review will discuss the current trends and challenges in the clinical translation of NNMs as well as the potential pathways for translational development and commercialization. Key issues related to the clinical development of NNMs will be covered, including biological challenges, large-scale manufacturing, biocompatibility and safety, intellectual property (IP), government regulations, and overall cost-effectiveness in comparison to current therapies. These factors can impose significant hurdles limiting the appearance of NNMs on the market, irrelevant of whether they are therapeutically beneficial or not.
Collapse
Affiliation(s)
- Susan Hua
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW, Australia
| | - Maria B C de Matos
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands.,Department of Experimental Molecular Imaging, RWTH University Clinic Aachen, Aachen, Germany
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands.,Imaging Division, University Medical Centre Utrecht, Utrecht, Netherlands
| |
Collapse
|
33
|
Wang C, Chen S, Wang Y, Liu X, Hu F, Sun J, Yuan H. Lipase-Triggered Water-Responsive "Pandora's Box" for Cancer Therapy: Toward Induced Neighboring Effect and Enhanced Drug Penetration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706407. [PMID: 29484719 DOI: 10.1002/adma.201706407] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/12/2018] [Indexed: 05/20/2023]
Abstract
Insufficient drug release as well as poor drug penetration are major obstacles for effective nanoparticles (NPs)-based cancer therapy. Herein, the high aqueous instability of amorphous calcium carbonate (ACC) is employed to construct doxorubicin (DOX) preloaded and monostearin (MS) coated "Pandora's box" (MS/ACC-DOX) NPs for lipase-triggered water-responsive drug release in lipase-overexpressed tumor tissue to induce a neighboring effect and enhance drug penetration. MS as a solid lipid can prevent potential drug leakage of ACC-DOX NPs during the circulatory process, while it can be readily be disintegrated in lipase-overexpressed SKOV3 cells to expose the ACC-DOX core. The high aqueous instability of ACC will lead to burst release of the encapsulated DOX to induce apoptosis and cytotoxicity to kill the tumor cells. The liberated NPs from the dead or dying cells continue to respond to the ubiquitous aqueous environment to sufficiently release DOX once unpacked, like the "Pandora's box", leading to severe cytotoxicity to neighboring cells (neighboring effect). Moreover, the continuously released free DOX molecules can readily diffused through the tumor extracellular matrix to enhance drug penetration to deep tumor tissue. Both effects contribute to achieve elevated antitumor benefits.
Collapse
Affiliation(s)
- Cheng Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Shaoqing Chen
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Yunxin Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Xuerong Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Jihong Sun
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Hangzhou, 310016, China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| |
Collapse
|
34
|
Lambruschini C, Villa S, Banfi L, Canepa F, Morana F, Relini A, Riani P, Riva R, Silvetti F. Enzymatically promoted release of organic molecules linked to magnetic nanoparticles. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2018; 9:986-999. [PMID: 29719751 PMCID: PMC5905276 DOI: 10.3762/bjnano.9.92] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/27/2018] [Indexed: 06/08/2023]
Abstract
Magnetite-based magnetic nanoparticles have been successfully coupled to an organic system constituted of a fluorescent molecule, a tripeptide specifier and a spacer. The system is able to selectively release the fluorescent molecule upon targeted enzymatic hydrolysis promoted by a lysine/arginine specific protease.
Collapse
Affiliation(s)
- Chiara Lambruschini
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Silvia Villa
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Luca Banfi
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Fabio Canepa
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Fabio Morana
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Annalisa Relini
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Paola Riani
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Renata Riva
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Fulvio Silvetti
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| |
Collapse
|
35
|
Børresen B, Hansen AE, Kjaer A, Andresen TL, Kristensen AT. Liposome-encapsulated chemotherapy: Current evidence for its use in companion animals. Vet Comp Oncol 2017; 16:E1-E15. [PMID: 29027350 DOI: 10.1111/vco.12342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/31/2017] [Indexed: 12/01/2022]
Abstract
Cytotoxic drugs encapsulated into liposomes were originally designed to increase the anticancer response, while minimizing off-target adverse effects. The first liposomal chemotherapeutic drug was approved for use in humans more than 20 years ago, and the first publication regarding its use in a canine cancer patient was published shortly thereafter. Regardless, no general application for liposomal cytotoxic drugs has been established in veterinary oncology till now. Due to the popularity of canines as experimental models for pharmacokinetic analyses and toxicity studies, multiple publications exist describing various liposomal drugs in healthy dogs. Also, some evidence for its use in veterinary cancer patients exists, especially in canine lymphoma, canine splenic hemangiosarcoma and feline soft tissue sarcoma, however, the results have not been overwhelming. Reasons for this may be related to inherent issues with the enhanced permeability and retention effect, the tumour phenomenon which liposomal drugs exploit. This effect seems very heterogeneously distributed in the tumour. Also, it is potentially not as ubiquitously occurring as once thought, and it may prove important to select patients for liposomal therapy on an individual, non-histology-oriented, basis. Concurrently, new developments with active-release modified liposomes in experimental models and humans will likely be relevant for veterinary patients as well, and holds the potential to improve the therapeutic response. It, however, does not resolve the other challenges that liposomal chemotherapy faces, and more work still needs to be done to determine which veterinary patients may benefit the most from liposomal chemotherapy.
Collapse
Affiliation(s)
- B Børresen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - A E Hansen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - A Kjaer
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - T L Andresen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - A T Kristensen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
36
|
Revisiting the use of sPLA 2 -sensitive liposomes in cancer therapy. J Control Release 2017; 261:163-173. [DOI: 10.1016/j.jconrel.2017.06.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/21/2017] [Accepted: 06/24/2017] [Indexed: 11/24/2022]
|
37
|
Dai Y, Xu C, Sun X, Chen X. Nanoparticle design strategies for enhanced anticancer therapy by exploiting the tumour microenvironment. Chem Soc Rev 2017; 46:3830-3852. [PMID: 28516983 PMCID: PMC5521825 DOI: 10.1039/c6cs00592f] [Citation(s) in RCA: 643] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanovehicles can efficiently carry and deliver anticancer agents to tumour sites. Compared with normal tissue, the tumour microenvironment has some unique properties, such as vascular abnormalities, hypoxia and acidic pH. There are many types of cells, including tumour cells, macrophages, immune and fibroblast cells, fed by defective blood vessels in the solid tumour. Exploiting the tumour microenvironment can benefit the design of nanoparticles for enhanced therapeutic effectiveness. In this review article, we summarized the recent progress in various nanoformulations for cancer therapy, with a special emphasis on tumour microenvironment stimuli-responsive ones. Numerous tumour microenvironment modulation strategies with promising cancer therapeutic efficacy have also been highlighted. Future challenges and opportunities of design consideration are also discussed in detail. We believe that these tumour microenvironment modulation strategies offer a good chance for the practical translation of nanoparticle formulas into clinic.
Collapse
Affiliation(s)
- Yunlu Dai
- Centre for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen 361102, China. and Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | - Can Xu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | - Xiaolian Sun
- Centre for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiang'an South Road, Xiamen 361102, China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
38
|
Guo Y, Zhang Y, Ma J, Li Q, Li Y, Zhou X, Zhao D, Song H, Chen Q, Zhu X. Light/magnetic hyperthermia triggered drug released from multi-functional thermo-sensitive magnetoliposomes for precise cancer synergetic theranostics. J Control Release 2017; 272:145-158. [PMID: 28442407 DOI: 10.1016/j.jconrel.2017.04.028] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/06/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022]
Abstract
Precise delivery of antineoplastic drugs to specific tumor region has drawn much attention in recent years. Herein, a light/magnetic hyperthermia triggered drug delivery with multiple functionality is designed based on methotrexate (MTX) modified thermo-sensitive magnetoliposomes (MTX-MagTSLs). In this system, MTX and oleic acid modified magnetic nanoparticles (MNPs) can be applied in biological and magnetic targeting. Meanwhile, lipophilic fluorescent dye Cy5.5 and MNPs are encapsulated into the bilayer of liposomes, which can not only achieve dual-imaging effect to verify the MTX-MagTSLs accumulation in tumor region, but also provide an appropriate laser irradiation region to release Doxorubicin (Dox) under alternating magnetic field (AMF). Both in vitro and in vivo results revealed that MTX-MagTSLs possessed an excellent targeting ability towards HeLa cells and HeLa tumor-bearing mice. Furthermore, the heating effect of MTX-MagTSLs was amplified 4.2-fold upon combination with AMF and local precise near-infrared laser irradiation (808nm) (DUAL-mode) to rapidly reach the phase change temperature (Tm) of MTX-MagTSLs in 5min compared with either AMF or laser stimulation alone, resulting in a significantly enhanced release of Dox at tumor region and precise cancer synergetic theranostics.
Collapse
Affiliation(s)
- Yuxin Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Jinyuan Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qi Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yang Li
- College of Materials, Xiamen University, Xiamen, China
| | - Xinyi Zhou
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Dan Zhao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Hua Song
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qing Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
39
|
Sun Q, Zhou Z, Qiu N, Shen Y. Rational Design of Cancer Nanomedicine: Nanoproperty Integration and Synchronization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1606628. [PMID: 28234430 DOI: 10.1002/adma.201606628] [Citation(s) in RCA: 726] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Indexed: 05/21/2023]
Abstract
Current cancer nanomedicines can only mitigate adverse effects but fail to enhance therapeutic efficacies of anticancer drugs. Rational design of next-generation cancer nanomedicines should aim to enhance their therapeutic efficacies. Taking this into account, this review first analyzes the typical cancer-drug-delivery process of an intravenously administered nanomedicine and concludes that the delivery involves a five-step CAPIR cascade and that high efficiency at every step is critical to guarantee high overall therapeutic efficiency. Further analysis shows that the nanoproperties needed in each step for a nanomedicine to maximize its efficiency are different and even opposing in different steps, particularly what the authors call the PEG, surface-charge, size and stability dilemmas. To resolve those dilemmas in order to integrate all needed nanoproperties into one nanomedicine, stability, surface and size nanoproperty transitions (3S transitions for short) are proposed and the reported strategies to realize these transitions are comprehensively summarized. Examples of nanomedicines capable of the 3S transitions are discussed, as are future research directions to design high-performance cancer nanomedicines and their clinical translations.
Collapse
Affiliation(s)
- Qihang Sun
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| |
Collapse
|
40
|
Tagami T, Ozeki T. Recent Trends in Clinical Trials Related to Carrier-Based Drugs. J Pharm Sci 2017; 106:2219-2226. [PMID: 28259767 DOI: 10.1016/j.xphs.2017.02.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023]
Abstract
Clinical trials related to carrier-based drugs have recently attracted attention because carrier-based drugs hold promise for high efficiency drug delivery and for reducing drug-related side effects. In this commentary, we introduce recent clinical trials involving the use of various carriers, including liposomes, nano and micro particles, micelles, emulsions, and polymeric carriers. Liposomal drug carriers are currently the most intensively tested carriers in clinical trials, but other carriers such as polymeric carriers, albumin-based carriers, and metal nanocarriers have also recently been studied in clinical trials. Each carrier has specific properties, advantages, and disadvantages. The recent clinical trials introduced herein provide information critical to understanding current trends in carrier-based drug research.
Collapse
Affiliation(s)
- Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
| |
Collapse
|
41
|
Plourde K, Derbali RM, Desrosiers A, Dubath C, Vallée-Bélisle A, Leblond J. Aptamer-based liposomes improve specific drug loading and release. J Control Release 2017; 251:82-91. [PMID: 28238787 DOI: 10.1016/j.jconrel.2017.02.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/27/2017] [Accepted: 02/07/2017] [Indexed: 01/02/2023]
Abstract
Aptamer technology has shown much promise in cancer therapeutics for its targeting abilities. However, its potential to improve drug loading and release from nanocarriers has not been thoroughly explored. In this study, we employed drug-binding aptamers to actively load drugs into liposomes. We designed a series of DNA aptamer sequences specific to doxorubicin, displaying multiple binding sites and various binding affinities. The binding ability of aptamers was preserved when incorporated into cationic liposomes, binding up to 15equivalents of doxorubicin per aptamer, therefore drawing the drug into liposomes. Optimization of the charge and drug/aptamer ratios resulted in ≥80% encapsulation efficiency of doxorubicin, ten times higher than classical passively-encapsulating liposomal formulations and similar to a pH-gradient active loading strategy. In addition, kinetic release profiles and cytotoxicity assay on HeLa cells demonstrated that the release and therapeutic efficacy of liposomal doxorubicin could be controlled by the aptamer's structure. Our results suggest that the aptamer exhibiting a specific intermediate affinity is the best suited to achieve high drug loading while maintaining efficient drug release and therapeutic activity. This strategy was successfully applied to tobramycin, a hydrophilic drug suffering from low encapsulation into liposomes, where its loading was improved six-fold using aptamers. Overall, we demonstrate that aptamers could act, in addition to their targeting properties, as multifunctional excipients for liposomal formulations.
Collapse
Affiliation(s)
- Kevin Plourde
- Faculty of Pharmacy, University of Montreal, QC H3T 1J4, Canada
| | | | | | - Céline Dubath
- Faculty of Pharmacy, University of Montreal, QC H3T 1J4, Canada
| | | | - Jeanne Leblond
- Faculty of Pharmacy, University of Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
42
|
Mizukami S, Kashibe M, Matsumoto K, Hori Y, Kikuchi K. Enzyme-triggered compound release using functionalized antimicrobial peptide derivatives. Chem Sci 2017; 8:3047-3053. [PMID: 28451373 PMCID: PMC5380883 DOI: 10.1039/c6sc04435b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/07/2017] [Indexed: 12/03/2022] Open
Abstract
Two strategies have been proposed to develop enzyme-triggered compound release systems.
Controlled release is one of the key technologies for medical innovation, and many stimulus-responsive nanocarriers have been developed to utilize this technology. Enzyme activity is one of the most useful stimuli, because many enzymes are specifically activated in diseased tissues. However, controlled release stimulated by enzyme activity has not been frequently reported. One of the reasons for this is the lack of versatility of carriers. Most of the reported stimulus-responsive systems involve a sophisticated design and a complicated process for the synthesis of stimulus-responsive nanocarrier components. The purpose of this study was to develop versatile controlled release systems triggered by various stimuli, including enzyme activity, without modifying the nanocarrier components. We developed two controlled release systems, both of which comprised a liposome as the nanocarrier and a membrane-damaging peptide, temporin L (TL), and its derivatives as the release-controllers. One system utilized branched peptides for proteases, and the other utilized phosphopeptides for phosphatases. In our systems, the target enzymes converted the non-membrane-damaging TL derivatives into membrane-damaging peptides and released the liposome inclusion. We demonstrated the use of our antimicrobial peptide-based controlled release systems for different enzymes and showed the promise of this technology as a novel theranostic tool.
Collapse
Affiliation(s)
- Shin Mizukami
- Institute of Multidisciplinary Research for Advanced Materials , Tohoku University , 2-1-1C Katahira, Aoba-ku , Sendai , Miyagi 980-8577 , Japan .
| | - Masayoshi Kashibe
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan .
| | - Kengo Matsumoto
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan .
| | - Yuichiro Hori
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan . .,Immunology Frontier Research Center , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan
| | - Kazuya Kikuchi
- Division of Advanced Science and Biotechnology , Graduate School of Engineering , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan . .,Immunology Frontier Research Center , Osaka University , 2-1 Yamadaoka, Suita , Osaka 565-0871 , Japan
| |
Collapse
|
43
|
Oude Blenke E, Sleszynska M, Evers MJW, Storm G, Martin NI, Mastrobattista E. Strategies for the Activation and Release of the Membranolytic Peptide Melittin from Liposomes Using Endosomal pH as a Trigger. Bioconjug Chem 2017; 28:574-582. [DOI: 10.1021/acs.bioconjchem.6b00677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- E. Oude Blenke
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - M. Sleszynska
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - M. J. W. Evers
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - G. Storm
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - N. I. Martin
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - E. Mastrobattista
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| |
Collapse
|
44
|
Luo D, Carter KA, Miranda D, Lovell JF. Chemophototherapy: An Emerging Treatment Option for Solid Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2017; 4:1600106. [PMID: 28105389 PMCID: PMC5238751 DOI: 10.1002/advs.201600106] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/21/2016] [Indexed: 05/17/2023]
Abstract
Near infrared (NIR) light penetrates human tissues with limited depth, thereby providing a method to safely deliver non-ionizing radiation to well-defined target tissue volumes. Light-based therapies including photodynamic therapy (PDT) and laser-induced thermal therapy have been validated clinically for curative and palliative treatment of solid tumors. However, these monotherapies can suffer from incomplete tumor killing and have not displaced existing ablative modalities. The combination of phototherapy and chemotherapy (chemophototherapy, CPT), when carefully planned, has been shown to be an effective tumor treatment option preclinically and clinically. Chemotherapy can enhance the efficacy of PDT by targeting surviving cancer cells or by inhibiting regrowth of damaged tumor blood vessels. Alternatively, PDT-mediated vascular permeabilization has been shown to enhance the deposition of nanoparticulate drugs into tumors for enhanced accumulation and efficacy. Integrated nanoparticles have been reported that combine photosensitizers and drugs into a single agent. More recently, light-activated nanoparticles have been developed that release their payload in response to light irradiation to achieve improved drug bioavailability with superior efficacy. CPT can potently eradicate tumors with precise spatial control, and further clinical testing is warranted.
Collapse
Affiliation(s)
- Dandan Luo
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260
| | - Kevin A. Carter
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260
| | - Dyego Miranda
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260
| |
Collapse
|
45
|
Goyal R, Macri LK, Kaplan HM, Kohn J. Nanoparticles and nanofibers for topical drug delivery. J Control Release 2016; 240:77-92. [PMID: 26518723 PMCID: PMC4896846 DOI: 10.1016/j.jconrel.2015.10.049] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/11/2023]
Abstract
This review provides the first comprehensive overview of the use of both nanoparticles and nanofibers for topical drug delivery. Researchers have explored the use of nanotechnology, specifically nanoparticles and nanofibers, as drug delivery systems for topical and transdermal applications. This approach employs increased drug concentration in the carrier, in order to increase drug flux into and through the skin. Both nanoparticles and nanofibers can be used to deliver hydrophobic and hydrophilic drugs and are capable of controlled release for a prolonged period of time. The examples presented provide significant evidence that this area of research has - and will continue to have - a profound impact on both clinical outcomes and the development of new products.
Collapse
Affiliation(s)
- Ritu Goyal
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Lauren K Macri
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Hilton M Kaplan
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, 145 Bevier Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
46
|
Li Z, Ye E, Lakshminarayanan R, Loh XJ. Recent Advances of Using Hybrid Nanocarriers in Remotely Controlled Therapeutic Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:4782-4806. [PMID: 27482950 DOI: 10.1002/smll.201601129] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/27/2016] [Indexed: 06/06/2023]
Abstract
The development of hybrid biomaterials has been attracting great attention in the design of materials for biomedicine. The nanosized level of inorganic and organic or even bioactive components can be combined into a single material by this approach, which has created entirely new advanced compositions with truly unique properties for drug delivery. The recent advances in using hybrid nanovehicles as remotely controlled therapeutic delivery carriers are summarized with respect to different nanostructures, including hybrid host-guest nanoconjugates, micelles, nanogels, core-shell nanoparticles, liposomes, mesoporous silica, and hollow nanoconstructions. In addition, the controlled release of guest molecules from these hybrid nanovehicles in response to various remote stimuli such as alternating magnetic field, near infrared, or ultrasound triggers is further summarized to introduce the different mechanisms of remotely triggered release behavior. Through proper chemical functionalization, the hybrid nanovehicle system can be further endowed with many new properties toward specific biomedical applications.
Collapse
Affiliation(s)
- Zibiao Li
- Institute of Materials Research and Engineering (IMRE), A*STAR, 2 Fusionopolis Way. Innovis, #08-03, Singapore, 138634, Singapore
| | - Enyi Ye
- Institute of Materials Research and Engineering (IMRE), A*STAR, 2 Fusionopolis Way. Innovis, #08-03, Singapore, 138634, Singapore
| | | | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), A*STAR, 2 Fusionopolis Way. Innovis, #08-03, Singapore, 138634, Singapore.
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore, 117576, Singapore.
- Singapore Eye Research Institute, 11 Third Hospital Avenue, Singapore, 168751, Singapore.
| |
Collapse
|
47
|
Sybachin AV, Zaborova OV, Imelbaeva KM, Samoshin VV, Migulin VA, Plamper FA, Yaroslavov AA. Effects of the electrostatic complexation between anionic pH-sensitive liposomes and star-shaped polycations on the release of the liposomal content. MENDELEEV COMMUNICATIONS 2016. [DOI: 10.1016/j.mencom.2016.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
48
|
Hopkins SL, Siewert B, Askes SHC, Veldhuizen P, Zwier R, Heger M, Bonnet S. An in vitro cell irradiation protocol for testing photopharmaceuticals and the effect of blue, green, and red light on human cancer cell lines. Photochem Photobiol Sci 2016; 15:644-53. [PMID: 27098927 PMCID: PMC5044800 DOI: 10.1039/c5pp00424a] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/30/2016] [Indexed: 12/21/2022]
Abstract
Traditionally, ultraviolet light (100-400 nm) is considered an exogenous carcinogen while visible light (400-780 nm) is deemed harmless. In this work, a LED irradiation system for in vitro photocytotoxicity testing is described. The LED irradiation system was developed for testing photopharmaceutical drugs, but was used here to determine the basal level response of human cancer cell lines to visible light of different wavelengths, without any photo(chemo)therapeutic. The effects of blue (455 nm, 10.5 mW cm(-2)), green (520 nm, 20.9 mW cm(-2)), and red light (630 nm, 34.4 mW cm(-2)) irradiation was measured for A375 (human malignant melanoma), A431 (human epidermoid carcinoma), A549 (human lung carcinoma), MCF7 (human mammary gland adenocarcinoma), MDA-MB-231 (human mammary gland adenocarcinoma), and U-87 MG (human glioblastoma-grade IV) cell lines. In response to a blue light dose of 19 J cm(-2), three cell lines exhibited a minimal (20%, MDA-MB-231) to moderate (30%, A549 and 60%, A375) reduction in cell viability, compared to dark controls. The other cell lines were not affected. Effective blue light doses that produce a therapeutic response in 50% of the cell population (ED50) compared to dark conditions were found to be 10.9 and 30.5 J cm(-2) for A375 and A549 cells, respectively. No adverse effects were observed in any of the six cell lines irradiated with a 19 J cm(-2) dose of 520 nm (green) or 630 nm (red) light. The results demonstrate that blue light irradiation can have an effect on the viability of certain human cancer cell types and controls should be used in photopharmaceutical testing, which uses high-energy (blue or violet) visible light activation.
Collapse
Affiliation(s)
- S. L. Hopkins
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| | - B. Siewert
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| | - S. H. C. Askes
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| | - P. Veldhuizen
- Leiden Institute of Physics , Leiden University , Niels Bohrweg 2 , 2333CA Leiden , The Netherlands
| | - R. Zwier
- Leiden Institute of Physics , Leiden University , Niels Bohrweg 2 , 2333CA Leiden , The Netherlands
| | - Michal Heger
- Department of Experimental Surgery , Academic Medical Center , University of Amsterdam , Meibergdreef 9 , 1105 AZ Amsterdam , The Netherlands
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry , Leiden University , Einsteinweg 55 , 2300RA Leiden , The Netherlands .
| |
Collapse
|
49
|
Miller K, Dixit S, Bredlau AL, Moore A, McKinnon E, Broome AM. Delivery of a drug cache to glioma cells overexpressing platelet-derived growth factor receptor using lipid nanocarriers. Nanomedicine (Lond) 2016; 11:581-95. [PMID: 27003178 DOI: 10.2217/nnm.15.218] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Glioblastoma multiforme is a devastating disease with no curative options due to the difficulty in achieving sufficient quantities of effective chemotherapies into the tumor past the blood-brain barrier. Micelles loaded with temozolomide (TMZ) were designed to increase the delivery of this drug into the brain. MATERIALS & METHODS pH-responsive micelles composed of distearoyl phosphoethanolamine-PEG-2000-amine and N-palmitoyl homocysteine were surface-functionalized with PDGF peptide and Dylight 680 fluorophore. RESULTS & CONCLUSION PDGF-micelles containing TMZ have specific uptake and increased killing in glial cells compared with untargeted micelles. In vivo studies demonstrated selective accumulation of PDGF-micelles containing TMZ in orthotopic gliomas implanted in mice. Targeted micelle-based drug carrier systems hold potential for delivery of a wide variety of hydrophobic drugs thereby reducing its systemic toxicity.
Collapse
Affiliation(s)
- Kayla Miller
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA
| | - Suraj Dixit
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA.,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Amy-Lee Bredlau
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alfred Moore
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA.,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Emilie McKinnon
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ann-Marie Broome
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA.,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
50
|
Lorenzato C, Oerlemans C, van Elk M, Geerts WJC, Denis de Senneville B, Moonen C, Bos C. MRI monitoring of nanocarrier accumulation and release using Gadolinium-SPIO co-labelled thermosensitive liposomes. CONTRAST MEDIA & MOLECULAR IMAGING 2016; 11:184-94. [PMID: 26750715 DOI: 10.1002/cmmi.1679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/09/2015] [Accepted: 11/19/2015] [Indexed: 12/20/2022]
Abstract
Encapsulation of anticancer drugs in triggerable nanocarriers can beneficially modify pharmacokinetics and biodistribution of chemotherapeutic drugs, and consequently increase tumor drug concentration and efficacy, while reducing side effects. Thermosensitive liposomes release their contents triggered by hyperthermia, which can be, for example, precisely delivered using an MR Imaging-guided focused ultrasound procedure. In such a scenario, it is attractive to demonstrate the accumulation of liposomes before applying hyperthermia, as well as to document the release of liposome content using MRI. To address this need, thermosensitive liposomes were developed and characterized, which were doubly loaded by iron oxide nanoparticles and Gd-chelate, as opposed to loading with a single contrast agent. When intact, the transverse relaxivity of the liposomes is high allowing detection of carriers in tissue. After heating the longitudinal relaxivity steeply increases indicating release of the small molecular contents. By choosing the appropriate MR sequences, availability and release can be evaluated without interference of one contrast agent with the other. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Cyril Lorenzato
- University Medical Center Utrecht, Department of Radiology, Imaging Division, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Chris Oerlemans
- University Medical Center Utrecht, Department of Radiology, Imaging Division, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Merel van Elk
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, 3584 CG, Utrecht, The Netherlands
| | - Willie J C Geerts
- Biomolecular Imaging, Bijvoet Center, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Baudouin Denis de Senneville
- University Medical Center Utrecht, Department of Radiology, Imaging Division, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.,Institut de Mathématiques de Bordeaux, UMR 5251 CNRS/Université de Bordeaux, F-33400, Talence, France
| | - Chrit Moonen
- University Medical Center Utrecht, Department of Radiology, Imaging Division, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Clemens Bos
- University Medical Center Utrecht, Department of Radiology, Imaging Division, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|