1
|
Romero-Ben E, Goswami U, Soto-Cruz J, Mansoori-Kermani A, Mishra D, Martin-Saldaña S, Muñoz-Ugartemendia J, Sosnik A, Calderón M, Beloqui A, Larrañaga A. Polymer-based nanocarriers to transport therapeutic biomacromolecules across the blood-brain barrier. Acta Biomater 2025; 196:17-49. [PMID: 40032217 DOI: 10.1016/j.actbio.2025.02.065] [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: 09/16/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Therapeutic biomacromolecules such as genetic material, antibodies, growth factors and enzymes represent a novel therapeutic alternative for neurological diseases and disorders. In comparison to traditional therapeutics, which are mainly based on small molecular weight drugs that address the symptoms of these disorders, therapeutic biomacromolecules can reduce undesired side effects and target specific pathological pathways, thus paving the way towards personalized medicine. However, these biomacromolecules undergo degradation/denaturation processes in the physiological environment and show poor capacity to cross the blood-brain barrier (BBB). Consequently, they rarely reach the central nervous system (CNS) in their active form. Herein, we critically overview several polymeric nanocarriers that can protect and deliver therapeutic biomacromolecules across the BBB. Polymeric nanocarriers are first categorized based on their architecture (biodegradable solid nanoparticles, nanogels, dendrimers, self-assembled nanoparticles) that ultimately determines their physico-chemical properties and function. The available polymeric formulations are then thoroughly analyzed, placing particular attention on those strategies that ensure the stability of the biomacromolecules during their encapsulation process and promote their passage across the BBB by controlling their physical (e.g., mechanical properties, size, surface charge) and chemical (e.g., surface functional groups, targeting motifs) properties. Accordingly, this review gives a unique perspective on polymeric nanocarriers for the delivery of therapeutic biomacromolecules across the BBB, representing a concise, complete and easy-to-follow guide, which will be of high interest for chemists, material scientists, pharmacologists, and biologists. Besides, it also provides a critical perspective about the limited clinical translation of these systems. STATEMENT OF SIGNIFICANCE: The increasing incidence of central nervous system disorders is a major health concern. The use of therapeutic biomacromolecules has been placed in the spotlight of many investigations. However, reaching therapeutic concentration levels of biomacromolecules in the central nervous system is restricted by the blood-brain barrier and, thus, this represents the main clinical challenge when developing efficient therapies. Herein, we provide a critical discussion about the use of polymeric nanocarriers to deliver therapeutic biomacromolecules into the central nervous system, highlighting potential future directions to overcome the current challenges.
Collapse
Affiliation(s)
- Elena Romero-Ben
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Upashi Goswami
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain
| | - Jackeline Soto-Cruz
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Amirreza Mansoori-Kermani
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain; Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy; Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo PIaggio 34, Pontedera 56025, Italy
| | - Dhiraj Mishra
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Jone Muñoz-Ugartemendia
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Ana Beloqui
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain.
| |
Collapse
|
2
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
3
|
Sood S, Matar MM, Kim J, Kinsella M, Rayavara K, Signer O, Henderson J, Rogers J, Chawla B, Narvaez B, Van Ry A, Kar S, Arnold A, Rice JS, Smith AM, Su D, Sparks J, Le Goff C, Boyer JD, Anwer K. Strong immunogenicity & protection in mice with PlaCCine: A COVID-19 DNA vaccine formulated with a functional polymer. Vaccine 2024; 42:1300-1310. [PMID: 38302336 DOI: 10.1016/j.vaccine.2024.01.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/30/2023] [Accepted: 01/20/2024] [Indexed: 02/03/2024]
Abstract
DNA- based vaccines have demonstrated the potential as a safe and effective modality. PlaCCine, a DNA-based vaccine approach described subsequently relies on a synthetic DNA delivery system and is independent of virus or device. The synthetic functionalized polymer combined with DNA demonstrated stability over 12 months at 4C and for one month at 25C. Transfection efficiency compared to naked DNA increased by 5-15-fold in murine skeletal muscle. Studies of DNA vaccines expressing spike proteins from variants D614G (pVAC15), Delta (pVAC16), or a D614G + Delta combination (pVAC17) were conducted. Mice immunized intramuscular injection (IM) with pVAC15, pVAC16 or pVAC17 formulated with functionalized polymer and adjuvant resulted in induction of spike-specific humoral and cellular responses. Antibody responses were observed after one immunization. And endpoint IgG titers increased to greater than 1x 105 two weeks after the second injection. Neutralizing antibodies as determined by a pseudovirus competition assay were observed following vaccination with pVAC15, pVAC16 or pVAC17. Spike specific T cell immune responses were also observed following vaccination and flow cytometry analysis demonstrated the cellular immune responses included both CD4 and CD8 spike specific T cells. The immune responses in vaccinated mice were maintained for up to 14 months after vaccination. In an immunization and challenge study of K18 hACE2 transgenic mice pVAC15, pVAC16 and pVAC17 induced immune responses lead to decreased lung viral loads by greater than 90 % along with improved clinical score. These findings suggest that PlaCCine DNA vaccines are effective and stable and further development against emerging SARS-CoV-2 variants is warranted.
Collapse
Affiliation(s)
| | | | - Jessica Kim
- Imunon Inc., Lawrenceville, NJ, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | - Daishui Su
- Imunon Inc., Lawrenceville, NJ, United States
| | - Jeff Sparks
- Imunon Inc., Lawrenceville, NJ, United States
| | | | | | | |
Collapse
|
4
|
Gandla K, Islam F, Zehravi M, Karunakaran A, Sharma I, Haque MA, Kumar S, Pratyush K, Dhawale SA, Nainu F, Khan SL, Islam MR, Al-Mugren KS, Siddiqui FA, Emran TB, Khandaker MU. Natural polymers as potential P-glycoprotein inhibitors: Pre-ADMET profile and computational analysis as a proof of concept to fight multidrug resistance in cancer. Heliyon 2023; 9:e19454. [PMID: 37662819 PMCID: PMC10472248 DOI: 10.1016/j.heliyon.2023.e19454] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
P-glycoprotein (P-gp) is known as the "multidrug resistance protein" because it contributes to tumor resistance to several different classes of anticancer drugs. The effectiveness of such polymers in treating cancer and delivering drugs has been shown in a wide range of in vitro and in vivo experiments. The primary objective of the present study was to investigate the inhibitory effects of several naturally occurring polymers on P-gp efflux, as it is known that P-gp inhibition can impede the elimination of medications. The objective of our study is to identify polymers that possess the potential to inhibit P-gp, a protein involved in drug resistance, with the aim of enhancing the effectiveness of anticancer drug formulations. The ADMET profile of all the selected polymers (Agarose, Alginate, Carrageenan, Cyclodextrin, Dextran, Hyaluronic acid, and Polysialic acid) has been studied, and binding affinities were investigated through a computational approach using the recently released crystal structure of P-gp with PDB ID: 7O9W. The advanced computational study was also done with the help of molecular dynamics simulation. The aim of the present study is to overcome MDR resulting from the activity of P-gp by using such polymers that can inhibit P-gp when used in formulations. The docking scores of native ligand, Agarose, Alginate, Carrageenan, Chitosan, Cyclodextrin, Dextran, Hyaluronic acid, and Polysialic acid were found to be -10.7, -8.5, -6.6, -8.7, -8.6, -24.5, -6.7, -8.3, and -7.9, respectively. It was observed that, Cyclodextrin possess multiple properties in drug delivery science and here also demonstrated excellent binding affinity. We propose that drug efflux-related MDR may be prevented by the use of Agarose, Carregeenan, Chitosan, Cyclodextrin, Hyaluronic acid, and/or Polysialic acid in the administration of anticancer drugs.
Collapse
Affiliation(s)
- Kumaraswamy Gandla
- Department of Pharmaceutical Analysis, Chaitanya (Deemed to be University), Himayath Nagar, Hyderabad 500075, Telangana, India
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University, Al-Kharj 11942, Saudi Arabia
| | - Anandakumar Karunakaran
- Department of Pharmaceutical Analysis, Vivekanandha Pharmacy College for Women, Beerachipalayam, Sankari West, Sankari, Salem, Tamil Nadu, - 637 303, India
| | - Indu Sharma
- Department of Physics, Career Point University, Hamirpur, Himachal Pradesh 176041, India
| | - M. Akiful Haque
- Department of Pharmaceutical Analysis, School of Pharmacy, Anurag University, Hyderabad, India
| | - Sanjay Kumar
- Department of Pharmacognosy, Laureate Institute of Pharmacy, VPO Kathog, Dehra, Kangra, Himachal Pradesh 176031, India
| | - Kumar Pratyush
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Sachin A. Dhawale
- Shreeyash Institute of Pharmaceutical Education and Research Aurangabad, 431 005, Maharashtra, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Sharuk L. Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa 413520, Maharashtra, India
- Department of Pharmaceutical Chemistry, School of Pharmacy, Anurag University, Hyderabad, India
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Kholoud Saad Al-Mugren
- Department of Physics, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428 Riyadh 11671, Saudi Arabia
| | - Falak A. Siddiqui
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa 413520, Maharashtra, India
- Department of Pharmaceutical Chemistry, School of Pharmacy, Anurag University, Hyderabad, India
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Selangor, Malaysia
| |
Collapse
|
5
|
Carrera Espinoza MJ, Lin KS, Weng MT, Kunene SC, Lin YS, Wu CM. Synthesis and Characterization of Supermagnetic Nanocomposites Coated with Pluronic F127 as a Contrast Agent for Biomedical Applications. Pharmaceutics 2023; 15:740. [PMID: 36986601 PMCID: PMC10053918 DOI: 10.3390/pharmaceutics15030740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Nanomedicine has garnered significant interest owing to advances in drug delivery, effectively demonstrated in the treatment of certain diseases. Here, smart supermagnetic nanocomposites based on iron oxide nanoparticles (MNPs) coated with Pluronic F127 (F127) were developed for the delivery of doxorubicin (DOX) to tumor tissues. The XRD patterns for all samples revealed peaks consistent with Fe3O4, as shown by their indices (220), (311), (400), (422), (511), and (440), demonstrating that the structure of Fe3O4 did not change after the coating process. After loading with DOX, the as-prepared smart nanocomposites demonstrated drug-loading efficiency and drug-loading capacity percentages of 45 ± 0.10 and 17 ± 0.58% for MNP-F127-2-DOX and 65 ± 0.12 and 13 ± 0.79% for MNP-F127-3-DOX, respectively. Moreover, a better DOX release rate was observed under acidic conditions, which may be credited to the pH sensitivity of the polymer. In vitro analysis demonstrated the survival rate of approximately 90% in HepG2 cells treated with PBS and MNP-F127-3 nanocomposites. Furthermore, after treatment with MNP-F127-3-DOX, the survival rate decreased, confirming cellular inhibition. Hence, the synthesized smart nanocomposites showed great promise for drug delivery in liver cancer treatment, overcoming the limitations of traditional therapies.
Collapse
Affiliation(s)
- Maria Janina Carrera Espinoza
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung–Li District, Taoyuan City 320, Taiwan
| | - Kuen-Song Lin
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung–Li District, Taoyuan City 320, Taiwan
| | - Meng-Tzu Weng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302, Taiwan
| | - Sikhumbuzo Charles Kunene
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung–Li District, Taoyuan City 320, Taiwan
| | - You-Sheng Lin
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung–Li District, Taoyuan City 320, Taiwan
| | - Chun-Ming Wu
- National Synchrotron Radiation Research Center, Hsinchu Science Park, Hsinchu 300, Taiwan
| |
Collapse
|
6
|
Verma P, Gupta GD, Markandeywar TS, Singh D. A Critical Sojourn of Polymeric Micelles: Technological Concepts, Recent Advances, and Future Prospects. Assay Drug Dev Technol 2023; 21:31-47. [PMID: 36856457 DOI: 10.1089/adt.2022.079] [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: 03/02/2023] Open
Abstract
Poorly soluble drug molecules/phytoconstituents are still a growing concern for biopharmaceutical delivery in the body. Polymeric micelles are the amphiphilic block copolymers and have been widely investigated as targeted nanocarriers for the treatment of various ailments. The versatility of nanocarriers is the self-assembling properties in the aqueous medium and forms a stable isotropic system in vivo. The hydrophobic core-hydrophilic shell configuration of the polymers used to the mixed micelles makes easy encapsulation of hydrophobic and hydrophilic drugs into the core. Polymeric micelles can also be combined with targeting ligands that increase their uptake by specific cells, decreasing off-target effects, and provide enhanced therapeutic effect. In the present review, we primarily focused on a critical appraisal of Polymeric micelles along with the method of preparation, mechanism of micelle formulation, and the ongoing formulations under clinical trials. In addition, the biological applications of this isotropic nanocarrier have been duly presented in each route of administration along with suitable case studies.
Collapse
Affiliation(s)
- Princi Verma
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | | | - Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
7
|
Influence of the Hydrophobicity of Pluronic Micelles Encapsulating Curcumin on the Membrane Permeability and Enhancement of Photoinduced Antibacterial Activity. Pharmaceutics 2022; 14:pharmaceutics14102137. [PMID: 36297572 PMCID: PMC9608470 DOI: 10.3390/pharmaceutics14102137] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Apart from its well-known activity as an antimicrobial agent, Curcumin (CURC) has recently started to arouse interest as a photosensitizer in the photodynamic therapy of bacterial infections. The aim of the present study was to evidence the influence of the encapsulation of Curcumin into polymeric micelles on the efficiency of photoinduced microbial inhibition. The influence of the hydrophobicity of the selected Pluronics (P84, P123, and F127) on the encapsulation, stability, and antimicrobial efficiency of CURC-loaded micelles was investigated. In addition, the size, morphology, and drug-loading capacity of the micellar drug delivery systems have been characterized. The influence of the presence of micellar aggregates and unassociated molecules of various Pluronics on the membrane permeability was investigated on both normal and resistant microbial strains of E. coli, S. aureus, and C. albicans. The antimicrobial efficiency on the common pathogens was assessed for CURC-loaded polymeric micelles in dark conditions and activated by blue laser light (470 nm). Significant results in the reduction of the microorganism’s growth were found in cultures of C. albicans, even at very low concentrations of surfactants and Curcumin. Unlike the membrane permeabilization effect of the monomeric solution of Pluronics, reported in the case of tumoral cells, a limited permeabilization effect was found on the studied microorganisms. Encapsulation of the Curcumin in Pluronic P84 and P123 at very low, nontoxic concentrations for photosensitizer and drug-carrier, produced CURC-loaded micelles that prove to be effective in the light-activated inhibition of resistant species of Gram-positive bacteria and fungi.
Collapse
|
8
|
Nanofiber Carriers of Therapeutic Load: Current Trends. Int J Mol Sci 2022; 23:ijms23158581. [PMID: 35955712 PMCID: PMC9368923 DOI: 10.3390/ijms23158581] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
The fast advancement in nanotechnology has prompted the improvement of numerous methods for the creation of various nanoscale composites of which nanofibers have gotten extensive consideration. Nanofibers are polymeric/composite fibers which have a nanoscale diameter. They vary in porous structure and have an extensive area. Material choice is of crucial importance for the assembly of nanofibers and their function as efficient drug and biomedicine carriers. A broad scope of active pharmaceutical ingredients can be incorporated within the nanofibers or bound to their surface. The ability to deliver small molecular drugs such as antibiotics or anticancer medications, proteins, peptides, cells, DNA and RNAs has led to the biomedical application in disease therapy and tissue engineering. Although nanofibers have shown incredible potential for drug and biomedicine applications, there are still difficulties which should be resolved before they can be utilized in clinical practice. This review intends to give an outline of the recent advances in nanofibers, contemplating the preparation methods, the therapeutic loading and release and the various therapeutic applications.
Collapse
|
9
|
Singh P, Singh N, Mishra N, Nisha R, Alka, Maurya P, Pal RR, Singh S, Saraf SA. Functionalized Bosutinib Liposomes for Target Specific Delivery in management of Estrogen-Positive Cancer. Colloids Surf B Biointerfaces 2022; 218:112763. [DOI: 10.1016/j.colsurfb.2022.112763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022]
|
10
|
Akib AA, Shakil R, Rumon MMH, Roy CK, Chowdhury EH, Chowdhury AN. Natural and Synthetic Micelles for Delivery of Small Molecule Drugs, Imaging Agents and Nucleic Acids. Curr Pharm Des 2022; 28:1389-1405. [PMID: 35524674 DOI: 10.2174/1381612828666220506135301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
The poor solubility, lack of targetability, quick renal clearance, and degradability of many therapeutic and imaging agents strongly limit their applications inside the human body. Amphiphilic copolymers having self-assembling properties can form core-shell structures called micelles, a promising nanocarrier for hydrophobic drugs, plasmid DNA, oligonucleotides, small interfering RNAs (siRNAs) and imaging agents. Fabrication of micelles loaded with different pharmaceutical agents provides numerous advantages including therapeutic efficacy, diagnostic sensitivity, and controlled release to the desired tissues. Moreover, due to their smaller particle size (10-100 nm) and modified surfaces with different functional groups (such as ligands) help them to accumulate easily in the target location, enhancing cellular uptake and reducing unwanted side effects. Furthermore, the release of the encapsulated agents may also be triggered from stimuli-sensitive micelles at different physiological conditions or by an external stimulus. In this review article, we discuss the recent advancement in formulating and targeting different natural and synthetic micelles including block copolymer micelles, cationic micelles, and dendrimers-, polysaccharide- and protein-based micelles for the delivery of different therapeutic and diagnostic agents. Finally, their applications, outcomes, and future perspectives have been summarized.
Collapse
Affiliation(s)
- Anwarul Azim Akib
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Ragib Shakil
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Md Mahamudul Hasan Rumon
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Chanchal Kumar Roy
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Malaysia
| | - Al-Nakib Chowdhury
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| |
Collapse
|
11
|
|
12
|
Cao J, Bhatnagar S, Wang J, Qi X, Prabha S, Panyam J. Cancer stem cells and strategies for targeted drug delivery. Drug Deliv Transl Res 2021; 11:1779-1805. [PMID: 33095384 PMCID: PMC8062588 DOI: 10.1007/s13346-020-00863-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 12/23/2022]
Abstract
Cancer stem cells (CSCs) are a small proportion of cancer cells with high tumorigenic activity, self-renewal ability, and multilineage differentiation potential. Standard anti-tumor therapies including conventional chemotherapy, radiation therapy, and molecularly targeted therapies are not effective against CSCs, and often lead to enrichment of CSCs that can result in tumor relapse. Therefore, it is hypothesized that targeting CSCs is key to increasing the efficacy of cancer therapies. In this review, CSC properties including CSC markers, their role in tumor growth, invasiveness, metastasis, and drug resistance, as well as CSC microenvironment are discussed. Further, CSC-targeted strategies including the use of targeted drug delivery systems are examined.
Collapse
Affiliation(s)
- Jin Cao
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Shubhmita Bhatnagar
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Jiawei Wang
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- College of Pharmacy, University of Texas at Austin, Austin, TX, 78712, USA
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Swayam Prabha
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- Cancer Research & Molecular Biology and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jayanth Panyam
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA.
- School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
13
|
Ganguly R, Kumar S, Nath S, Basu M, Aswal VK. Unusual Growth and Hydration Characteristics of Oil Solubilized Micelles in Aqueous Pluronic Systems. J Phys Chem B 2021; 125:10578-10588. [PMID: 34495673 DOI: 10.1021/acs.jpcb.1c04450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lipophile induced modulations of self-assembly characteristics in aqueous Pluronic systems merit attention because of wide-ranging uses of Pluronics as solubilizing agents of lipophilic substances. In this paper, we report unusual evolutions of structural and hydration properties in lavender essential oil (LO) solubilized Pluronic P85 aqueous micellar systems as a function of micellar volume fraction and temperature. Our DLS, SANS, and viscometry studies show that the spherical-to-wormlike micellar structural transition observed in 1% P85 solutions upon solubilization of LO quite unexpectedly gets suppressed with increased P85 concentration to ≥5%. Detailed SANS studies reveal that the core sizes of the oil solubilized micelles cannot attain the threshold value required for the onset of structural transition at higher copolymer concentrations due to their progressive shrinking with an increase in P85 concentration. Oil solubilized P85 solutions show two cloud points and very interestingly exhibit micellar growth upon cooling to their lower cloud points. Steady state fluorescence studies explain this based on increasing dehydration of micellar corona with a decrease in temperature, very much opposite to what is observed in pure aqueous Pluronic systems. The results give new insight into viscous flow properties and low temperature storage possibilities of oil solubilized aqueous Pluronic systems.
Collapse
Affiliation(s)
- R Ganguly
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - S Kumar
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - S Nath
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - M Basu
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - V K Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| |
Collapse
|
14
|
Micellar solubilization of Lavender oil in aqueous P85/P123 systems: Investigating the associated micellar structural transitions, therapeutic properties and existence of double cloud points. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
15
|
Shaikh SJ, Patel HS, Ray D, Aswal VK, Singh S, Vijayvargia R, Sheth U, Sharma RK. Enhanced Solubility and Oral Bioavailability of Hydrophobic Drugs Using Pluronic Nanomicelles: An In‐Vitro Evaluation. ChemistrySelect 2021. [DOI: 10.1002/slct.202102123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sofiya J. Shaikh
- Applied Chemistry Department Faculty of Technology and Engineering The Maharaja Sayajirao University of Baroda Vadodara 390001 Gujarat India
| | - Hemil S. Patel
- Applied Chemistry Department Faculty of Technology and Engineering The Maharaja Sayajirao University of Baroda Vadodara 390001 Gujarat India
| | - Debes Ray
- Solid State Physics Division Bhabha Atomic Research Centre Mumbai Maharashtra India
| | - Vinod K. Aswal
- Solid State Physics Division Bhabha Atomic Research Centre Mumbai Maharashtra India
| | - Sushmita Singh
- Department of Biochemistry Faculty of Science The Maharaja Sayajirao University of Baroda Vadodara 390001 Gujarat India
| | - Ravi Vijayvargia
- Department of Biochemistry Faculty of Science The Maharaja Sayajirao University of Baroda Vadodara 390001 Gujarat India
| | - Urjita Sheth
- C. G. Bhakta Institute of Biotechnology UkaTarsadia University Bardoli Gujarat India
| | - Rakesh K. Sharma
- Applied Chemistry Department Faculty of Technology and Engineering The Maharaja Sayajirao University of Baroda Vadodara 390001 Gujarat India
| |
Collapse
|
16
|
Yu J, Qiu H, Yin S, Wang H, Li Y. Polymeric Drug Delivery System Based on Pluronics for Cancer Treatment. Molecules 2021; 26:3610. [PMID: 34204668 PMCID: PMC8231161 DOI: 10.3390/molecules26123610] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Pluronic polymers (pluronics) are a unique class of synthetic triblock copolymers containing hydrophobic polypropylene oxide (PPO) and hydrophilic polyethylene oxide (PEO) arranged in the PEO-PPO-PEO manner. Due to their excellent biocompatibility and amphiphilic properties, pluronics are an ideal and promising biological material, which is widely used in drug delivery, disease diagnosis, and treatment, among other applications. Through self-assembly or in combination with other materials, pluronics can form nano carriers with different morphologies, representing a kind of multifunctional pharmaceutical excipients. In recent years, the utilization of pluronic-based multi-functional drug carriers in tumor treatment has become widespread, and various responsive drug carriers are designed according to the characteristics of the tumor microenvironment, resulting in major progress in tumor therapy. This review introduces the specific role of pluronic-based polymer drug delivery systems in tumor therapy, focusing on their physical and chemical properties as well as the design aspects of pluronic polymers. Finally, using newer literature reports, this review provides insights into the future potential and challenges posed by different pluronic-based polymer drug delivery systems in tumor therapy.
Collapse
Affiliation(s)
- Jialin Yu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (J.Y.); (H.Q.); (S.Y.)
| | - Huayu Qiu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (J.Y.); (H.Q.); (S.Y.)
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, China
| | - Shouchun Yin
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (J.Y.); (H.Q.); (S.Y.)
| | - Hebin Wang
- College of Chemical Engineering and Technology, Tianshui Normal University, Tianshui 741099, China
| | - Yang Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (J.Y.); (H.Q.); (S.Y.)
| |
Collapse
|
17
|
Nasongkla N, Tuchinda P, Munyoo B, Eawsakul K. Preparation and Characterization of MUC-30-Loaded Polymeric Micelles against MCF-7 Cell Lines Using Molecular Docking Methods and In Vitro Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5597681. [PMID: 34135981 PMCID: PMC8179782 DOI: 10.1155/2021/5597681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
MUC-30 is a hydrophobic compound which is active against the MCF-7 cancer cell line. In this study, MUC-30 was loaded in polymeric micelles to improve the water solubility and release rate. For prolonged MUC-30 release, MUC-30 was encapsulated in polymeric micelles using PEG-b-PLA and PEG-b-PCL as materials. Micelles prepared with 1 : 9 w per w ratios by film hydration achieved the highest entrapment efficiency (EE%). The EE% of MUC-30-loaded PEG-b-PCL micelles was approximately 30% greater than that of PEG-b-PLA micelles, due to the different H-bond formations between MUC-30 and the polymer membrane (PCL, 4; PLA, 3). The cytotoxic activity of MUC-30 against EGFR theoretically presented 399.31 nM (IC50 = 282.26 ng/mL) by molecular docking. In vitro cytotoxic activity of MUC-30 was confirmed by MTT assay. MUC-30 (IC50 = 11 ± 0.39 ng/mL) showed three-fold higher activity over MUC-30-loaded PEG-b-PLA micelles (IC50 = 37 ± 1.18 ng/mL) and two-fold higher over PEG-b-PCL micelles (IC50 = 75 ± 3.97 ng/mL). This was due to the higher release rate of MUC-30 from PEG-b-PLA micelles compared to PEG-b-PCL micelles. Therefore, MUC-30-loaded PEG-b-PLA micelles could be a promising candidate for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Norased Nasongkla
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom 73170, Thailand
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Mahidol University, Nakhon Bangkok 10400, Thailand
| | - Patoomratana Tuchinda
- Excellence Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Bamroong Munyoo
- Department of Chemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Komgrit Eawsakul
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom 73170, Thailand
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- School of Allied Health Sciences and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
18
|
Wiwatchaitawee K, Quarterman JC, Geary SM, Salem AK. Enhancement of Therapies for Glioblastoma (GBM) Using Nanoparticle-based Delivery Systems. AAPS PharmSciTech 2021; 22:71. [PMID: 33575970 DOI: 10.1208/s12249-021-01928-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of malignant brain tumor. Current FDA-approved treatments include surgical resection, radiation, and chemotherapy, while hyperthermia, immunotherapy, and most relevantly, nanoparticle (NP)-mediated delivery systems or combinations thereof have shown promise in preclinical studies. Drug-carrying NPs are a promising approach to brain delivery as a result of their potential to facilitate the crossing of the blood-brain barrier (BBB) via two main types of transcytosis mechanisms: adsorptive-mediated transcytosis (AMT) and receptor-mediated transcytosis (RMT). Their ability to accumulate in the brain can thus provide local sustained release of tumoricidal drugs at or near the site of GBM tumors. NP-based drug delivery has the potential to significantly reduce drug-related toxicity, increase specificity, and consequently improve the lifespan and quality of life of patients with GBM. Due to significant advances in the understanding of the molecular etiology and pathology of GBM, the efficacy of drugs loaded into vectors targeting this disease has increased in both preclinical and clinical settings. Multitargeting NPs, such as those incorporating multiple specific targeting ligands, are an innovative technology that can lead to decreased off-target effects while simultaneously having increased accumulation and action specifically at the tumor site. Targeting ligands can include antibodies, or fragments thereof, and peptides or small molecules, which can result in a more controlled drug delivery system compared to conventional drug treatments. This review focuses on GBM treatment strategies, summarizing current options and providing a detailed account of preclinical findings with prospective NP-based approaches aimed at improving tumor targeting and enhancing therapeutic outcomes for GBM patients.
Collapse
|
19
|
Mahdi WA, Hussain A, Ramzan M, Faruk A, Bukhari SI, Dev A. Pluronic-Coated Biogenic Gold Nanoparticles for Colon Delivery of 5-Fluorouracil: In vitro and Ex vivo Studies. AAPS PharmSciTech 2021; 22:64. [PMID: 33533992 DOI: 10.1208/s12249-021-01922-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
The aim of the study was to prepare 5-fluorouracil (5-FU)-loaded biogenic gold nanoparticles with pluronic-based coating (PFGNPs), their optimization (full factorial predicted OBPN-1) and in vitro-ex vivo evaluation. Several formulations were prepared, selected for optimization using Design Expert®, and compared for morphology, 5-FU release kinetics, compatibility, cell line toxicity, in vitro hemocompatibility, and ex vivo intestinal permeation across the rat duodenum, jejunum, and ileum. The pluronic-coated 5-FU-carrying GNPs were spherical, 29.11-178.21 nm in diameter, with a polydispersity index (PDI) range of 0.191-292, and a zeta potential (ZP) range of 11.19-29.21 (-mV). The optimized OBPN-1 (desirability = 0.95) demonstrated optimum size (175.1 nm), %DL as 73.8%, ZP as 21.7 mV, % drug release (DR) as 75.7%, and greater cytotoxicity (viability ~ 8.9%) against the colon cancer cell lines than 5-FU solution (~ 24.91%), and less hemocompatibility. Moreover, OBPN-1 exhibited 4.5-fold permeation across the rat jejunum compared with 5-FU solution. Thus, the PFGNPs exhibit high DL capacity, sustained delivery, hemocompatibility, improved efficacy, and enhanced permeation profiles compared with 5-FU solution and several other NPs preparations suggesting it is a promising formulation for effective colon cancer control with reduced side effects.
Collapse
|
20
|
Gregoriou Y, Gregoriou G, Yilmaz V, Kapnisis K, Prokopi M, Anayiotos A, Strati K, Dietis N, Constantinou AI, Andreou C. Resveratrol loaded polymeric micelles for theranostic targeting of breast cancer cells. Nanotheranostics 2021; 5:113-124. [PMID: 33391978 PMCID: PMC7738945 DOI: 10.7150/ntno.51955] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
Treatment of breast cancer underwent extensive progress in recent years with molecularly targeted therapies. However, non-specific pharmaceutical approaches (chemotherapy) persist, inducing severe side-effects. Phytochemicals provide a promising alternative for breast cancer prevention and treatment. Specifically, resveratrol (res) is a plant-derived polyphenolic phytoalexin with potent biological activity but displays poor water solubility, limiting its clinical use. Here we have developed a strategy for delivering res using a newly synthesized nano-carrier with the potential for both diagnosis and treatment. Methods: Res-loaded nanoparticles were synthesized by the emulsion method using Pluronic F127 block copolymer and Vitamin E-TPGS. Nanoparticle characterization was performed by SEM and tunable resistive pulse sensing. Encapsulation Efficiency (EE%) and Drug Loading (DL%) content were determined by analysis of the supernatant during synthesis. Nanoparticle uptake kinetics in breast cancer cell lines MCF-7 and MDA-MB-231 as well as in MCF-10A breast epithelial cells were evaluated by flow cytometry and the effects of res on cell viability via MTT assay. Results: Res-loaded nanoparticles with spherical shape and a dominant size of 179±22 nm were produced. Res was loaded with high EE of 73±0.9% and DL content of 6.2±0.1%. Flow cytometry revealed higher uptake efficiency in breast cancer cells compared to the control. An MTT assay showed that res-loaded nanoparticles reduced the viability of breast cancer cells with no effect on the control cells. Conclusions: These results demonstrate that the newly synthesized nanoparticle is a good model for the encapsulation of hydrophobic drugs. Additionally, the nanoparticle delivers a natural compound and is highly effective and selective against breast cancer cells rendering this type of nanoparticle an excellent candidate for diagnosis and therapy of difficult to treat mammary malignancies.
Collapse
Affiliation(s)
- Yiota Gregoriou
- Department of Biological Sciences, Faculty of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus.,Department of Electrical and Computer Engineering University of Cyprus, Nicosia, Cyprus.,Emphasis Research Centre, University of Cyprus, Nicosia, Cyprus
| | - Gregoria Gregoriou
- Department of Biological Sciences, Faculty of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | - Vural Yilmaz
- Department of Biological Sciences, Faculty of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | - Konstantinos Kapnisis
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limassol, Cyprus
| | - Marianna Prokopi
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limassol, Cyprus
| | - Andreas Anayiotos
- Department of Mechanical Engineering and Material Science and Engineering, Cyprus University of Technology, Limassol, Cyprus
| | - Katerina Strati
- Department of Biological Sciences, Faculty of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | | | - Andreas I Constantinou
- Department of Biological Sciences, Faculty of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chrysafis Andreou
- Department of Electrical and Computer Engineering University of Cyprus, Nicosia, Cyprus.,Emphasis Research Centre, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
21
|
Samith VD, Navarro S, Dabirian R. Morphological and Semi-empirical Study of the Pluronic F68/Imogolite/Sudan III Intersurfaces Composite for the Controlled Temperature Release of Hydrophobic Drugs. ACS OMEGA 2020; 5:20707-20723. [PMID: 32875204 PMCID: PMC7450501 DOI: 10.1021/acsomega.9b02965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 03/03/2020] [Indexed: 06/11/2023]
Abstract
Some PluronicF68 (F68) triblock copolymer properties demonstrate surprising applications in selective drug administration, such as the transportation of hydrophobic anti-inflammatories through epithelial barriers. Nuclear magnetic resonance (1H-NMR) spectroscopy was carried out for micelle precursor dispersions and F68 films modified with a synthetic imogolite (IMO) biocompatible hydrogel. Theoretical calculations and morphological assessment for the process of morphogenesis of dendritic crystallization were performed by molecular docking and atomic force microscopy (AFM) of the Sudan III-IMO-F68 composite, which was more hydrophobic than Sudan III-F68 and carried out the prolonged release of the Sudan III "drug" captured by a water-octanol interface determined by standard absorbance. Surface fusions were measured and compared to the unmodified matrix. However, despite the superior properties of the composite, the critical micelle concentration (CMC) was practically unmodified because solitary IMO strands attached to Sudan III formed Sudan III-IMO. These strands unraveled in a stable manner by expanding like a "spiderweb" in hydrophilic interfaces according to NMR analysis of the hydrogen one H1 polarization of Sudan III and F68 methyl, whose correlation relates hydrophobicity of Sudan III-IMO-F68 with dendrite properties from F68 concentrations. CMC and surface fusions equivalent to F68 surface properties, calculated by differential scanning calorimetry and dynamic Raman spectroscopy, were determined by AFM and high-resolution ellipsometry. Our results show highly specialized pharmacological applications since micelle surfaces expand, triggering maximum deliveries of "Drugs" from its interior to the physiological environment. The implanted sensor prototype determined equilibria reached Sudan III according to temperature (32-50 °C) and time it took to cross the membrane model 1-octanol (48 h). The findings suggest that the targested design of a F68-IMO-"Drug" would function as a microdevice for the prolonged release of hydrophobic drugs. In addition, the said microdevice could regenerate the damaged tissue in the central nervous system or other organs of the body. This is due to the fact that it could perform both tasks simultaneously, given the properties and characteristics acquired by the compatible material depending on the temperature of the physiological environment.
Collapse
Affiliation(s)
- Vicente D. Samith
- Departamento de
Química, Facultad de Ciencias, Universidad
de Chile, Las Palmeras 3425, Santiago 244355, Chile
- Institute for Medical
and Biological Engineering, Schools of Engineering, Biological Sciences
and Medicine, Pontificia Universidad Católica
de Chile, Santiago 7800003, Chile
| | - Sebastián Navarro
- Departamento de
Química, Facultad de Ciencias, Universidad
de Chile, Las Palmeras 3425, Santiago 244355, Chile
| | - Reza Dabirian
- Departamento de
Química, Facultad de Ciencias, Universidad
de Chile, Las Palmeras 3425, Santiago 244355, Chile
- Istituto per la Sintesi
Organica e la Fotoreattività, Consiglio
Nazionale delle Ricerche, Via Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
22
|
Mishra MK, Gupta J, Gupta R. Self-Assemble Amphiphilic PEO-PPO-PEO Tri-Block Co-Polymeric Methotrexate Nanomicelles to Combat MCF7 Cancer Cells. Curr Drug Deliv 2020; 18:794-804. [PMID: 32778025 DOI: 10.2174/1567201817666200810110914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/15/2020] [Accepted: 06/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Methotrexate (MTX) is a water-insoluble, anti-tumor agent that causes adverse effects like bone marrow suppression, chronic interstitial obstructive pulmonary disease, hepatotoxicity, leukopenia, interstitial pneumonitis and nephrotoxicity with slow drug release rate. OBJECTIVE The present study aimed to successfully incorporate MTX into novel-targeted Pluronic (PEOPPO- PEO tri-block co-polymer) F127 polymeric micelles intended for intravenous administration with improved drug loading and sustained release behavior necessary to achieve better efficacy of MTX. METHODS MTX-loaded Pluronic F127 micelles were characterized for critical micelle concentration, particle size and zeta potential,1H NMR, drug loading, encapsulation efficiency characterization, cell uptake, in vitro release study along with partition coefficient and solubilization thermodynamics. RESULTS The micellar formulation resulted in nano size 27.32±1.43nm of PF127/SDS, as compared to Pluronic F127 micelles or PF127/Phosphatidyl choline which were 30.52±1.18nm and 154.35±5.5nm in size, respectively. The uptake of PF127/SDS micellar formulation incorporating Rhodamine 123 in MCF7 cancer cells was found to be higher (84.25%) than PF127/PC, PF127 and MTX i.e. 66.26%, 73.59% and 53% respectively. The in vitro MTX release from PF127, PF127/SDS and PF127/PC polymeric micelles formulations was observed to be 69%, 69.5% and 66% at 12 h whereas 80.89%, 77.67% and 78.54% after 24 h, respectively and revealed a sustained release. MTX-loaded PF127/SDS micelles showed high partition coefficient and negative free energy of solubilization compared to PF127 and PF127/PC which signify self-assembly behavior and thermodynamic stability towards higher dissociation. CONCLUSION It was finally concluded that MTX-loaded PF127/SDS micelles act as a potential anticancer delivery system in comparison to PF127/PC and PF127 to combat tumor cells by enhancing their cellular uptake targeting with sustained release pattern and reducing the thermodynamic instability. Thus, PF127/SDS micellar formulation can provide a useful alternative dosage form for intravenous administration of MTX.
Collapse
Affiliation(s)
- Manoj Kumar Mishra
- Shambhunath Institute of Pharmacy, Jhalwa, Prayagraj, Uttar Pradesh, India
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Reena Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
23
|
Targeting anticancer drugs with pluronic aggregates: Recent updates. Int J Pharm 2020; 586:119544. [DOI: 10.1016/j.ijpharm.2020.119544] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022]
|
24
|
Eradication of cancer stem cells in triple negative breast cancer using doxorubicin/pluronic polymeric micelles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 24:102124. [DOI: 10.1016/j.nano.2019.102124] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 11/15/2022]
|
25
|
Wang J, Li N, Cao L, Gao C, Zhang Y, Shuai Q, Xie J, Luo K, Yang J, Gu Z. DOX-loaded peptide dendritic copolymer nanoparticles for combating multidrug resistance by regulating the lysosomal pathway of apoptosis in breast cancer cells. J Mater Chem B 2020; 8:1157-1170. [PMID: 31951231 DOI: 10.1039/c9tb02130b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Multidrug resistance (MDR) is a common phenomenon in clinical oncology and is a major obstacle to cancer chemotherapy. Many nanoparticle (NP)-based drug delivery systems have been developed to overcome MDR depending on increasing intracellular drug concentrations via increased cellular uptake and rapid drug release. The objective of this work was to investigate the performance and possible mechanisms of enzyme-sensitive mPEGylated dendron-GFLG-DOX conjugate based nanoparticles for blockading the MDR phenotype of MCF-7/ADR. In vitro, mPEGylated dendron-GFLG-DOX conjugate based nanoparticles could significantly promote cellular uptake and accumulation, potent cytotoxicity and apoptosis compared to free DOX in resistant cells. mPEGylated dendron-GFLG-DOX conjugate based nanoparticles were found to translocate across the membranes of resistant cells via active endocytic pathways leading to more DOX accumulating in the nuclei of MCF-7/ADR cells. Importantly, we found that mPEGylated dendron-GFLG-DOX conjugate based nanoparticles could induce cathepsin B in the cytoplasm and enhance lysosomal-mediated cell death compared to free DOX. Furthermore, mPEGylated dendron-GFLG-DOX conjugate based nanoparticles enhanced the drug's penetration, toxicity, and growth inhibition compared to free DOX in the three-dimensional multicellular tumor spheroid model. In vivo, mPEGylated dendron-GFLG-DOX conjugate based nanoparticles significantly improved the therapeutic efficacy against MDR xenograft tumors, and showed better biocompatibility than free DOX. These results indicated that mPEGylated dendron-GFLG-DOX conjugate based nanoparticles could be used as an alternative drug delivery system for MDR tumor treatment through initiating the lysosomal apoptosis pathway.
Collapse
Affiliation(s)
- Jianxi Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Ning Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China. and School of Pharmacy, Fujian Medical University, Fuzhou 350122, P. R. China
| | - Lei Cao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Chao Gao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Yan Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Qizhi Shuai
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Jinghui Xie
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, P. R. China.
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China. and College of Materials Science and Engineering, Nanjing Tech University, Nanjing 210009, P. R. China
| |
Collapse
|
26
|
De Clercq K, Xie F, De Wever O, Descamps B, Hoorens A, Vermeulen A, Ceelen W, Vervaet C. Preclinical evaluation of local prolonged release of paclitaxel from gelatin microspheres for the prevention of recurrence of peritoneal carcinomatosis in advanced ovarian cancer. Sci Rep 2019; 9:14881. [PMID: 31619730 PMCID: PMC6795903 DOI: 10.1038/s41598-019-51419-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 09/25/2019] [Indexed: 01/12/2023] Open
Abstract
Patients with advanced ovarian cancer develop recurrence despite initial treatment response to standard treatment of surgery and intravenous/intraperitoneal (IP) chemotherapy, partly due to a limited peritoneal exposure time of chemotherapeutics. Paclitaxel-loaded genipin-crosslinked gelatin microspheres (PTX-GP-MS) are evaluated for the treatment of microscopic peritoneal carcinomatosis and prevention of recurrent disease. The highest drug load (39.2 µg PTX/mg MS) was obtained by immersion of GP-MS in aqueous PTX nanosuspension (PTXnano-GP-MS) instead of ethanolic PTX solution (PTXEtOH-GP-MS). PTX release from PTX-GP-MS was prolonged. PTXnano-GP-MS displayed a more controlled release compared to a biphasic release from PTXEtOH-GP-MS. Anticancer efficacy of IP PTX-GP-MS (PTXEtOH-GP-MS, D = 7.5 mg PTX/kg; PTXnano-GP-MS D = 7.5 and 35 mg PTX/kg), IP nanoparticular albumin-bound PTX (D = 35 mg PTX/kg) and controls (0.9% NaCl, blank GP-MS) was evaluated in a microscopic peritoneal carcinomatosis xenograft mouse model. PTXnano-GP-MS showed superior anticancer efficacy with significant increased survival time, decreased peritoneal carcinomatosis index score and ascites incidence. However, prolonged PTX release over 14 days from PTXnano-GP-MS caused drug-related toxicity in 27% of high-dosed PTXnano-GP-MS-treated mice. Dose simulations for PTXnano-GP-MS demonstrated an optimal survival without drug-induced toxicity in a range of 7.5-15 mg PTX/kg. Low-dosed PTXnano-GP-MS can be a promising IP drug delivery system to prevent recurrent ovarian cancer.
Collapse
Affiliation(s)
- Kaat De Clercq
- Laboratory of Pharmaceutical Technology, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Feifan Xie
- Laboratory for Medical Biochemistry and Clinical Analysis, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Benedicte Descamps
- Infinity (IBiTech-MEDISIP), Department of Electronics and Information Systems, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - An Vermeulen
- Department of Gastro-intestinal Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Wim Ceelen
- Department of Gastro-intestinal Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Chris Vervaet
- Laboratory of Pharmaceutical Technology, Ghent University, Ottergemsesteenweg 460, 9000, Ghent, Belgium.
| |
Collapse
|
27
|
Katrajkar K, Darji L, Kethavath D, Thakkar S, Kshirsagar B, Misra M. Shedding light on interaction of so called inactive ingredients (excipients) with permeability-glycoprotein. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Hu H, Petrosyan A, Osna NA, Liu T, Olou AA, Alakhova DY, Singh PK, Kabanov AV, Faber EA, Bronich TK. Pluronic block copolymers enhance the anti-myeloma activity of proteasome inhibitors. J Control Release 2019; 306:149-164. [PMID: 31121280 PMCID: PMC6822276 DOI: 10.1016/j.jconrel.2019.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/30/2019] [Accepted: 05/15/2019] [Indexed: 02/08/2023]
Abstract
Proteasome inhibitors (PIs) have markedly improved response rates as well as the survival of multiple myeloma (MM) patients over the past decade and have become an important foundation in the treatment of MM patients. Unfortunately, the majority of patients either relapses or becomes refractory to proteasome inhibition. This report describes that both PI sensitive and resistant MM cells display enhanced sensitivity to PI in the presence of synthetic amphiphilic block copolymers, Pluronics (SP1017). SP1017 effectively overcomes both acquired resistance and tumor microenvironment-mediated resistance to PIs. The combination of bortezomib and SP1017 augments accumulation of ubiquitinated proteins, increases markers of proteotoxic and ER stress, and ultimately induces both the intrinsic and extrinsic drug-induced apoptotic pathways in MM cells. Notably, co-treatment of bortezomib and SP1017 intensifies SP1017-induced disorganization of the Golgi complex and significantly reduces secretion of paraproteins. Using a human MM/SCID mice model, the combination of bortezomib and SP1017 exerted enhanced antitumor efficacy as compared to bortezomib alone, delaying disease progression, but without additional toxicity. Collectively, these findings provide proof of concept for the utility of combining PI with SP1017 and present a new approach to enhance the efficacy of current treatment options for MM patients.
Collapse
Affiliation(s)
- Hangting Hu
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Armen Petrosyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Natalia A Osna
- Liver Study Unit, VA Medical Center, Research Service (151), 4101 Woolworth Avenue, Omaha, NE 68105, United States of America
| | - Tong Liu
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Appolinaire A Olou
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Daria Y Alakhova
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina 27599, United States of America
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Alexander V Kabanov
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina 27599, United States of America; Carolina Institute for Nanomedicine, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599, United States of America
| | - Edward A Faber
- Department of Internal Medicine, Division of Hematology-Oncology, University of Nebraska Medical Center, Omaha, NE 68198, United States of America
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, United States of America.
| |
Collapse
|
29
|
Singh A, Thakur S, Sharma T, Kaur M, Sahajpal NS, Aurora R, Jain SK. Harmonious Biomaterials for Development of In situ Approaches for Locoregional Delivery of Anti-cancer Drugs: Current Trends. Curr Med Chem 2019; 27:3463-3498. [PMID: 31223077 DOI: 10.2174/1573406415666190621095726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 11/22/2022]
Abstract
Locoregional drug delivery is a novel approach for the effective delivery of anti-cancer agents as it exposes the tumors to high concentration of drugs. In situ gelling systems have fetched paramount attention in the field of localized cancer chemotherapy due to their targeted delivery, ease of preparation, prolonged or sustained drug release and improved patient compliance. Numerous polymers have been investigated for their properties like swelling along with biodegradation, drug release and physicochemical properties for successful targeting of the drugs at the site of implantation. The polymers such as chitosan, Hyaluronic Acid (HA), poloxamer, Poly Glycolic Lactic Acid (PGLA) and Poly Lactic Acid (PLA) tend to form in situ hydrogels and have been exploited to develop localized delivery vehicles. These formulations are administered in the solution form and on exposure to physiological environment such as temperature, pH or ionic composition they undergo phase conversion into a hydrogel drug depot. The use of in situ gelling approach has provided prospects to increase overall survival and life quality of cancer patient by enhancing the bioavailability of drug to the site of tumor by minimizing the exposure to normal cells and alleviating systemic side effects. Because of its favorable safety profile and clinical benefits, United States Food and Drug Administration (U.S. FDA) has approved polymer based in situ systems for prolonged locoregional activity. This article discusses the rationale for developing in situ systems for targeted delivery of anti-cancer agents with special emphasis on types of polymers used to formulate the in situ system. In situ formulations for locoregional anti-cancer drug delivery that are marketed and are under clinical trials have also been discussed in detail in this article.
Collapse
Affiliation(s)
- Amrinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Tushit Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Manjot Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Nikhil Shri Sahajpal
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Rohan Aurora
- The International School Bangalore, Karnataka, India
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
30
|
khan Z, Minhas MU, Ahmad M, Khan KU, Sohail M, Khalid I. Functionalized pectin hydrogels by cross-linking with monomer: synthesis, characterization, drug release and pectinase degradation studies. Polym Bull (Berl) 2019. [DOI: 10.1007/s00289-019-02745-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
31
|
Pollard J, Rajabi-Siahboomi A, Badhan RKS, Mohammed AR, Perrie Y. High-throughput screening of excipients with a biological effect: a kinetic study on the effects of surfactants on efflux-mediated transport. J Pharm Pharmacol 2019; 71:889-897. [DOI: 10.1111/jphp.13072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/12/2019] [Indexed: 11/28/2022]
Abstract
Abstract
Objective
In this study, we develop and apply a high-throughput screening protocol to investigate the activity of non-ionic surfactants, with a broad range of hydrophilic–lipophilic balance values, against ABCB1-mediated efflux transport and ABCC2-mediated efflux transport.
Methods
Caco-2 cells were grown for 7 days in 96-well plates, then washed and incubated with the test materials for 2 h in the presence of 2.5 μm of either rhodamine 123 (R-123) or 5(6)-Carboxy-2′,7′ dichlorofluorescein diacetate as probes of ABCB1 and ABCC2, respectively.
Key findings
Of the surfactants tested, no activity against ABCC2 was detected and all surfactants showing efficacy against ABCB1 had a HLB value of 22 or below. Inhibition of ABCB1 was seen in the order of efficacy to be poloxamer 335 > poloxamer 40 > Crovol A-70 > Myrj S-40 > poloxamer 184 > poloxamer 182 > Etocas 40 > Tween 20 > Etocas 29 > Tween 80 > Acconon C-44 > Span 20. With regard to this inhibition, the distribution of hydrophilic regions is more important than the HLB value.
Conclusion
This work demonstrates a high-throughput protocol for detecting materials that can modulate ABCB1-mediated efflux. These surfactants could be exploited to improve oral delivery of drugs prone to efflux.
Collapse
Affiliation(s)
- John Pollard
- Aston Pharmacy School, School of Life and Health Sciences, Aston University, Birmingham, UK
| | | | - Raj K S Badhan
- Aston Pharmacy School, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Afzal R Mohammed
- Aston Pharmacy School, School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
32
|
Molecular Targets of the Hydrophobic Block of Pluronics in Cells: a Photo Affinity Labelling Approach. Pharm Res 2018; 35:205. [DOI: 10.1007/s11095-018-2484-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/21/2018] [Indexed: 12/31/2022]
|
33
|
Cellular fluorescein hyperfluorescence is dynamin-dependent and increased by Tetronic 1107 treatment. Int J Biochem Cell Biol 2018; 101:54-63. [PMID: 29800726 DOI: 10.1016/j.biocel.2018.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/04/2018] [Accepted: 05/21/2018] [Indexed: 11/22/2022]
Abstract
Sodium fluorescein ('fluorescein') staining of the ocular surface is frequently an indicator of compromised ocular health, and increases in the presence of certain contact lens multi-purpose solutions (MPS), a phenomenon known as solution induced corneal staining (SICS). The mechanism(s) underpinning fluorescein hyperfluorescence are uncertain, though may reflect increased cellular uptake of fluorescein by corneal epithelial cells. We have developed an in vitro model to study fluorescein uptake in both 'generic' mammalian cells (murine fibroblasts) and human corneal cells. Fluorescein hyperfluorescence increased after treatment with two MPS associated with clinical corneal fluorescein staining, yet there was no cellular hyperfluorescence for two MPS that do not cause this staining. Increased fluorescein uptake did not correlate with presence of a necrotic or an apoptotic marker (propidium iodide and caspase-3 respectively). Incubation of MPS-treated cells with dynasore (an inhibitor of dynamin, implicated in endocytic pathways) reduced fluorescein uptake irrespective of MPS treatment. The non-ionic surfactant Tetronic 1107 (present in both MPS associated with corneal fluorescein staining) increased uptake of fluorescein for both cell types, whereas an unrelated surfactant (Triton X-100) did not. We conclude that the clinical hyperfluorescence profile observed after exposure to four MPS can be reproduced using a simple model of cellular fluorescein uptake, suggesting this is the biological basis for SICS. Fluorescein entry does not correlate with necrosis or apoptosis, but instead involves a dynamin-dependent active process. Moreover the surfactant Tetronic 1107 appears to be a key MPS constituent triggering increased fluorescein entry, and may be the major factor responsible for SICS.
Collapse
|
34
|
Mohamed EA, Abu Hashim II, Yusif RM, Shaaban AAA, El-Sheakh AR, Hamed MF, Badria FAE. Polymeric micelles for potentiated antiulcer and anticancer activities of naringin. Int J Nanomedicine 2018; 13:1009-1027. [PMID: 29497294 PMCID: PMC5823073 DOI: 10.2147/ijn.s154325] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [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
Naringin is one of the most interesting phytopharmaceuticals that has been widely investigated for various biological actions. Yet, its low water solubility, limited permeability, and suboptimal bioavailability limited its use. Therefore, in this study, polymeric micelles of naringin based on pluronic F68 (PF68) were developed, fully characterized, and optimized. The optimized formula was investigated regarding in vitro release, storage stability, and in vitro cytotoxicity vs different cell lines. Also, cytoprotection against ethanol-induced ulcer in rats and antitumor activity against Ehrlich ascites carcinoma in mice were investigated. Nanoscopic and nearly spherical 1:50 micelles with the mean diameter of 74.80±6.56 nm and narrow size distribution were obtained. These micelles showed the highest entrapment efficiency (EE%; 96.14±2.29). The micelles exhibited prolonged release up to 48 vs 10 h for free naringin. The stability of micelles was confirmed by insignificant changes in drug entrapment, particle size, and retention (%) (91.99±3.24). At lower dose than free naringin, effective cytoprotection of 1:50 micelles against ethanol-induced ulcer in rat model has been indicated by significant reduction in mucosal damage, gastric level of malondialdehyde, gastric expression of tumor necrosis factor-alpha, caspase-3, nuclear factor kappa-light-chain-enhancer of activated B cells, and interleukin-6 with the elevation of gastric reduced glutathione and superoxide dismutase when compared with the positive control group. As well, these micelles provoked pronounced antitumor activity assessed by potentiated in vitro cytotoxicity particularly against colorectal carcinoma cells and tumor growth inhibition when compared with free naringin. In conclusion, 1:50 naringin-PF68 micelles can be represented as a potential stable nanodrug delivery system with prolonged release and enhanced antiulcer as well as antitumor activities.
Collapse
Affiliation(s)
| | | | - Rehab Mohammad Yusif
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawarah, Saudi Arabia
| | - Ahmed Abdel Aziz Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed Ramadan El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohammed Fawzy Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | |
Collapse
|
35
|
Gad SF, Park J, Park JE, Fetih GN, Tous SS, Lee W, Yeo Y. Enhancing Docetaxel Delivery to Multidrug-Resistant Cancer Cells with Albumin-Coated Nanocrystals. Mol Pharm 2018; 15:10.1021/acs.molpharmaceut.7b00783. [PMID: 29341617 PMCID: PMC6064681 DOI: 10.1021/acs.molpharmaceut.7b00783] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Intravenous delivery of poorly water-soluble anticancer drugs such as docetaxel (DTX) is challenging due to the low bioavailability and the toxicity related to solubilizing excipients. Colloidal nanoparticles are used as alternative carriers, but low drug loading capacity and circulation instability limit their clinical translation. To address these challenges, DTX nanocrystals (NCs) were prepared using Pluronic F127 as an intermediate stabilizer and albumin as a functional surface modifier, which were previously found to be effective in producing small and stable NCs. We hypothesize that the albumin-coated DTX NCs (DTX-F-alb) will remain stable in serum-containing medium so as to effectively leverage the enhanced permeability and retention effect. In addition, the surface-bound albumin, in its native form, may contribute to cellular transport of NCs through interactions with albumin-binding proteins such as secreted protein acidic and rich in cysteine (SPARC). DTX-F-alb NCs showed sheet-like structure with an average length, width, and thickness of 284 ± 96, 173 ± 56, and 40 ± 8 nm and remained stable in 50% serum solution at a concentration greater than 10 μg/mL. Cytotoxicity and cellular uptake of DTX-F-alb and unformulated (free) DTX were compared on three cell lines with different levels of SPARC expression and DTX sensitivity. While the uptake of free DTX was highly dependent on DTX sensitivity, DTX-F-alb treatment resulted in relatively consistent cellular levels of DTX. Free DTX was more efficient in entering drug-sensitive B16F10 and SKOV-3 cells than DTX-F-alb, with consistent cytotoxic effects. In contrast, multidrug-resistant NCI/ADR-RES cells took up DTX-F-alb more than free DTX with time and responded better to the former. This difference was reduced by SPARC knockdown. The high SPARC expression level of NCI/ADR-RES cells, the known affinity of albumin for SPARC, and the opposing effect of SPARC knockdown support that DTX-F-alb have exploited the surface-bound albumin-SPARC interaction in entering NCI/ADR-RES cells. Albumin-coated NC system is a promising formulation for the delivery of hydrophobic anticancer drugs to multidrug-resistant tumors.
Collapse
Affiliation(s)
- Sheryhan F. Gad
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Joonyoung Park
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Republic of Korea
| | - Gihan N. Fetih
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Sozan S. Tous
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Republic of Korea
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
36
|
Gervais M, Forens A, Ibarboure E, Carlotti S. Anionic polymerization of activated oxetane and its copolymerization with ethylene oxide for the synthesis of amphiphilic block copolymers. Polym Chem 2018. [DOI: 10.1039/c8py00307f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The anionic polymerization of oxetane in toluene was achieved using a combination of tetraoctylammonium bromide and simple triisobutylaluminum. By its copolymerization with ethylene oxide, di- and triblock copolymers were prepared in one and two steps respectively.
Collapse
Affiliation(s)
- Matthieu Gervais
- Bordeaux INP
- Univ. of Bordeaux
- CNRS
- Laboratoire de Chimie des Polymères Organiques
- UMR 5629
| | - Antoine Forens
- Bordeaux INP
- Univ. of Bordeaux
- CNRS
- Laboratoire de Chimie des Polymères Organiques
- UMR 5629
| | - Emmanuel Ibarboure
- Bordeaux INP
- Univ. of Bordeaux
- CNRS
- Laboratoire de Chimie des Polymères Organiques
- UMR 5629
| | - Stephane Carlotti
- Bordeaux INP
- Univ. of Bordeaux
- CNRS
- Laboratoire de Chimie des Polymères Organiques
- UMR 5629
| |
Collapse
|
37
|
Singh MS, Tammam SN, Shetab Boushehri MA, Lamprecht A. MDR in cancer: Addressing the underlying cellular alterations with the use of nanocarriers. Pharmacol Res 2017; 126:2-30. [PMID: 28760489 DOI: 10.1016/j.phrs.2017.07.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/29/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023]
Abstract
Multidrug resistance (MDR) is associated with a wide range of pathological changes at different cellular and intracellular levels. Nanoparticles (NPs) have been extensively exploited as the carriers of MDR reversing payloads to resistant tumor cells. However, when properly formulated in terms of chemical composition and physicochemical properties, NPs can serve as beyond delivery systems and help overcome MDR even without carrying a load of chemosensitizers or MDR reversing molecular cargos. Whether serving as drug carriers or beyond, a wise design of the nanoparticulate systems to overcome the cellular and intracellular alterations underlying the resistance is imperative. Within the current review, we will initially discuss the cellular changes occurring in resistant cells and how such changes lead to chemotherapy failure and cancer cell survival. We will then focus on different mechanisms through which nanosystems with appropriate chemical composition and physicochemical properties can serve as MDR reversing units at different cellular and intracellular levels according to the changes that underlie the resistance. Finally, we will conclude by discussing logical grounds for a wise and rational design of MDR reversing nanoparticulate systems to improve the cancer therapeutic approaches.
Collapse
Affiliation(s)
- Manu S Singh
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany
| | - Salma N Tammam
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Department of Pharmaceutical Technology, German University of Cairo, Egypt
| | | | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France.
| |
Collapse
|
38
|
Raval A, Pillai SA, Bahadur A, Bahadur P. Systematic characterization of Pluronic® micelles and their application for solubilization and in vitro release of some hydrophobic anticancer drugs. J Mol Liq 2017. [DOI: 10.1016/j.molliq.2017.01.065] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
39
|
pH-sensitive micelles for the intracellular co-delivery of curcumin and Pluronic L61 unimers for synergistic reversal effect of multidrug resistance. Sci Rep 2017; 7:42465. [PMID: 28195164 PMCID: PMC5307950 DOI: 10.1038/srep42465] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 01/11/2017] [Indexed: 12/17/2022] Open
Abstract
Pluronic L61 unimers, which are biomacromolecular modulators, and curcumin, a small-molecule modulator, were co-formulated into pH-sensitive micelles to reveal the full synergistic potential of combination drug treatments to reverse multidrug resistance (MDR). Compared to monotherapy, combined therapy significantly improved the cytotoxicity, cellular uptake and apoptotic effects of doxorubicin (DOX) against MCF-7/ADR cells. In mechanistic studies, both L61 and curcumin enhanced the cytotoxic effect by acting on mitochondrial signalling pathways. The compounds selectively accumulated in the mitochondria and disabled the mitochondria by dissipating the mitochondrial membrane potential, decreasing the ATP levels, and releasing cytochrome c, which initiated a cascade of caspase-9 and caspase-3 reactions. Furthermore, both curcumin and L61 down-regulated the expression and function of P-gp in response to drug efflux from the MCF-7/ADR cells. In the MCF-7/ADR tumour-bearing mouse model, intravenous administration of the combined therapy directly targeted the tumour, as revealed by the accumulation of DiR in the tumour site, which led to a significant inhibition of tumour growth without measurable side effects. In conclusion, co-formulation consisting of L61 and curcumin in pH-sensitive micelles induced significant synergistic effects on the reversal of MDR. Therefore, the intracellular co-delivery of various MDR modulators has great potential to reverse MDR in tumours.
Collapse
|
40
|
Pluronic Nanotechnology for Overcoming Drug Resistance. BIOACTIVITY OF ENGINEERED NANOPARTICLES 2017. [DOI: 10.1007/978-981-10-5864-6_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
41
|
Optimization and evaluation of Oridonin-loaded Soluplus ®-Pluronic P105 mixed micelles for oral administration. Int J Pharm 2016; 518:193-202. [PMID: 28012993 DOI: 10.1016/j.ijpharm.2016.12.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/07/2016] [Accepted: 12/21/2016] [Indexed: 01/07/2023]
Abstract
In this study, a new type of mixed micelles was developed using Soluplus® (SOL) and Pluronic® P105 (P105) for the encapsulation of Oridonin (ORN). Oridonin-loaded micelles (ORN-M) were simply prepared using solvent evaporation and characterized for particle size, particle morphology, encapsulation efficiency, and drug loading. In addition, the in vitro drug release behavior of ORN-M was assessed using the widely applied dialysis bag technique. The pharmacokinetic property of ORN was explored in rats after oral administration of ORN-M. Optimized ORN-M were of a small size (137.2±1.65nm) and spherical shape when the ratio of SOL:P105 was 3:1, with entrapment efficiency 90.48±1.85% and drug loading 15.08±0.38%. Oral absorption capacity of ORN was greatly enhanced with a relative bioavailability of 210.55% in comparison to that of in-house suspensions, which suggests that ORN-M shows significantly improved bioavailability and drug absorption characteristics. Overall, the optimized SOL-P105 dual mixed micelles show great potential for use as oral drug carriers for cancer treatment.
Collapse
|
42
|
Braunová A, Kostka L, Sivák L, Cuchalová L, Hvězdová Z, Laga R, Filippov S, Černoch P, Pechar M, Janoušková O, Šírová M, Etrych T. Tumor-targeted micelle-forming block copolymers for overcoming of multidrug resistance. J Control Release 2016; 245:41-51. [PMID: 27871991 DOI: 10.1016/j.jconrel.2016.11.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/31/2016] [Accepted: 11/17/2016] [Indexed: 11/17/2022]
Abstract
New amphiphilic diblock polymer nanotherapeutics serving simultaneously as a drug delivery system and an inhibitor of multidrug resistance were designed, synthesized, and evaluated for their physico-chemical and biological characteristics. The amphiphilic character of the diblock polymer, containing a hydrophilic block based on the N-(2-hydroxypropyl)methacrylamide copolymer and a hydrophobic poly(propylene oxide) block (PPO), caused self-assembly into polymer micelles with an increased hydrodynamic radius (Rh of approximately 15nm) in aqueous solutions. Doxorubicin (Dox), as a cytostatic drug, was bound to the diblock polymer through a pH-sensitive hydrazone bond, enabling prolonged circulation in blood, the delivery of Dox into a solid tumor and the subsequent stimuli-sensitive controlled release within the tumor mass and tumor cells at a decreased pH. The applicability of micellar nanotherapeutics as drug carriers was confirmed by an in vivo evaluation using EL4 lymphoma-bearing C57BL/6 mice. We observed significantly higher accumulation of micellar conjugates in a solid tumor because of the EPR effect compared with similar polymer-drug conjugates that do not form micellar structures or with the parent free drug. In addition, highly increased anti-tumor efficacy of the micellar polymer nanotherapeutics, even at a sub-optimal dose, was observed. The presence of PPO in the structure of the diblock polymer ensured, during in vitro tests on human and mouse drug-sensitive and resistant cancer cell lines, the inhibition of P-glycoprotein, one of the most frequently expressed ATP-dependent efflux pump that causes multidrug resistance. In addition, we observed highly increased rate of the uptake of the diblock polymer nanotherapeutics within the cells. We suppose that combination of unique properties based on MDR inhibition, stimuli sensitiveness (pH sensitive activation of drug), improved pharmacokinetics and increased uptake into the cells made the described polymer micelle a good candidate for investigation as potential drug delivery system.
Collapse
Affiliation(s)
- Alena Braunová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Libor Kostka
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Ladislav Sivák
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Lucie Cuchalová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Zuzana Hvězdová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Richard Laga
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Sergey Filippov
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Peter Černoch
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic
| | - Milada Šírová
- Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06 Prague, Czech Republic.
| |
Collapse
|
43
|
Enhanced in vitro cytotoxicity and anti-tumor activity of vorinostat-loaded pluronic micelles with prolonged release and reduced hepatic and renal toxicities. Eur J Pharm Sci 2016; 96:232-242. [PMID: 27667706 DOI: 10.1016/j.ejps.2016.09.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/16/2016] [Accepted: 09/21/2016] [Indexed: 11/21/2022]
Abstract
Vorinostat is the first histone deacetylase inhibitor approved by US FDA for use in cancer therapy. However, its limited aqueous solubility, low permeability, and suboptimal pharmacokinetics hinder its delivery. Thus, in this study, micelles of vorinostat with each of pluronic F68 (PF68) and pluronic F127 (PF127) were developed and optimized based on drug loading and entrapment. The optimized micelles were characterized using Fourier transform infrared spectroscopy (FT-IR), X-ray diffractometry (XRD), differential scanning calorimetry (DSC), zeta analyzer, and electron transmission microscopy. Their in vitro release, stability, in vitro cytotoxicity against HepG2, Caco-2, and MCF-7 cell lines, and finally, in vivo antitumor activity in mice bearing Ehrlich Ascites Carcinoma (EAC) were assessed. The highest entrapment efficiency was 99.09±2.16% and 94.19±2.37% for micelles of 1:50 drug to polymer ratio with each of PF127 and PF68, respectively. These micelles were nearly spherical with nanoscopic mean diameters of 72.61±10.66nm for PF68 and 91.88±10.70nm for PF127 with narrow size distribution. The micelles provided prolonged release at phosphate buffer saline pH7.4 up to 24h for PF68 and 72h for PF127. Potentiation of in vitro cytotoxicity of vorinostat was more pronounced with PF127 micelles particularly against MCF-7 cells. Compared with free vorinostat, the micelles with PF127 were more effective in inhibiting tumor growth as well as exhibiting significantly (p<0.05) diminished hepatic and renal toxicities. In conclusion, 1:50 vorinostat-PF127 micelles may facilitate i.v. formulations and can be suggested as a promising stable and safe nanoparticulate delivery system with prolonged release and potentiated cytotoxicity.
Collapse
|
44
|
Nguyen-Kim V, Prévost S, Seidel K, Maier W, Marguerre AK, Oetter G, Tadros T, Gradzielski M. Solubilization of active ingredients of different polarity in Pluronic® micellar solutions – Correlations between solubilizate polarity and solubilization site. J Colloid Interface Sci 2016; 477:94-102. [DOI: 10.1016/j.jcis.2016.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 11/28/2022]
|
45
|
Abstract
Thermo-reversible polymeric gels represent an interesting class of materials that can be tailored for a wide range of applications. The triblock poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO) based systems, PEO-PPO-PEO, show thermoreversible gelation around body temperature and, therefore, are particularly suitable for biomedical applications such as drug delivery, gene therapy and tissue engineering. The PEO-PPO-PEO tri-block copolymers have amphiphilic characteristics and self-assemble into micelles to form a variety of close packed structures. By varying the block composition (PEO/PPO ratio) and the molecular weight, it is possible to tailor the final properties of these systems to meet the specific application needs. In this report the thermodynamic basis of micellization of PEO-PPO-PEO systems is described. The factors influencing the micelles formation are discussed along with the methods used to investigate the micellization process and morphology as well as with the main applications of these systems in biomedical fields.
Collapse
|
46
|
Janas C, Mostaphaoui Z, Schmiederer L, Bauer J, Wacker MG. Novel polymeric micelles for drug delivery: Material characterization and formulation screening. Int J Pharm 2016; 509:197-207. [PMID: 27234698 DOI: 10.1016/j.ijpharm.2016.05.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/11/2016] [Accepted: 05/13/2016] [Indexed: 12/24/2022]
Abstract
A rising number of new chemical entities that exhibit only poor aqueous solubility are identified in drug discovery processes. Polymeric micelles composed of block copolymers (BP) facilitate the delivery of such lipophilic molecules in drug therapy. Consequently, a rational screening and selection procedure for novel BP was established. Further, the interplay of polymer structure, micelle formation and drug binding was studied. Therefore seven polymers (BP001 to BP007) were synthesized from different monomer compositions resulting in nanocarriers varying in surface decoration and lipophilicity. These polymers were characterized by H(1)-NMR and SEC. The molecular weight was ranging between 13 and 37kDa. The critical micelle concentration and micellar integrity in presence of human plasma were determined. Micelles were loaded with dexamethasone and characterized with regards to their size, morphology and surface charge. Polymeric micelles with a size of 49.21-236.37nm were obtained. A half-life of 11h was determined for five of the copolymers in presence of human plasma. Two nanocarrier formulations (BP006 and BP007) were exhibiting optimal micellar integrity in vitro and a modified release profile under biorelevant conditions. Strongest drug-polymer interaction was observed for nanocarrier compositions providing benzyl and carboxylic groups and were composed of BP006 and BP007.
Collapse
Affiliation(s)
- Christine Janas
- Goethe University, Institute of Pharmaceutical Technology, D-60438 Frankfurt, Germany
| | - Zouhair Mostaphaoui
- Goethe University, Institute of Pharmaceutical Technology, D-60438 Frankfurt, Germany
| | | | - Johann Bauer
- Merck KGaA, Frankfurter Straße 250, D-64293, Darmstadt, Germany
| | - Matthias G Wacker
- Fraunhofer-Institute for Molecular Biology and Applied Ecology, Project Group for Translational Medicine & Pharmacology, D-60438 Frankfurt, Germany.
| |
Collapse
|
47
|
Chen LC, Chen YC, Su CY, Hong CS, Ho HO, Sheu MT. Development and characterization of self-assembling lecithin-based mixed polymeric micelles containing quercetin in cancer treatment and an in vivo pharmacokinetic study. Int J Nanomedicine 2016; 11:1557-66. [PMID: 27143878 PMCID: PMC4841422 DOI: 10.2147/ijn.s103681] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Quercetin (Que) is known to have biological benefits including an anticancer effect, but low water solubility limits its clinical application. The aim of this study was to develop a lecithin-based mixed polymeric micelle (LMPM) delivery system to improve the solubility and bioavailability of Que. The optimal Que-LMPM, composed of Que, lecithin, Pluronic® P123, and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy[poly(ethylene glycol)-2000] in a proportion of 3:1:17.5:2.5 (w/w), was prepared by a thin-film method. The average size, polydispersion index, encapsulating efficiency, and drug loading of Que-LMPM were 61.60±5.02 nm, 0.589±0.198, 96.87%±9.04%, and 12.18%±1.11%, respectively. The solubility of Que in the Que-LMPM system increased to 5.81 mg/mL, compared to that of free Que in water of 0.17–7.7 μg/mL. The Que-LMPM system presented a sustained-release property in vitro. The in vitro cytotoxicity assay showed that the 50% inhibitory concentration values toward MCF-7 breast cancer cells for free Que, blank LMPMs, and Que-LMPMs were >200, >200, and 110 μM, respectively, indicating the nontoxicity of the LMPM carrier, but the LMPM formulation enhanced the cytotoxicity of Que against MCF-7 cells. A cellular uptake assay also confirmed the intake of Que-LMPM by MCF-7 cells. An in vivo pharmacokinetic study demonstrated that Que-LMPMs had higher area under the concentration–time curve and a longer half-life, leading to better bioavailability compared to a free Que injection. Due to their nanosize, core–shell structure, and solubilization potential, LMPMs were successfully developed as a drug delivery system for Que to improve its solubility and bioavailability.
Collapse
Affiliation(s)
- Ling-Chun Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ying-Chen Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chia-Yu Su
- School of Pharmacy, College of Pharmacy, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chung-Shu Hong
- School of Pharmacy, College of Pharmacy, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-O Ho
- School of Pharmacy, College of Pharmacy, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan; Clinical Research Center and Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
48
|
Drug-Loaded Perfluorocarbon Nanodroplets for Ultrasound-Mediated Drug Delivery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:221-41. [DOI: 10.1007/978-3-319-22536-4_13] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Herzberger J, Niederer K, Pohlit H, Seiwert J, Worm M, Wurm FR, Frey H. Polymerization of Ethylene Oxide, Propylene Oxide, and Other Alkylene Oxides: Synthesis, Novel Polymer Architectures, and Bioconjugation. Chem Rev 2015; 116:2170-243. [PMID: 26713458 DOI: 10.1021/acs.chemrev.5b00441] [Citation(s) in RCA: 465] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The review summarizes current trends and developments in the polymerization of alkylene oxides in the last two decades since 1995, with a particular focus on the most important epoxide monomers ethylene oxide (EO), propylene oxide (PO), and butylene oxide (BO). Classical synthetic pathways, i.e., anionic polymerization, coordination polymerization, and cationic polymerization of epoxides (oxiranes), are briefly reviewed. The main focus of the review lies on more recent and in some cases metal-free methods for epoxide polymerization, i.e., the activated monomer strategy, the use of organocatalysts, such as N-heterocyclic carbenes (NHCs) and N-heterocyclic olefins (NHOs) as well as phosphazene bases. In addition, the commercially relevant double-metal cyanide (DMC) catalyst systems are discussed. Besides the synthetic progress, new types of multifunctional linear PEG (mf-PEG) and PPO structures accessible by copolymerization of EO or PO with functional epoxide comonomers are presented as well as complex branched, hyperbranched, and dendrimer like polyethers. Amphiphilic block copolymers based on PEO and PPO (Poloxamers and Pluronics) and advances in the area of PEGylation as the most important bioconjugation strategy are also summarized. With the ever growing toolbox for epoxide polymerization, a "polyether universe" may be envisaged that in its structural diversity parallels the immense variety of structural options available for polymers based on vinyl monomers with a purely carbon-based backbone.
Collapse
Affiliation(s)
- Jana Herzberger
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany
| | - Kerstin Niederer
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Hannah Pohlit
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany.,Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany.,Department of Dermatology, University Medical Center , Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Jan Seiwert
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | - Matthias Worm
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany
| | - Frederik R Wurm
- Max Planck Graduate Center , Staudingerweg 6, D-55128 Mainz, Germany.,Max Planck Institute for Polymer Research , Ackermannweg 10, D-55128 Mainz, Germany
| | - Holger Frey
- Institute of Organic Chemistry, Johannes Gutenberg-University Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany.,Graduate School Materials Science in Mainz , Staudingerweg 9, D-55128 Mainz, Germany
| |
Collapse
|
50
|
Martin P, Giardiello M, McDonald TO, Smith D, Siccardi M, Rannard SP, Owen A. Augmented Inhibition of CYP3A4 in Human Primary Hepatocytes by Ritonavir Solid Drug Nanoparticles. Mol Pharm 2015; 12:3556-68. [DOI: 10.1021/acs.molpharmaceut.5b00204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Philip Martin
- Department
of Molecular and Clinical Pharmacology, University of Liverpool, Block H, 70 Pembroke Place, Liverpool L69 3GF, U.K
| | - Marco Giardiello
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 3BX, U.K
| | - Tom O. McDonald
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 3BX, U.K
| | - Darren Smith
- Department
of Applied Sciences, University of Northumbria at Newcastle, Ellison
Building, Newcastle NE1
8ST, U.K
| | - Marco Siccardi
- Department
of Molecular and Clinical Pharmacology, University of Liverpool, Block H, 70 Pembroke Place, Liverpool L69 3GF, U.K
| | - Steven P. Rannard
- Department
of Chemistry, University of Liverpool, Crown Street, Liverpool L69 3BX, U.K
| | - Andrew Owen
- Department
of Molecular and Clinical Pharmacology, University of Liverpool, Block H, 70 Pembroke Place, Liverpool L69 3GF, U.K
| |
Collapse
|