1
|
Hernández-Velázquez ED, Granados-López AJ, López JA, Solorio-Alvarado CR. Multidrug Resistance Reversed by Maleimide Interactions. A Biological and Synthetic Overview for an Emerging Field. Chembiochem 2025; 26:e202400640. [PMID: 39383297 DOI: 10.1002/cbic.202400640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/11/2024]
Abstract
Multidrug Resistance (MDR) can be considered one of the most frightening adaptation types in bacteria, fungi, protozoa, and eukaryotic cells. It allows the organisms to survive the attack of many drugs used in the daily basis. This forces the development of new and more complex, highly specific drugs to fight diseases. Given the high usage of medicaments, poor variation in active chemical cores, and self-medication, the appearance of MDR is more frequent each time, and has been established as a serious medical and social problem. Over the years it has been possible the identification of several genes and proteins responsible for MDR and with that the development of blockers of them to reach MDR reversion and try to avoid a global problem. These mechanisms also have been observed in cancer cells, and several calcium channel blockers have been successful in MDR reversion, and the maleimide can be found included in them. In this review, we explore particularly the tree main proteins involved in cancer chemoresistance, MRP1 (encoded by ABCC1), BCRP (encoded by ABCG2) and P-gp (encoded by ABCB1). The participation of P-gp is remarkably important, and several aspects of its regulations are discussed. Additionally, we address the history, mechanisms, reversion efforts, and we specifically focused on the maleimide synthesis as MDR-reversers in co-administration, as well as on how their biological applications are imperative to expand the available information and explore a very plausible MDR reversion source.
Collapse
Affiliation(s)
- Edson D Hernández-Velázquez
- Campus Guanajuato, División de Ciencias Naturales y Exactas, Departamento de Química, Universidad de Guanajuato, Noria Alta S/N, 36050, Guanajuato, Gto., México
| | | | - Jesús Adrián López
- Laboratorio de MicroRNAs y Cáncer, Universidad Autónoma de Zacatecas, 98066, Zacatecas, México
| | - César R Solorio-Alvarado
- Campus Guanajuato, División de Ciencias Naturales y Exactas, Departamento de Química, Universidad de Guanajuato, Noria Alta S/N, 36050, Guanajuato, Gto., México
| |
Collapse
|
2
|
Li C, Chen L, McClements DJ, Peng X, Xu Z, Meng M, Ji H, Qiu C, Long J, Jin Z. Encapsulation of polyphenols in protein-based nanoparticles: Preparation, properties, and applications. Crit Rev Food Sci Nutr 2024; 64:11341-11355. [PMID: 37486163 DOI: 10.1080/10408398.2023.2237126] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Polyphenols have a variety of physiological activities, including antioxidant, antimicrobial, and anti-inflammatory properties. However, their applications are often limited because due to the instability of polyphenols. Encapsulation technologies can be employed to overcome these problems and increase the utilization of polyphenols. In this article, the utilization of protein-based nanoparticles for encapsulating polyphenols is reviewed due to their good biocompatibility, biodegradability, and functional attributes. Initially, the various kinds of animal and plant proteins available for forming protein nanoparticles are discussed, as well as the fabrication methods that can be used to assemble these nanoparticles. The molecular interaction mechanisms between proteins and polyphenols are then summarized. Applications of protein-based nanoparticles for encapsulating polyphenols are then discussed, including as nutrient delivery systems, in food packaging materials, and in the creation of functional foods. Finally, areas where further research is need on the development, characterization, and application of protein-based polyphenol-loaded nanoparticles are highlighted.
Collapse
Affiliation(s)
- Cuicui Li
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Long Chen
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, South China Agricultural University, Guangzhou, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, China
| | | | - Xinwen Peng
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, China
| | - Zhenlin Xu
- School of Food Science and Technology, South China Agricultural University, Guangzhou, China
| | - Man Meng
- Licheng Detection & Certification Group Co., Ltd, Zhongshan, China
| | - Hangyan Ji
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chao Qiu
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jie Long
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhengyu Jin
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
3
|
Chen X, Fan R, Wang Y, Munir M, Li C, Wang C, Hou Z, Zhang G, Liu L, He J. Bovine milk β-casein: Structure, properties, isolation, and targeted application of isolated products. Compr Rev Food Sci Food Saf 2024; 23:e13311. [PMID: 38445543 DOI: 10.1111/1541-4337.13311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/07/2024]
Abstract
β-Casein, an important protein found in bovine milk, has significant potential for application in the food, pharmaceutical, and other related industries. This review first introduces the composition, structure, and functional properties of β-casein. It then reviews the techniques for isolating β-casein. Chemical and enzymatic isolation methods result in inactivity of β-casein and other components in the milk, and it is difficult to control the production conditions, limiting the utilization range of products. Physical technology not only achieves high product purity and activity but also effectively preserves the biological activity of the components. The isolated β-casein needs to be utilized effectively and efficiently for various purity products in order to achieve optimal targeted application. Bovine β-casein, which has a purity higher than or close to that of breast β-casein, can be used in infant formulas. This is achieved by modifying its structure through dephosphorylation, resulting in a formula that closely mimics the composition of breast milk. Bovine β-casein, which is lower in purity than breast β-casein, can be maximized for the preparation of functional peptides and for use as natural carriers. The remaining byproducts can be utilized as food ingredients, emulsifiers, and carriers for encapsulating and delivering active substances. Thus, realizing the intensive processing and utilization of bovine β-casein isolation. This review can promote the industrial production process of β-casein, which is beneficial for the sustainable development of β-casein as a food and material. It also provides valuable insights for the development of other active substances in milk.
Collapse
Affiliation(s)
- Xiaoqian Chen
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Rui Fan
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Yuanbin Wang
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Maliha Munir
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Chun Li
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Caiyun Wang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Zhanqun Hou
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Guofang Zhang
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Libo Liu
- Key Laboratory of Dairy Science, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Science Research Institute, Harbin, China
| | - Jian He
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| |
Collapse
|
4
|
Peng H, Wang J, Chen J, Peng Y, Wang X, Chen Y, Kaplan DL, Wang Q. Challenges and opportunities in delivering oral peptides and proteins. Expert Opin Drug Deliv 2023; 20:1349-1369. [PMID: 37450427 PMCID: PMC10990675 DOI: 10.1080/17425247.2023.2237408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Rapid advances in bioengineering enable the use of complex proteins as therapeutic agents to treat diseases. Compared with conventional small molecule drugs, proteins have multiple advantages, including high bioactivity and specificity with low toxicity. Developing oral dosage forms with active proteins is a route to improve patient compliance and significantly reduce production costs. However, the gastrointestinal environment remains a challenge to this delivery path due to enzymatic degradation, low permeability, and weak absorption, leading to reduced delivery efficiency and poor clinical outcomes. AREAS COVERED This review describes the barriers to oral delivery of peptides and complex proteins, current oral delivery strategies utilized and the opportunities and challenges ahead to try and circumvent these barriers. Oral protein drugs on the market and clinical trials provide insights and approaches for advancing delivery strategies. EXPERT OPINION Although most current studies on oral protein delivery rely on in vitro and in vivo animal data, the safety and limitations of the approach in humans remain uncertain. The shortage of clinical data limits the development of new or alternative strategies. Therefore, designing appropriate oral delivery strategies remains a significant challenge and requires new ideas, innovative design strategies and novel model systems.
Collapse
Affiliation(s)
- Haisheng Peng
- Department of Pharmacology, Medical College, University of Shaoxing, Shaoxing, China
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Jiahe Wang
- Department of Humanities, Daqing Branch, Harbin Medical University, Daqing, China
| | - Jiayu Chen
- Department of Pharmacology, Medical College, University of Shaoxing, Shaoxing, China
| | - Yanbo Peng
- Department of Pharmaceutical Engineering, China Pharmaceutical University, 639 Longmian Rd, Nanjing 211198, China
| | - Xiaoxian Wang
- The Affiliated Hospital of Medical College, University of Shaoxing, Shaoxing, Zhejiang Province, China
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
5
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [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: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
6
|
Obozina AS, Komedchikova EN, Kolesnikova OA, Iureva AM, Kovalenko VL, Zavalko FA, Rozhnikova TV, Tereshina ED, Mochalova EN, Shipunova VO. Genetically Encoded Self-Assembling Protein Nanoparticles for the Targeted Delivery In Vitro and In Vivo. Pharmaceutics 2023; 15:231. [PMID: 36678860 PMCID: PMC9861179 DOI: 10.3390/pharmaceutics15010231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Targeted nanoparticles of different origins are considered as new-generation diagnostic and therapeutic tools. However, there are no targeted drug formulations within the composition of nanoparticles approved by the FDA for use in the clinic, which is associated with the insufficient effectiveness of the developed candidates, the difficulties of their biotechnological production, and inadequate batch-to-batch reproducibility. Targeted protein self-assembling nanoparticles circumvent this problem since proteins are encoded in DNA and the final protein product is produced in only one possible way. We believe that the combination of the endless biomedical potential of protein carriers as nanoparticles and the standardized protein purification protocols will make significant progress in "magic bullet" creation possible, bringing modern biomedicine to a new level. In this review, we are focused on the currently existing platforms for targeted self-assembling protein nanoparticles based on transferrin, lactoferrin, casein, lumazine synthase, albumin, ferritin, and encapsulin proteins, as well as on proteins from magnetosomes and virus-like particles. The applications of these self-assembling proteins for targeted delivery in vitro and in vivo are thoroughly discussed, including bioimaging applications and different therapeutic approaches, such as chemotherapy, gene delivery, and photodynamic and photothermal therapy. A critical assessment of these protein platforms' efficacy in biomedicine is provided and possible problems associated with their further development are described.
Collapse
Affiliation(s)
| | | | | | - Anna M. Iureva
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Vera L. Kovalenko
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Fedor A. Zavalko
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | | | - Elizaveta N. Mochalova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Victoria O. Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
7
|
Romano A, Engelberg Y, Landau M, Lesmes U. Alpha-lactalbumin amyloid-like fibrils for intestinal delivery: Formation, physiochemical characterization, and digestion fate of capsaicin-loaded fibrils. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2022.108248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
8
|
A comparative evaluation of anti-tumor activity following oral and intravenous delivery of doxorubicin in a xenograft model of breast tumor. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00595-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose
Natural materials have been extensively studied for oral drug delivery due to their biodegradability and other unique properties. In the current research, we fabricated sodium caseinate nanomicelles (NaCNs) using casein as a natural polymer to develop a controlled-release oral delivery system that would improve the therapeutic potential of doxorubicin (DOX) and reduce its toxicity.
Methods
DOX-loaded NaCNs were synthesized and thoroughly characterized, then subjected to in vivo anti-tumor evaluation and bio-distribution analysis in a 4T1-induced breast cancer model.
Results
Our findings indicated that the tumor would shrink by eight-fold in the group orally treated with DOX-NaCNs when compared to free DOX. The tumor accumulated drug 1.27-fold more from the orally administered DOX-NaCNs compared to the intravenously administered DOX-NaCNs, 6.8-fold more compared to free DOX, and 8.34-times more compared to orally administered free DOX. In comparison, the orally administered DOX-NaCNs lead to a significant reduction in tumor size (5.66 ± 4.36 mm3) compared to intravenously administered DOX-NaCNs (10.29 ± 4.86 mm3) on day 17 of the experiment. NaCNs were well tolerated at a single dose of 2000 mg/kg in an acute oral toxicity study.
Conclusion
The enhanced anti-tumor effects of oral DOX-NaCNs might be related to the controlled release of DOX from the delivery system when compared to free DOX and the intravenous formulation of DOX-NaCNs. Moreover, NaCNs is recognized as a safe and non-toxic delivery system with excellent bio-distribution profile and high anti-tumor effects that has a potential for oral chemotherapy.
Collapse
|
9
|
Li M, Wen X, Wang K, Liu Z, Ni Y. Maillard induced glycation of β-casein for enhanced stability of the self-assembly micelles against acidic and calcium environment. Food Chem 2022; 387:132914. [PMID: 35421650 DOI: 10.1016/j.foodchem.2022.132914] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 03/12/2022] [Accepted: 04/04/2022] [Indexed: 11/04/2022]
Abstract
Bovine β-casein (β-CN) has attracted increasingly interest as biocompatible nanocarrier for hydrophobic flavonoid due to its self-assembly ability to form micelles. This paper reported Maillard induced glycation reaction of β-CN using dextran in order to improve stability of naringenin-loaded β-CN micelles under acidic and high calcium environments. Our results showed that solubility of β-CN-graft-dextran was remarkable increased at acidic pH and the conjugation with 20 kDa dextran had the highest level of graft degree. Glycation restrained β-CN from aggregating around pH 5.0 where was close to the isoelectric point, forming spherical micelles with irregular and rough surfaces, which were significantly larger than the micelles at pH 7.0. β-CN-graft-dextran also overcame destabilization of the micelles induced by excess calcium and had no impact on the chelating ability of calcium. These findings appeared to be promising for future applications of modified β-CN-graft-dextran based on Maillard reaction as fairly stable nanocarrier under extreme condition.
Collapse
Affiliation(s)
- Mo Li
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Beijing 100083, China; National Academy of Agriculture Green Development, China Agricultural University, 100193 Beijing, China; National Engineering Research Center for Fruits and Vegetables Processing, No. 17 Qinghua East Road, Beijing 100083, China
| | - Xin Wen
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Beijing 100083, China; National Engineering Research Center for Fruits and Vegetables Processing, No. 17 Qinghua East Road, Beijing 100083, China
| | - Kunli Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Beijing 100083, China; National Engineering Research Center for Fruits and Vegetables Processing, No. 17 Qinghua East Road, Beijing 100083, China
| | - Zihao Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Beijing 100083, China; National Engineering Research Center for Fruits and Vegetables Processing, No. 17 Qinghua East Road, Beijing 100083, China
| | - Yuanying Ni
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Beijing 100083, China; National Engineering Research Center for Fruits and Vegetables Processing, No. 17 Qinghua East Road, Beijing 100083, China.
| |
Collapse
|
10
|
Bovine alpha-lactalbumin particulates for controlled delivery: Impact of dietary fibers on stability, digestibility, and gastro-intestinal release of capsaicin. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2022.107536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
11
|
Gong G, Qian W, Zhang L, Jia J, Xie J, Zhu Q, Liu W, Tu P, Gao M, Zhang L, Tang H, Su H, Wei K, Zhou C, Wang KK, Zhang Z, Pan Q. A curcumin-induced assembly of a transferrin nanocarrier system and its antitumor effect. Colloids Surf B Biointerfaces 2022; 217:112613. [DOI: 10.1016/j.colsurfb.2022.112613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 10/17/2022]
|
12
|
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
|
13
|
Nanoparticle-based drug delivery systems to overcome gastric cancer drug resistance. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
14
|
Gandhi S, Roy I. Drug delivery applications of casein nanostructures: A minireview. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
15
|
Larsen JB, Taebnia N, Dolatshahi-Pirouz A, Eriksen AZ, Hjørringgaard C, Kristensen K, Larsen NW, Larsen NB, Marie R, Mündler AK, Parhamifar L, Urquhart AJ, Weller A, Mortensen KI, Flyvbjerg H, Andresen TL. Imaging therapeutic peptide transport across intestinal barriers. RSC Chem Biol 2021; 2:1115-1143. [PMID: 34458827 PMCID: PMC8341777 DOI: 10.1039/d1cb00024a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Oral delivery is a highly preferred method for drug administration due to high patient compliance. However, oral administration is intrinsically challenging for pharmacologically interesting drug classes, in particular pharmaceutical peptides, due to the biological barriers associated with the gastrointestinal tract. In this review, we start by summarizing the pharmacological performance of several clinically relevant orally administrated therapeutic peptides, highlighting their low bioavailabilities. Thus, there is a strong need to increase the transport of peptide drugs across the intestinal barrier to realize future treatment needs and further development in the field. Currently, progress is hampered by a lack of understanding of transport mechanisms that govern intestinal absorption and transport of peptide drugs, including the effects of the permeability enhancers commonly used to mediate uptake. We describe how, for the past decades, mechanistic insights have predominantly been gained using functional assays with end-point read-out capabilities, which only allow indirect study of peptide transport mechanisms. We then focus on fluorescence imaging that, on the other hand, provides opportunities to directly visualize and thus follow peptide transport at high spatiotemporal resolution. Consequently, it may provide new and detailed mechanistic understanding of the interplay between the physicochemical properties of peptides and cellular processes; an interplay that determines the efficiency of transport. We review current methodology and state of the art in the field of fluorescence imaging to study intestinal barrier transport of peptides, and provide a comprehensive overview of the imaging-compatible in vitro, ex vivo, and in vivo platforms that currently are being developed to accelerate this emerging field of research.
Collapse
Affiliation(s)
- Jannik Bruun Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Nayere Taebnia
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Alireza Dolatshahi-Pirouz
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Anne Zebitz Eriksen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Claudia Hjørringgaard
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Kasper Kristensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Nanna Wichmann Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Niels Bent Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Rodolphe Marie
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Ann-Kathrin Mündler
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Ladan Parhamifar
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Andrew James Urquhart
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Arjen Weller
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Kim I Mortensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Henrik Flyvbjerg
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Thomas Lars Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| |
Collapse
|
16
|
Yuan Z, Deng S, Chen L, Hu Y, Gu J, He L. pH-driven entrapment of enrofloxacin in casein-based nanoparticles for the enhancement of oral bioavailability. Food Sci Nutr 2021; 9:4057-4067. [PMID: 34401057 PMCID: PMC8358345 DOI: 10.1002/fsn3.2224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/01/2021] [Accepted: 02/14/2021] [Indexed: 11/11/2022] Open
Abstract
Enrofloxacin (ENR), a broad-spectrum antibacterial drug, has extremely poor water solubility contributing to low bioavailability, which prevents drug formulation design and limits its wide application in livestock farming and aquaculture. Compared to conventional formulations of ENR, casein (Cas)-based drug delivery system has been reported to have significant advantages in the improvement of solubility and bioavailability of drugs. In this paper, we report the preparation process of ENR-loaded Cas nanoparticles (ENR-Cas) using magnetic agitation without any organic agent and the optimization of the formulation. Transmission electron microscopy (TEM), dynamic light scattering (DLS), differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), and Fourier transform infrared spectroscopy (FTIR) were all adopted to characterize the ENR-Cas. Results showed that the obtained ENR-Cas were approximately spherical with a particle size of 171.6 ± 13.8 nm with a polydispersity index of 0.322 ± 0.053. In vitro release behavior of ENR-Cas showed a sustained release profile. Additionally, in vivo study in rats displayed that the mean plasma concentration of ENR after oral administration of ENR-Cas was significantly higher than that treated with ENR suspension. The mean residence time (MRT0-24) of ENR was enhanced by Cas nanoparticles from 9.287 ± 0.524 to 11.372 ± 1.139 hr in comparison with ENR suspension. Accordingly, the area under the curve (AUC0-24) of ENR-Cas was 80.521 ± 6.624 μg·hr/ml, 3.8-fold higher than that of ENR suspension (20.850 ± 1.715 μg·hr/ml). Therefore, it can be concluded that ENR-Cas enhanced the absorption, prolonged the retention time, and improved oral bioavailability of ENR. Taken the good oral safety of Cas into consideration, ENR-Cas should be a more promising oral preparation of ENR for clinical application.
Collapse
Affiliation(s)
- Zhi‐xiang Yuan
- College of PharmacySouthwest Minzu UniversityChengduChina
| | - Shichen Deng
- College of Animal & Veterinary SciencesSouthwest Minzu UniversityChengduChina
| | - Li Chen
- Department of PharmacyCollege of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - You Hu
- Department of PharmacyCollege of Veterinary MedicineSichuan Agricultural UniversityChengduChina
| | - Jian Gu
- College of PharmacySouthwest Minzu UniversityChengduChina
| | - Lili He
- College of PharmacySouthwest Minzu UniversityChengduChina
| |
Collapse
|
17
|
Metallo-Liposomes Derived from the [Ru(bpy)3]2+ Complex as Nanocarriers of Therapeutic Agents. CHEMOSENSORS 2021. [DOI: 10.3390/chemosensors9050090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The obtaining of nanocarriers of gene material and small drugs is still an interesting research line. Side-effects produced by the toxicity of several pharmaceutics, the high concentrations needed to get therapeutic effects, or their excessive use by patients have motivated the search for new nanostructures. For these reasons, cationic metallo-liposomes composed by phosphatidylcholine (PC), cholesterol (CHO) and RuC1C19 (a surfactant derived from the metallic complex [Ru(bpy)3]2+) were prepared and characterized by using diverse techniques (zeta potential, dynamic light scattering and electronic transmission microscopy –TEM-). Unimodal or bimodal populations of spherical aggregates with small sizes were obtained depending on the composition of the liposomes. The presence of cholesterol favored the formation of small aggregates. ct-DNA was condensed in the presence of the liposomes investigated. In-vitro assays demonstrated the ability of these nanoaggregates to internalize into different cell lines. A positive gene transfection into human bone osteosarcoma epithelial cells (U2OS) was also observed. The RuC1C19 surfactant was used as sensor to quantify the binding of DNA to the liposomes. Doxorubicin was encapsulated into the metallo-liposomes, demonstrating their ability to be also used as nanocarriers of drugs. A relationship between then encapsulation percentage of the antibiotic and the composition of the aggregates has been established.
Collapse
|
18
|
Sotoudegan F, Sotoudegan F, Talebkhan Garoosi Y, Afshar SH, Barkhordari F, Davami F. Anti-Aβ-scFv-loaded polymeric nano-micelles with enhanced plasma stability. J Pharm Pharmacol 2021; 73:460-472. [PMID: 33793837 DOI: 10.1093/jpp/rgaa068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 12/23/2020] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Immunotherapy using recombinant monoclonal antibodies specifically Anti-amyloid-beta (Anti-Aβ) scFv is envisaged as an appropriate therapeutic for Alzheimer through reduction of amyloid-beta aggregation. The solubilization of therapeutics using polymeric micelles facilitates an improved bioavailability and extended blood half-life. In this study, the optimum production condition for Anti-amyloid-beta (Anti-Aβ) scFv was obtained. To increase the stability of plasma, Anti-Aβ-loaded polymeric micelles were synthesized. METHODS Escherichia coli SHuffle expression strain was used and purified by Ni-NTA. Pluronics P85 and F127 micelles were used for the Anti-Aβ delivery and were characterized in terms of morphology, drug loading and drug release in phosphate buffer and artificial cerebrospinal fluid. The stability profile was quantified at 4°C over a 30 days storage period. The stability in human plasma was also evaluated. KEY FINDINGS Proteins expressed in SHuffle resulted in increased levels of protein expression and solubility. Low critical micelle concentration value and high micelle encapsulation efficiency (<200 nm) achieved via direct dissolution method. Anti-Aβ-loaded micelles were around 2.2-fold more stable than Anti-Aβ in plasma solution. A sustained in-vitro release of Anti-Aβ from micelles was observed. CONCLUSIONS Results confirmed that Pluronic-micelles pose benefits as a nano-carrier to increase the stability of Anti-Aβ scFvin in the plasma.
Collapse
Affiliation(s)
- Farnaz Sotoudegan
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Farzaneh Sotoudegan
- Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sahar H Afshar
- Faculty of Pharmacy International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
19
|
Das T, Anand U, Pandey SK, Ashby CR, Assaraf YG, Chen ZS, Dey A. Therapeutic strategies to overcome taxane resistance in cancer. Drug Resist Updat 2021; 55:100754. [PMID: 33691261 DOI: 10.1016/j.drup.2021.100754] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/17/2022]
Abstract
One of the primary causes of attenuated or loss of efficacy of cancer chemotherapy is the emergence of multidrug resistance (MDR). Numerous studies have been published regarding potential approaches to reverse resistance to taxanes, including paclitaxel (PTX) and docetaxel, which represent one of the most important classes of anticancer drugs. Since 1984, following the FDA approval of paclitaxel for the treatment of advanced ovarian carcinoma, taxanes have been extensively used as drugs that target tumor microtubules. Taxanes, have been shown to affect an array of oncogenic signaling pathways and have potent cytotoxic efficacy. However, the clinical success of these drugs has been restricted by the emergence of cancer cell resistance, primarily caused by the overexpression of MDR efflux transporters or by microtubule alterations. In vitro and in vivo studies indicate that the mechanisms underlying the resistance to PTX and docetaxel are primarily due to alterations in α-tubulin and β-tubulin. Moreover, resistance to PTX and docetaxel results from: 1) alterations in microtubule-protein interactions, including microtubule-associated protein 4, stathmin, centriole, cilia, spindle-associated protein, and kinesins; 2) alterations in the expression and activity of multidrug efflux transporters of the ABC superfamily including P-glycoprotein (P-gp/ABCB1); 3) overexpression of anti-apoptotic proteins or inhibition of apoptotic proteins and tumor-suppressor proteins, as well as 4) modulation of signal transduction pathways associated with the activity of several cytokines, chemokines and transcription factors. In this review, we discuss the abovementioned molecular mechanisms and their role in mediating cancer chemoresistance to PTX and docetaxel. We provide a detailed analysis of both in vitro and in vivo experimental data and describe the application of these findings to therapeutic practice. The current review also discusses the efficacy of different pharmacological modulations to achieve reversal of PTX resistance. The therapeutic roles of several novel compounds, as well as herbal formulations, are also discussed. Among them, many structural derivatives had efficacy against the MDR phenotype by either suppressing MDR or increasing the cytotoxic efficacy compared to the parental drugs, or both. Natural products functioning as MDR chemosensitizers offer novel treatment strategies in patients with chemoresistant cancers by attenuating MDR and increasing chemotherapy efficacy. We broadly discuss the roles of inhibitors of P-gp and other efflux pumps, in the reversal of PTX and docetaxel resistance in cancer cells and the significance of using a nanomedicine delivery system in this context. Thus, a better understanding of the molecular mechanisms mediating the reversal of drug resistance, combined with drug efficacy and the application of target-based inhibition or specific drug delivery, could signal a new era in modern medicine that would limit the pathological consequences of MDR in cancer patients.
Collapse
Affiliation(s)
- Tuyelee Das
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India
| | - Uttpal Anand
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Swaroop Kumar Pandey
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, West Bengal, India.
| |
Collapse
|
20
|
Romano A, Lajterer C, Shpigelman A, Lesmes U. Bovine alpha-lactalbumin assemblies with capsaicin: Formation, interactions, loading and physiochemical characterization. Food Chem 2021; 352:129306. [PMID: 33677213 DOI: 10.1016/j.foodchem.2021.129306] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 11/26/2022]
Abstract
Numerous human conditions can benefit from diets rich in proteins and bioactives, such as capsaicin (CAP), yet their effective delivery is a sensorial, scientific and technological challenge. This study hypothesized that CAP can form various complexes with native bovine alpha-lactalbumin (holo-ALA) and decalcified-ALA (apo-ALA). Calorimetric and spectroscopic techniques reveals ALA-CAP molecular complexation is spontaneous, exothermic and accompanied by various conformational changes. ITC shows the interaction stoichiometry (n) and binding constant (Kb) for holo-ALA to be 0.87 ± 0.03, 1.54 ± 0.23 × 105 M-1 and for apo-ALA to be 0.64 ± 0.09, 9.41 ± 2.16 × 104 M-1. Molecular docking further elucidates that hydrogen bonds govern CAP binding to holo-ALA while hydrophobic interactions dominate binding to apo-ALA in a structural cleft. Finally, this work shows these interactions along with controlled aggregation can be utilized to form CAP-loaded colloids with encapsulation efficiency of 47.1 ± 1.0%. Thus, this study shows great promise in the prospective use of ALA as an edible delivery vehicle for CAP.
Collapse
Affiliation(s)
- Alon Romano
- Laboratory of Chemistry of Foods and Bioactives, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Carolina Lajterer
- Laboratory of Chemistry of Foods and Bioactives, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Avi Shpigelman
- Laboratory of Chemistry of Foods and Bioactives, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Uri Lesmes
- Laboratory of Chemistry of Foods and Bioactives, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
21
|
Atanase LI. Micellar Drug Delivery Systems Based on Natural Biopolymers. Polymers (Basel) 2021; 13:477. [PMID: 33540922 PMCID: PMC7867356 DOI: 10.3390/polym13030477] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/30/2022] Open
Abstract
The broad diversity of structures and the presence of numerous functional groups available for chemical modifications represent an enormous advantage for the development of safe, non-toxic, and cost-effective micellar drug delivery systems (DDS) based on natural biopolymers, such as polysaccharides, proteins, and peptides. Different drug-loading methods are used for the preparation of these micellar systems, but it appeared that dialysis is generally recommended, as it avoids the formation of large micellar aggregates. Moreover, the preparation method has an important influence on micellar size, morphology, and drug loading efficiency. The small size allows the passive accumulation of these micellar systems via the permeability and retention effect. Natural biopolymer-based micellar DDS are high-value biomaterials characterized by good compatibility, biodegradability, long blood circulation time, non-toxicity, non-immunogenicity, and high drug loading, and they are biodegraded to non-toxic products that are easily assimilated by the human body. Even if some recent studies reported better antitumoral effects for the micellar DDS based on polysaccharides than for commercial formulations, their clinical use is not yet generalized. This review is focused on the studies from the last decade concerning the preparation as well as the colloidal and biological characterization of micellar DDS based on natural biopolymers.
Collapse
Affiliation(s)
- Leonard Ionut Atanase
- Department of Biomaterials, Faculty of Medical Dentistry, "Apollonia" University of Iasi, Pacurari Street, No. 11, 700511 Iasi, Romania
| |
Collapse
|
22
|
Kashyap D, Tuli HS, Yerer MB, Sharma A, Sak K, Srivastava S, Pandey A, Garg VK, Sethi G, Bishayee A. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin Cancer Biol 2021; 69:5-23. [PMID: 31421264 DOI: 10.1016/j.semcancer.2019.08.014] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 01/09/2023]
Abstract
Application of natural product-based nanoformulations for the treatment of different human diseases, such as cancer, is an emerging field. The conventional cancer therapeutic modalities, including surgery, chemotherapy, immunotherapy, radiotherapy has limited achievements. A larger number of drawbacks are associated with these therapies, including damage to proliferating healthy tissues, structural deformities, systemic toxicity, long-term side effects, resistance to the drug by tumor cells, and psychological problems. The advent of nanotechnology in cancer therapeutics is recent; however, it has progressed and transformed the field of cancer treatment at a rapid rate. Nanotherapeutics have promisingly overcome the limitations of conventional drug delivery system, i.e., low aqueous solubility, low bioavailability, multidrug resistance, and non-specificity. Specifically, natural product-based nanoformulations are being intentionally studied in different model systems. Where it is found that these nanoformulations has more proximity and reduced side effects. The nanoparticles can specifically target tumor cells, enhancing the specificity and efficacy of cancer therapeutic modalities which in turn improves patient response and survival. The integration of phytotherapy and nanotechnology in the clinical setting may improve pharmacological response and better clinical outcome of patients.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh - 160 012, Punjab, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala - 133 207, Haryana, India.
| | - Mukerrem Betul Yerer
- Department of Pharmacology, Faculty of Pharmacy, University of Erciyes, Kayseri 38039, Turkey
| | - Ajay Sharma
- Department of Chemistry, Career Point University, Tikker-Kharwarian, Hamirpur - 176 041, Himachal Pradesh, India
| | | | - Saumya Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad - 211 004, Uttar Pradesh, India
| | - Anjana Pandey
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad - 211 004, Uttar Pradesh, India
| | - Vivek Kumar Garg
- Department of Biochemistry, Government Medical College and Hospital, Sector 32, Chandigarh - 160 031, Punjab, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
23
|
Crosslinked casein micelles bound paclitaxel as enzyme activated intracellular drug delivery systems for cancer therapy. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110237] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
24
|
Sinitsyna OV, Vorob’ev MM. Atomic force microscopy characterization of β-casein nanoparticles on mica and graphite. MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.01.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Rehan F, Ahemad N, Islam RA, Gupta M, Gan SH, Chowdhury EH. Optimization and Formulation of Nanostructured and Self-Assembled Caseinate Micelles for Enhanced Cytotoxic Effects of Paclitaxel on Breast Cancer Cells. Pharmaceutics 2020; 12:pharmaceutics12100984. [PMID: 33080962 PMCID: PMC7589039 DOI: 10.3390/pharmaceutics12100984] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Paclitaxel (PTX) is a widely used anti-cancer drug for treating various types of solid malignant tumors including breast, ovarian and lung cancers. However, PTX has a low therapeutic response and is linked with acquired resistance, as well as a high incidence of adverse events, such as allergic reactions, neurotoxicity and myelosuppression. The situation is compounded when its complex chemical structure contributes towards hydrophobicity, shortening its circulation time in blood, causing off-target effects and limiting its therapeutic activity against cancer cells. Formulating a smart nano-carrier may overcome the solubility and toxicity issues of the drug and enable its more selective delivery to the cancerous cells. Among the nano-carriers, natural polymers are of great importance due to their excellent biodegradability, non-toxicity and good accessibility. The aim of the present research is to develop self-assembled sodium caseinate nanomicelles (NaCNs) with PTX loaded into the hydrophobic core of NaCNs for effective uptake of the drug in cancer cells and its subsequent intracellular release. METHODS The PTX-loaded micelle was characterized with high-performance liquid chromatography (HPLC), Fourier Transform Infrared Spectra (FTIR), High Resolution-Transmission Electron Microscope (HR-TEM), Field Emission Scanning Electron Microscope (FESEM) and Energy Dispersive X-Ray (EDX). Following treatment with PTX-loaded NaCNs, cell viability, cellular uptake and morphological changes were analyzed using MCF-7 and MDA-MB 231 human breast cancer cell lines. RESULTS We found that PTX-loaded NaCNs efficiently released PTX in an acidic tumor environment, while showing an enhanced cytotoxicity, cellular uptake and in-vivo anti-tumor efficacy in a mouse model of breast cancer when compared to free drug and blank micelles. Additionally, the nanomicelles also presented improved colloidal stability for three months at 4 °C and -20 °C and when placed at a temperature of 37 °C. CONCLUSIONS We conclude that the newly developed NaCNs is a promising carrier of PTX to enhance tumor accumulation of the drug while addressing its toxicity issues as well.
Collapse
Affiliation(s)
- Farah Rehan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
- Tropical Medicine and Biology Multidisciplinary Platform, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia
- Global Asia in the 21st century Research Platform, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia
| | - Rowshan Ara Islam
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia;
| | - Manish Gupta
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
- School of Pharmaceutical and Population Health Informatics, DIT University, Mussoorie-Diversion Road, Dehradun, Uttarakhand-248009, India
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia; (F.R.); (N.A.); (M.G.); (S.H.G.)
| | - Ezharul Hoque Chowdhury
- Tropical Medicine and Biology Multidisciplinary Platform, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway 47500, Petaling Jaya, Selangor, Malaysia;
- Correspondence:
| |
Collapse
|
26
|
Zhu YX, Jia HR, Duan QY, Liu X, Yang J, Liu Y, Wu FG. Photosensitizer-Doped and Plasma Membrane-Responsive Liposomes for Nuclear Drug Delivery and Multidrug Resistance Reversal. ACS APPLIED MATERIALS & INTERFACES 2020; 12:36882-36894. [PMID: 32666795 DOI: 10.1021/acsami.0c09110] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Clinically approved doxorubicin (Dox)-loaded liposomes (e.g., Doxil) guarantee good biosafety, but their insufficient nuclear delivery of Dox (<0.4%) after cellular uptake significantly hampers their final anticancer efficacy. Here, we report that simply doping protoporphyrin IX (PpIX, a commonly used hydrophobic photosensitizer) into the lipid bilayers of Dox-loaded liposomes (the resultant product is termed PpIX/Dox liposomes) is a feasible way to promote the nuclear delivery of Dox. This facile strategy relies on a unique property of PpIX-it presents considerably higher affinity for the real plasma membrane over its liposomal carrier, which drives the doped PpIX molecules to detach from the liposomes when encountering cancer cells. We demonstrate that this process can trigger the efficient release of the loaded Dox molecules and allow them to enter the nuclei of MCF-7 breast cancer cells without being trapped by lysosomes. Regarding the drug-resistant MCF-7/ADR cells, the aberrant activation of the efflux pumps in the plasma membranes expels the internalized Dox. However, we strikingly find that the robust drug resistance can be reversed upon mild laser irradiation because the photodynamic effect of PpIX disrupts the drug efflux system (e.g., P-glycoprotein) and facilitates the nuclear entry of Dox. As a proof-of-concept, this PpIX doping strategy is also applicable for enhancing the effectiveness of cisplatin-loaded liposomes against both A549 and A549/DDP lung cancer cells. In vivo experimental results prove that a single injection of PpIX/Dox liposomes completely impedes the growth of MCF-7 tumors in nude mice within 2 weeks and, in combination with laser irradiation, can synergistically ablate MCF-7/ADR tumors. Biosafety assessments reveal no significant systemic toxicity caused by PpIX/Dox liposomes. This work exemplifies a facile method to modulate the subcellular fate of liposomal drugs and may inspire the optimization of nanopharmaceuticals in the near future.
Collapse
Affiliation(s)
- Ya-Xuan Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Hao-Ran Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Jing Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Yi Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| |
Collapse
|
27
|
Gupta SK, Singh P, Ali V, Verma M. Role of membrane-embedded drug efflux ABC transporters in the cancer chemotherapy. Oncol Rev 2020; 14:448. [PMID: 32676170 PMCID: PMC7358983 DOI: 10.4081/oncol.2020.448] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
One of the major problems being faced by researchers and clinicians in leukemic treatment is the development of multidrug resistance (MDR) which restrict the action of several tyrosine kinase inhibitors (TKIs). MDR is a major obstacle to the success of cancer chemotherapy. The mechanism of MDR involves active drug efflux transport of ABC superfamily of proteins such as Pglycoprotein (P-gp/ABCB1), multidrug resistance-associated protein 2 (MRP2/ABCC2), and breast cancer resistance protein (BCRP/ABCG2) that weaken the effectiveness of chemotherapeutics and negative impact on the future of anticancer therapy. In this review, the authors aim to provide an overview of various multidrug resistance (MDR) mechanisms observed in cancer cells as well as the various strategies developed to overcome these MDR. Extensive studies have been carried out since last several years to enhance the efficacy of chemotherapy by defeating these MDR mechanisms with the use of novel anticancer drugs that could escape from the efflux reaction, MDR modulators or chemosensitizers, multifunctional nanotechnology, and RNA interference (RNAi) therapy.
Collapse
Affiliation(s)
- Sonu Kumar Gupta
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Punjab, India
| | - Priyanka Singh
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Punjab, India
| | - Villayat Ali
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Punjab, India
| | - Malkhey Verma
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Punjab, India
| |
Collapse
|
28
|
Liu Y, Bai H, Guo K, Wang P. Hypocrellin B triggered sonodynamic therapy reverses multidrug resistance of doxorubicin-resistant SGC7901/ADR cells via down-regulation of P-gp expression. J Chemother 2020; 32:385-393. [DOI: 10.1080/1120009x.2020.1778242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Yichen Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Hong Bai
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
- Medical College, Xi’an Peihua University, Xi’an, Shaanxi, China
| | - Kaili Guo
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, China
| |
Collapse
|
29
|
Vorob'ev M, Sinitsyna O. Degradation and assembly of β-casein micelles during proteolysis by trypsin. Int Dairy J 2020. [DOI: 10.1016/j.idairyj.2020.104652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Towards the overcoming of anticancer drug resistance mediated by p53 mutations. Drug Resist Updat 2020; 49:100671. [DOI: 10.1016/j.drup.2019.100671] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022]
|
31
|
Dallavalle S, Dobričić V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, Tsakovska I, Zidar N, Fruttero R. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat 2020; 50:100682. [PMID: 32087558 DOI: 10.1016/j.drup.2020.100682] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/28/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is the dominant cause of the failure of cancer chemotherapy. The design of antitumor drugs that are able to evade MDR is rapidly evolving, showing that this area of biomedical research attracts great interest in the scientific community. The current review explores promising recent approaches that have been developed with the aim of circumventing or overcoming MDR. Encouraging results have been obtained in the investigation of the MDR-modulating properties of various classes of natural compounds and their analogues. Inhibition of P-gp or downregulation of its expression have proven to be the main mechanisms by which MDR can be surmounted. The use of hybrid molecules that are able to simultaneously interact with two or more cancer cell targets is currently being explored as a means to circumvent drug resistance. This strategy is based on the design of hybrid compounds that are obtained either by merging the structural features of separate drugs, or by conjugating two drugs or pharmacophores via cleavable/non-cleavable linkers. The approach is highly promising due to the pharmacokinetic and pharmacodynamic advantages that can be achieved over the independent administration of the two individual components. However, it should be stressed that the task of obtaining successful multivalent drugs is a very challenging one. The conjugation of anticancer agents with nitric oxide (NO) donors has recently been developed, creating a particular class of hybrid that can combat tumor drug resistance. Appropriate NO donors have been shown to reverse drug resistance via nitration of ABC transporters and by interfering with a number of metabolic enzymes and signaling pathways. In fact, hybrid compounds that are produced by covalently attaching NO-donors and antitumor drugs have been shown to elicit a synergistic cytotoxic effect in a variety of drug resistant cancer cell lines. Another strategy to circumvent MDR is based on nanocarrier-mediated transport and the controlled release of chemotherapeutic drugs and P-gp inhibitors. Their pharmacokinetics are governed by the nanoparticle or polymer carrier and make use of the enhanced permeation and retention (EPR) effect, which can increase selective delivery to cancer cells. These systems are usually internalized by cancer cells via endocytosis and accumulate in endosomes and lysosomes, thus preventing rapid efflux. Other modalities to combat MDR are described in this review, including the pharmaco-modulation of acridine, which is a well-known scaffold in the development of bioactive compounds, the use of natural compounds as means to reverse MDR, and the conjugation of anticancer drugs with carriers that target specific tumor-cell components. Finally, the outstanding potential of in silico structure-based methods as a means to evaluate the ability of antitumor drugs to interact with drug transporters is also highlighted in this review. Structure-based design methods, which utilize 3D structural data of proteins and their complexes with ligands, are the most effective of the in silico methods available, as they provide a prediction regarding the interaction between transport proteins and their substrates and inhibitors. The recently resolved X-ray structure of human P-gp can help predict the interaction sites of designed compounds, providing insight into their binding mode and directing possible rational modifications to prevent them from becoming P-gp drug substrates. In summary, although major efforts were invested in the search for new tools to combat drug resistant tumors, they all require further implementation and methodological development. Further investigation and progress in the abovementioned strategies will provide significant advances in the rational combat against cancer MDR.
Collapse
Affiliation(s)
- Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Loretta Lazzarato
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Elena Gazzano
- Department of Oncology, Università degli Studi di Torino, Via Santena 5/bis, 10126 Turin, Italy
| | - Miguel Machuqueiro
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, C8 Building, Campo Grande, 1749-016, Lisbon, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| | - Ilza Pajeva
- QSAR and Molecular Modelling Department, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 105, 1113 Sofia, Bulgaria
| | - Ivanka Tsakovska
- QSAR and Molecular Modelling Department, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Block 105, 1113 Sofia, Bulgaria
| | - Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Roberta Fruttero
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Turin, Italy.
| |
Collapse
|
32
|
Coelho ALS, Feuser PE, Carciofi BAM, de Andrade CJ, de Oliveira D. Mannosylerythritol lipids: antimicrobial and biomedical properties. Appl Microbiol Biotechnol 2020; 104:2297-2318. [PMID: 31980917 DOI: 10.1007/s00253-020-10354-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/16/2022]
Abstract
Mannosylerythritol lipids (MELs) have attracted particular interest of medical, pharmaceutical, and cosmetic fields, due to their specific characteristics, including non-toxicity, easy biodegradability, and environmental compatibility. Therefore, this review aims to highlight recent findings on MEL biological properties, focusing on issues related to therapeutic applications. Among the main findings is that MELs can play a fundamental role due to their antimicrobial properties against several nosocomial pathogen microorganisms. Other remarkable biological properties of MELs are related to skincare, as antiaging (active agent), and in particular on recover of skin cells that were damaged by UV radiation. MEL is also related to the increased efficiency of DNA transfection in liposome systems. Regarding the health field, these glycolipids seem to be associated with disturbance in the membrane composition of cancerous cells, increasing expression of genes responsible for cytoplasmic stress and apoptosis. Moreover, MELs can be associated with nanoparticles, as a capping agent, also acting to increase the solubility and cytotoxicity of them. Furthermore, the differences in the chemical structure of MEL could improve and expand their biochemical diversity and applications. Such modifications could change their interfacial properties and, thus, reduce the surface tension value, enhance the solubility, lower critical micelle concentrations, and form unique self-assembly structures. The latest is closely related to molecular recognition and protein stabilization properties of MEL, that is, essential parameters for their effective cosmetical and pharmaceutical effects. Thus, this current research indicates the huge potential of MEL for use in biomedical formulations, either alone or in combination with other molecules.
Collapse
Affiliation(s)
- Ana Letícia Silva Coelho
- Department of Chemical Engineering and Food Engineering, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Paulo Emílio Feuser
- Department of Chemical Engineering and Food Engineering, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Bruno Augusto Mattar Carciofi
- Department of Chemical Engineering and Food Engineering, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Cristiano José de Andrade
- Department of Chemical Engineering and Food Engineering, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
| | - Débora de Oliveira
- Department of Chemical Engineering and Food Engineering, Federal University of Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| |
Collapse
|
33
|
Dong J, Qin Z, Zhang WD, Cheng G, Yehuda AG, Ashby CR, Chen ZS, Cheng XD, Qin JJ. Medicinal chemistry strategies to discover P-glycoprotein inhibitors: An update. Drug Resist Updat 2020; 49:100681. [PMID: 32014648 DOI: 10.1016/j.drup.2020.100681] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/16/2022]
Abstract
The presence of multidrug resistance (MDR) in malignant tumors is one of the primary causes of treatment failure in cancer chemotherapy. The overexpression of the ATP binding cassette (ABC) transporter, P-glycoprotein (P-gp), which significantly increases the efflux of certain anticancer drugs from tumor cells, produces MDR. Therefore, inhibition of P-gp may represent a viable therapeutic strategy to overcome cancer MDR. Over the past 4 decades, many compounds with P-gp inhibitory efficacy (referred to as first- and second-generation P-gp inhibitors) have been identified or synthesized. However, these compounds were not successful in clinical trials due to a lack of efficacy and/or untoward toxicity. Subsequently, third- and fourth-generation P-gp inhibitors were developed but dedicated clinical trials did not indicate a significant therapeutic effect. In recent years, an extraordinary array of highly potent, selective, and low-toxicity P-gp inhibitors have been reported. Herein, we provide a comprehensive review of the synthetic and natural products that have specific inhibitory activity on P-gp drug efflux as well as promising chemosensitizing efficacy in MDR cancer cells. The present review focuses primarily on the structural features, design strategies, and structure-activity relationships (SAR) of these compounds.
Collapse
Affiliation(s)
- Jinyun Dong
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China; College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zuodong Qin
- Research Center of Biochemical Engineering Technology, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Gang Cheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Assaraf G Yehuda
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Charles R Ashby
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, China; College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
34
|
Wang Y, Yu X, Wang L, Zhang F, Zhang Y. Research Progress on Chemical Constituents and Anticancer Pharmacological Activities of Euphorbia lunulata Bunge. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3618941. [PMID: 32420336 PMCID: PMC7201523 DOI: 10.1155/2020/3618941] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 12/18/2022]
Abstract
Euphorbia lunulata Bunge (ELB) is a traditional Chinese medicine possessing the functions of expectoration, cough relief, asthma relief, detoxification, and itching relief. Modern pharmacological studies have showed that ELB exhibits a variety of activities, such as antitumor, antibacterial, and antioxidant activities. In particular, the anticancer activities of ELB have attracted much attention. In this review, we summarize the recent research progress on the chemical constituents and anticancer activities of ELB by searching the PubMed, Web of Science, and China National Knowledge Infrastructure databases. The results show that more than 151 components have been identified from extracts of ELB, including 73 terpenoids, 28 flavonoids, 8 phenylpropanoids, 7 steroids, 19 phenols, and 5 alkaloids. ELB has been shown to exhibit significant inhibitory effects on lung, cervical, gastric, breast, and liver cancers, and its anticancer effects are mainly manifested in the 3 aspects, including cell cycle arrest, cell apoptosis, and inhibition of the migration of cancer cells.
Collapse
Affiliation(s)
- Yuwei Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiao Yu
- Shandong Medicine Technician College, Tai'an 271016, China
| | - Lingna Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Fang Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yongqing Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
35
|
Engelberg S, Netzer E, Assaraf YG, Livney YD. Selective eradication of human non-small cell lung cancer cells using aptamer-decorated nanoparticles harboring a cytotoxic drug cargo. Cell Death Dis 2019; 10:702. [PMID: 31541073 PMCID: PMC6754387 DOI: 10.1038/s41419-019-1870-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 01/06/2023]
Abstract
Targeted cancer therapy is currently the leading modality to enhance treatment selectivity and efficacy, as well as to minimize untoward toxicity to healthy tissues. Herein, we devised and studied nanoparticles (NPs) composed of the biocompatible block-copolymer PEG-PCL entrapping the hydrophobic chemotherapeutic drug paclitaxel (PTX), which are targeted to human non-small cell lung cancer (NSCLC) cells. To achieve selective NSCLC targeting, these NPs were decorated with single-stranded oligonucleotide-based S15 aptamers (S15-APTs), which we have recently shown to serve as efficient tumor cell targeting ligands. Prepared without using surfactants, these 15 nm PEG-PCL/PTX NPs entered NSCLC cells via clathrin-mediated endocytosis. These NPs demonstrated efficient encapsulation of PTX, high selectivity to- and potent eradication of human A549 NSCLC cells, with a remarkable half maximal inhibitory concentration (IC50) of 0.03 μM PTX. In contrast, very high IC50 values of 1.7, 4.2, 43, 87, and 980 µM PTX were obtained towards normal human bronchial epithelial BEAS2B, cervical carcinoma HeLa, colon adenocarcinoma CaCo-2, neonatal foreskin fibroblast FSE, and human embryonic kidney HEK-293 cells, respectively. These results demonstrate 2–5 orders of magnitude difference in the selective cytotoxicity towards NSCLCs, reflecting a potentially outstanding therapeutic window. Moreover, the dual utility of aptamer-decorated NPs for both drug stabilization and selective tumor targeting was studied by increasing APT concentrations during NP “decoration”. The optimal aptamer density on the surface of NPs for selective targeting, for high fluorescence diagnostic signal and for maintaining small particle size to enable endocytosis, was achieved by using 30 nM APTs during NP decoration. Collectively, our findings suggest that these APT-decorated NPs hold great preclinical promise in selective targeting and eradication of human NSCLC cells without harming normal tissues.
Collapse
Affiliation(s)
- Shira Engelberg
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 3200000, Haifa, Israel
| | - Einat Netzer
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 3200000, Haifa, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion - Israel Institute of Technology, 3200000, Haifa, Israel.
| | - Yoav D Livney
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 3200000, Haifa, Israel.
| |
Collapse
|
36
|
Milman N, Ginini L, Gil Z. Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resist Updat 2019; 45:1-12. [PMID: 31369918 DOI: 10.1016/j.drup.2019.07.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/19/2019] [Accepted: 07/21/2019] [Indexed: 12/25/2022]
Abstract
Exosomes are a class of extracellular vesicles ranging in size from 40 to 100 nm, which are secreted by both cancer cells and multiple stromal cells in the tumor microenvironment. Following their secretion, exosomes partake in endocrine, paracrine and autocrine signaling. Internalization of exosomes by tumor cells influences several cellular pathways which alter cancer cell physiology. Tumor-derived exosomes secreted by cancer or stromal cells can also confer anticancer drug-resistant traits upon cancer cells. These exosomes promote chemoresistance by transferring their cargo which includes nucleic acids, proteins, and metabolites to cancer cells or act as a decoy for immunotherapeutic targets. Depletion of exosomes can reverse some of the detrimental effects on tumor metabolism and restore drug sensitivity to chemotherapeutic treatment. Herein we discuss various approaches that have been developed to deplete exosomes for therapeutic purposes. The natural composition, low immunogenicity and cytotoxicity of exosomes, along with their ability to specifically target tumor cells, render them an appealing platform for drug delivery. The ability of exosomes to mediate autocrine and paracrine signaling in target cells, along with their natural structure and low immunogenicity render them an attractive vehicle for the delivery of anticancer drugs to tumors.
Collapse
Affiliation(s)
- Neta Milman
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, The Clinical Research Institute at Rambam Healthcare Campus, Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel
| | - Lana Ginini
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, The Clinical Research Institute at Rambam Healthcare Campus, Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ziv Gil
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, The Clinical Research Institute at Rambam Healthcare Campus, Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
37
|
Rehan F, Ahemad N, Gupta M. Casein nanomicelle as an emerging biomaterial—A comprehensive review. Colloids Surf B Biointerfaces 2019; 179:280-292. [DOI: 10.1016/j.colsurfb.2019.03.051] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 02/22/2019] [Accepted: 03/24/2019] [Indexed: 12/15/2022]
|
38
|
Kaundal B, Srivastava AK, Sardoiwala MN, Karmakar S, Choudhury SR. A NIR-responsive indocyanine green-genistein nanoformulation to control the polycomb epigenetic machinery for the efficient combinatorial photo/chemotherapy of glioblastoma. NANOSCALE ADVANCES 2019; 1:2188-2207. [PMID: 36131972 PMCID: PMC9419092 DOI: 10.1039/c9na00212j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/13/2019] [Indexed: 06/15/2023]
Abstract
Combinatorial photodynamics and chemotherapy have drawn enormous attention as therapeutic modalities via precise stimuli-responsive drug delivery for glioblastoma, which can overcome the limitations associated with conventional therapies. Herein, we have prepared an indocyanine green tagged, genistein encapsulated casein nanoformulation (ICG-Gen@CasNPs) that exhibits the near infra-red region responsive controlled release of genistein and enhanced cellular uptake in the human glioblastoma monolayer and a three-dimensional raft culture model via the enhanced retention effect. ICG-Gen@CasNPs, with the integrated photosensitizer indocyanine green within the nanoformulation, triggered oxidative stress, activating the apoptosis cascade, promoting cell cycle arrest and damaging the mitochondrial membrane potential, collectively directing glioblastoma cell death. The suppression of the polycomb group of proteins in the glioblastoma upon ICG-Gen@CasNPs/NIR exposure revealed the involvement of the epigenetic repression complex machinery in the regulation. Furthermore, ICG-Gen@CasNPs/PDT/PTT directed ubiquitination and proteasomal degradation of EZH2 and BMI1 indicates the implication of the polycomb in conferring glioblastoma survival. The increased activation of the apoptotic pathways and the generation of cellular reactive oxygen species upon inhibiting the expression of EZH2 and BMI1 strengthen our observations. It is worth noting that ICG-Gen@CasNPs robustly accumulated in the brain after crossing the blood-brain barrier, which represents the eminent biocompatibility and means that the system is devoid of any nonspecific toxicity in vivo. Moreover, a superior anti-tumor effect was demonstrated on a three-dimensional glioma spheroid model. Thus, this combinatorial chemo/photodynamic therapy revealed that ICG-Gen@CasNPs mediated epigenetic regulation, which is a crucial molecular mechanism of GBM suppression.
Collapse
Affiliation(s)
- Babita Kaundal
- Institute of Nano Science and Technology, Habitat Centre Phase-10, Sector 64 Mohali Punjab India
| | - Anup K Srivastava
- Institute of Nano Science and Technology, Habitat Centre Phase-10, Sector 64 Mohali Punjab India
| | | | - Surajit Karmakar
- Institute of Nano Science and Technology, Habitat Centre Phase-10, Sector 64 Mohali Punjab India
| | - Subhasree Roy Choudhury
- Institute of Nano Science and Technology, Habitat Centre Phase-10, Sector 64 Mohali Punjab India
| |
Collapse
|
39
|
Edelman R, Assaraf YG, Slavkin A, Dolev T, Shahar T, Livney YD. Developing Body-Components-Based Theranostic Nanoparticles for Targeting Ovarian Cancer. Pharmaceutics 2019; 11:E216. [PMID: 31060303 PMCID: PMC6572588 DOI: 10.3390/pharmaceutics11050216] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer mortality is the highest among gynecologic malignancies. Hence, the major challenges are early diagnosis and efficient targeted therapy. Herein, we devised model theranostic nanoparticles (NPs) for combined diagnostics and delivery of chemotherapeutics, targeted to ovarian cancer cells. These NPs were made of natural biocompatible and biodegradable body components: hyaluronic acid (HA) and serum albumin (SA). The hydrophilic HA served as the targeting ligand for cancer cells overexpressing CD44, the HA receptor. SA, the natural carrier of various ligands through the blood, served as the hydrophobic block of the self-assembling block copolymeric Maillard-conjugates. We show the successful construction of fluorescently-labeled SA-HA conjugate-based theranostic NPs, their loading with paclitaxel (PTX) (association constant (8.6 ± 0.8) × 103 M-1, maximal loading capacity of 4:1 PTX:BSA, and 96% encapsulation efficiency), selective internalization and cytotoxicity to CD44-overexpressing ovarian cancer cells (IC50: 26.4 ± 2.3 nM, compared to 115.0 ± 17.4 of free PTX, and to 58.6 ± 19.7 nM for CD44-lacking cognate ovarian cancer cells). Fluorescein isothiocyanate (FITC) was used for in vitro imaging, whereas long wavelength fluorophores or other suitable tracers would be used for future in vivo diagnostic imaging. Collectively, our findings demonstrate that fluorescent HA-SA NPs harboring a cytotoxic drug cargo can specifically target, label CD44-expressing ovarian cancer cells and efficiently eradicate them.
Collapse
Affiliation(s)
- Ravit Edelman
- The Lab of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| | - Anton Slavkin
- The Lab of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| | - Tamar Dolev
- The Lab of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| | - Tal Shahar
- The Lab of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| | - Yoav D Livney
- The Lab of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| |
Collapse
|
40
|
Homayun B, Lin X, Choi HJ. Challenges and Recent Progress in Oral Drug Delivery Systems for Biopharmaceuticals. Pharmaceutics 2019; 11:E129. [PMID: 30893852 PMCID: PMC6471246 DOI: 10.3390/pharmaceutics11030129] [Citation(s) in RCA: 454] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/09/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023] Open
Abstract
Routes of drug administration and the corresponding physicochemical characteristics of a given route play significant roles in therapeutic efficacy and short term/long term biological effects. Each delivery method has favorable aspects and limitations, each requiring a specific delivery vehicles design. Among various routes, oral delivery has been recognized as the most attractive method, mainly due to its potential for solid formulations with long shelf life, sustained delivery, ease of administration and intensified immune response. At the same time, a few challenges exist in oral delivery, which have been the main research focus in the field in the past few years. The present work concisely reviews different administration routes as well as the advantages and disadvantages of each method, highlighting why oral delivery is currently the most promising approach. Subsequently, the present work discusses the main obstacles for oral systems and explains the most recent solutions proposed to deal with each issue.
Collapse
Affiliation(s)
- Bahman Homayun
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada.
| | - Xueting Lin
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada.
| | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada.
| |
Collapse
|
41
|
β-Casein micelles for oral delivery of SN-38 and elacridar to overcome BCRP-mediated multidrug resistance in gastric cancer. Eur J Pharm Biopharm 2018; 133:240-249. [DOI: 10.1016/j.ejpb.2018.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 11/22/2022]
|
42
|
Batra H, Pawar S, Bahl D. Curcumin in combination with anti-cancer drugs: A nanomedicine review. Pharmacol Res 2018; 139:91-105. [PMID: 30408575 DOI: 10.1016/j.phrs.2018.11.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 10/31/2018] [Accepted: 11/04/2018] [Indexed: 12/31/2022]
Abstract
A huge surge of research is being conducted on combination therapy with anticancer compounds formulated in the form of nanoparticles (NPs). Numerous advantages like dose minimalization and synergism, reversal of multi drug resistance (MDRs), enhanced efficacy have emerged with nanoencapsulation of chemotherapeutic agents with chemo-sensitizing agent like curcumin. Within last couple of years various nano-sized formulations have been designed and tested both in vitro with cell lines for different types of cancers and in vivo with cancer types and drug resistance models. Despite the combinatorial models being advanced, translation to human trials has not been as smooth as one would have hoped, with as few as twenty ongoing clinical trials with curcumin combination, with less than 1/10th being nano-particulate formulations. Mass production of nano-formulation based on their physico-chemical and pharmacokinetics deficits poses as major hurdle up the ladder. Combination of these nano-sized dosage with poorly bioavailable drugs, unspecific target binding ability and naturally unstable curcumin further complicates the formulation aspects. Emphasis is now therefore being laid on altering natural forms of curcumin and usage of formulations like prodrug or coating of curcumin to overcome stability issues and focus more on enhancing the pharmaceutical and therapeutic ability of the nano-composites. Current studies and futuristic outlook in this direction are discussed in the review, which can serve as the basis for upcoming research which could boost commercial translational of improved nano-sized curcumin combination chemotherapy.
Collapse
Affiliation(s)
- Harshul Batra
- Neuroscience Institute & Center for Behavioral Neuroscience, Georgia State University, 789 Petit Science Center, Atlanta, GA, 30303, United States.
| | - Shrikant Pawar
- Department of Computer Science, Georgia State University, 34 Peachtree Street, Atlanta, GA, 30303, United States; Department of Biology, Georgia State University, 34 Peachtree Street, Atlanta, GA, 30303, United States
| | - Dherya Bahl
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
43
|
Perinelli DR, Bonacucina G, Cespi M, Bonazza F, Palmieri GF, Pucciarelli S, Polzonetti V, Attarian L, Polidori P, Vincenzetti S. A comparison among β-caseins purified from milk of different species: Self-assembling behaviour and immunogenicity potential. Colloids Surf B Biointerfaces 2018; 173:210-216. [PMID: 30296645 DOI: 10.1016/j.colsurfb.2018.09.079] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/24/2018] [Accepted: 09/28/2018] [Indexed: 12/13/2022]
Abstract
Caseins are a family of proteins constituted by α-caseins (αs-1 and αs-2 caseins), β-caseins and κ-caseins. β-caseins, in particular, show a temperature and concentration-dependent self-assembling behaviour. Recently, β-casein micelles have been proposed as natural nanocarriers for the delivery of hydrophobic compounds, promoting their bioavailability. Until now, all studies regarding both chemical-physical characterization and applications of β-caseins have employed the protein of bovine origin. However, it could be interesting to exploit the use of β-caseins from other milk sources for their potential encapsulation ability and immunogenicity but, at present, no information on the self-assembling behaviour is available for β-caseins from the milk of species different from bovine. In this work, for the first time, β-caseins from human milk and from donkey, goat, and sheep milk were purified and their self-assembling behaviour was compared to that of a commercial bovine β-casein, the only one for which the concentration and temperature aggregation behaviour is known. Furthermore, a preliminary evaluation of the immunogenicity potential of β-casein from other milk sources has been performed by cross-reaction experiments using anti-β-casein antibodies from bovine origin. The results indicated a similar self-assembling profile among all β-caseins examined compared to the bovine β-casein, suggesting the possible use of β-casein from other milk sources as nanocarriers. Since donkey and human β-casein do not cross-react with bovine anti-β-casein antibodies, they could be particularly interesting for the development of self-assembling systems with lower hypoallergenic potential.
Collapse
Affiliation(s)
| | | | - Marco Cespi
- School of Pharmacy, University of Camerino, Camerino, MC, Italy.
| | - Francesca Bonazza
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | | | - Stefania Pucciarelli
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | - Valeria Polzonetti
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | - Loucine Attarian
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | - Paolo Polidori
- School of Pharmacy, University of Camerino, Camerino, MC, Italy.
| | - Silvia Vincenzetti
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| |
Collapse
|
44
|
Xia Y, Fang M, Dong J, Xu C, Liao Z, Ning P, Zeng Q. pH sensitive liposomes delivering tariquidar and doxorubicin to overcome multidrug resistance of resistant ovarian cancer cells. Colloids Surf B Biointerfaces 2018; 170:514-520. [PMID: 29960952 DOI: 10.1016/j.colsurfb.2018.06.055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/03/2018] [Accepted: 06/25/2018] [Indexed: 11/16/2022]
Abstract
Multidrug resistance of tumour cells is one of the most important hurdles in tumour chemotherapy. To overcome the multidrug resistance, we constructed a pH-sensitive liposome formulation (pHSL) by loading tariquidar (TQR) and DOX simultaneously in this work. The formulation showed high stability at pH 7.4 and excellent sensitivity at acidic pH, which facilitated the delivery of TQR and DOX into cells. Cellular experiments demonstrated that the pHSL/TQR/DOX 0.05 could almost restore the drug sensitivity of OVCAR8/ADR cells. Therefore, the pH sensitive liposome formulation pHSL/TQR/DOX 0.05 was very promising in treating resistant tumours.
Collapse
Affiliation(s)
- Yuqiong Xia
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China.
| | - Mei Fang
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Jingyu Dong
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Chunzhong Xu
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Zhen Liao
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Pengbo Ning
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Qi Zeng
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| |
Collapse
|
45
|
Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev 2018; 47:3574-3620. [PMID: 29479622 PMCID: PMC6386136 DOI: 10.1039/c7cs00877e] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide- and protein-nanoparticle conjugates have emerged as powerful tools for biomedical applications, enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies. Within this framework, we provide a comprehensive overview of the key sequences and structures utilised to provide biological and physical stability to nano-constructs, direct particles to their target and influence their cellular and tissue distribution, induce and control biological responses, and form polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised nano-drug delivery vehicles, imaging species, and active therapeutics.
Collapse
Affiliation(s)
- Christopher D Spicer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden.
| | | | | | | |
Collapse
|
46
|
El-Far SW, Helmy MW, Khattab SN, Bekhit AA, Hussein AA, Elzoghby AO. Phytosomal bilayer-enveloped casein micelles for codelivery of monascus yellow pigments and resveratrol to breast cancer. Nanomedicine (Lond) 2018; 13:481-499. [PMID: 29376765 DOI: 10.2217/nnm-2017-0301] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
AIM Multireservoir nanocarriers were fabricated for delivering antineoplastic drug cocktail from herbal and fungal origin. Monascus yellow pigments (MYPs), monascin and ankaflavin, were isolated from red-mold rice, and incorporated within casein micelles (CAS MCs) along with the herbal drug, resveratrol (RSV). Both drugs (MYPs and RSV) were simultaneously incorporated into the hydrophobic core of CAS MCs. Alternatively, MYPs-loaded CAS MCs were enveloped within RSV-phytosomal bilayer elaborating multireservoir nanocarriers. RESULTS Cytotoxicity studies confirmed the superiority of multireservoir nanocarriers against MCF-7 breast cancer cells. The in vivo antitumor efficacy was revealed by reduction of the tumor volume and growth biomarkers. CONCLUSION Multireservoir CAS nanocarriers for codelivery of both MYPs and RSV may be promising alternative to traditional breast cancer therapy.
Collapse
Affiliation(s)
- Shaymaa W El-Far
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Biotechnology, Institute of Graduate Studies & Research, Alexandria University, Alexandria 21526, Egypt
| | - Maged W Helmy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Sherine N Khattab
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Chemistry, Faculty of Science, Alexandria University, Alexandria 21321, Egypt
| | - Adnan A Bekhit
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed A Hussein
- Department of Biotechnology, Institute of Graduate Studies & Research, Alexandria University, Alexandria 21526, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
47
|
Fu Z, Han X, Du J, Han X, Liu W, Shao S, Liu X. Euphorbia lunulata extract acts on multidrug resistant gastric cancer cells to inhibit cell proliferation, migration and invasion, arrest cell cycle progression, and induce apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2018; 212:8-17. [PMID: 28811220 DOI: 10.1016/j.jep.2017.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The milky sap or the aboveground part of the plant Euphorbia lunulata has long been used by Chinese people to treat noncancerous growths and cancerous ailments but the specific mode of action and the action mechanism remain to be elucidated. AIM OF THE STUDY To investigate the effects of Euphorbia lunulata extract on cell proliferation, migration, invasion, cell cycle progression, and apoptosis of multidrug resistant human gastric cancer cells; To study the mechanism of apoptosis induction by Euphorbia lunulata extract in multidrug resistant human gastric cancer cells. MATERIALS AND METHODS The aboveground part of fresh Euphorbia lunulata plant was extracted first with ethanol and then with n-hexane. The aseptic extract at varying concentrations was used to treat the multidrug resistant human gastric cancer SGC7901/ADR cells. After treatment, the inhibition of cell proliferation was examined by MTT assay. The inhibitions of cell migration and invasion were detected by Transwell method. The alteration of cell cycle progression was studied by flow cytometry. The morphological changes of cell nuclei were observed with fluorescence microscopy following Hoechst 33258 staining and the apoptotic indexes were calculated. The activation of caspase enzymes was analyzed by spectrophotometry. The sub-cellular distribution of cytochrome complex and the expression of Bax and Bcl-2 proteins were determined by Western blot. RESULTS The proliferation, migration, and invasion of SGC7901/ADR cells were significantly inhibited by Euphorbia lunulata extract, which showed time- and dose-dependent manners. Cell cycle was arrested in G2/M phase. Significant apoptotic morphological changes were observed in the nuclei of the treated cells, and apoptotic indexes were increased significantly; these changes were diminished when Z-VAD-FMK, a caspase inhibitor, was also presented. The activities of caspase-3, caspase-8, and caspase-9 were increased. The sub-cellular distribution of cytochrome complex was altered----reduced in the mitochondria and increased in the cytoplasm. The expression of Bax was upregulated, while that of Bcl-2 was downregulated. CONCLUSION Euphorbia lunulata extract inhibited the proliferation, migration, and invasion of SGC7901/ADR cells, arrested cell cycle progression, and induced cell apoptosis; the mechanism of apoptosis induction involved both the extrinsic and the intrinsic pathways.
Collapse
Affiliation(s)
- Zhaoying Fu
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China; Institute of Molecular Biology and Immunology, Yanan University, Yanan, Shaanxi Province 716000, China.
| | - Xiaodong Han
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China; Institute of Molecular Biology and Immunology, Yanan University, Yanan, Shaanxi Province 716000, China.
| | - Juan Du
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China; Institute of Molecular Biology and Immunology, Yanan University, Yanan, Shaanxi Province 716000, China.
| | - Xiaoxiao Han
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China.
| | - Weipeng Liu
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China.
| | - Shumei Shao
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China.
| | - Xiaobin Liu
- School of Medicine, Yanan University, Yanan, Shaanxi Province 716000, China.
| |
Collapse
|
48
|
Agwa M, Elessawy FM, Hussein A, El Demellawy MA, Elzoghby AO, Abd El-Salam MH, Eldiwany AI. Development and validation of a robust analytical method to quantify both etoposide and prodigiosin in polymeric nanoparticles by reverse-phase high-performance liquid chromatography. ANALYTICAL METHODS 2018; 10:2272-2280. [DOI: 10.1039/c8ay00030a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Preparation of the ETP-PRO loaded B-CN nanocarrier (1) and analysis of the % of ETP and PRO released (2) using the validated HPLC method (3).
Collapse
Affiliation(s)
- M. M. Agwa
- Department of Natural & Microbial Products
- National Research Centre
- Cairo
- Egypt
- Pharmaceutical & Fermentation Industries Development Center (PFIDC)
| | - F. M. Elessawy
- Pharmaceutical & Fermentation Industries Development Center (PFIDC)
- City for Scientific Research & Technology Applications
- Alexandria
- Egypt
- College of Pharmacy and Nutrition
| | - A. Hussein
- Department of Biotechnology
- Institute of Graduate Studies and Research
- Alexandria University
- Alexandria
- Egypt
| | - M. A. El Demellawy
- Pharmaceutical & Fermentation Industries Development Center (PFIDC)
- City for Scientific Research & Technology Applications
- Alexandria
- Egypt
| | - A. O. Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL)
- Faculty of Pharmacy
- Alexandria University
- Alexandria
- Egypt
| | | | - A. I. Eldiwany
- Department of Natural & Microbial Products
- National Research Centre
- Cairo
- Egypt
| |
Collapse
|
49
|
Ye L, Miao M, Li S, Hao K. Nanosuspensions of a new compound, ER-β005, for enhanced oral bioavailability and improved analgesic efficacy. Int J Pharm 2017; 531:246-256. [PMID: 28847666 DOI: 10.1016/j.ijpharm.2017.08.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/31/2017] [Accepted: 08/21/2017] [Indexed: 11/19/2022]
Abstract
Estrogen receptor-β005 (ER-β005) is a novel compound developed by our group; however, its application has been greatly hindered due to its low solubility. A nanosuspension of insoluble drugs is a nanoscale colloidal dispersion that has extremely higher drug-loading compared with other nanomedicines. In this study, nanosuspensions of ER-β005 (Nano-ER-β005) stabilized by a food protein, β-casein (β-CN), were prepared via an antisolvent-precipitation method to improve oral absorption and thus promote therapeutic efficacy. Nano-ER-β005, which has a diameter of 110nm and drug-loading of 50%, was developed. Analyses of fluorescence and circular dichroism (CD) spectra demonstrated a strong interaction between β-CN and drug particles in Nano-ER-β005, indicating that β-CN is a potent nanosuspension stabilizer. The oral bioavailability of Nano-ER-β005 was 1.6-fold greater than that of raw drug particles. Additionally, ER-β005 was confirmed to have a strong therapeutic effect against pain reactions in animal models, and inhibition of this effect was significantly increased with Nano-ER-β005 treatment. In conclusion, by using β-CN as a stabilizer, nanosuspensions of ER-β005 were developed and oral absorption was enhanced. Moreover, ER-β005 is a powerful drug that inhibits pain reactions, and its therapeutic efficacy was markedly increased in the Nano-ER-β005.
Collapse
Affiliation(s)
- Ling Ye
- Key Lab of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; School of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 51006, PR China
| | - Mingxing Miao
- Key Lab of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China; National Experimental Teaching Demonstration Center of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Suning Li
- China National Center for Biotechnology Development, Beijing 100039, PR China.
| | - Kun Hao
- Key Lab of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
50
|
Potential of Casein as a Carrier for Biologically Active Agents. Top Curr Chem (Cham) 2017; 375:71. [PMID: 28712055 PMCID: PMC5511616 DOI: 10.1007/s41061-017-0158-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/29/2017] [Indexed: 02/07/2023]
Abstract
Casein is the collective name for a family of milk proteins. In bovine milk, casein comprises four peptides: αS1, αS2, β, and κ, differing in their amino acid, phosphorus and carbohydrate content but similar in their amphiphilic character. Hydrophilic and hydrophobic regions of casein show block distribution in the protein chain. Casein peptides carry negative charge on their surface as a result of phosphorylation and tend to bind nanoclusters of amorphous calcium phosphate. Due to these properties, in suitable conditions, casein molecules agglomerate into spherical micelles. The high content of casein in milk (2.75 %) has made it one of the most popular proteins. Novel research techniques have improved understanding of its properties, opening up new applications. However, casein is not just a dietary protein. Its properties promise new and unexpected applications in science and the pharmaceutical and functional food industries. One example is an encapsulation of health-related substances in casein matrices. This review discusses gelation, coacervation, self-assembly and reassembly of casein peptides as means of encapsulation. We highlight information on encapsulation of health-related substances such as drugs and dietary supplements inside casein micro- and nanoparticles.
Collapse
|