1
|
Jiang P, Jiang W, Li X, Zhu Q. Combination of Formononetin and Sulforaphane Natural Drug Repress the Proliferation of Cervical Cancer Cells via Impeding PI3K/AKT/mTOR Pathway. Appl Biochem Biotechnol 2024:10.1007/s12010-024-04873-y. [PMID: 38401043 DOI: 10.1007/s12010-024-04873-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 02/26/2024]
Abstract
Natural substances have been demonstrated to be an unrivalled source of anticancer drugs in the present era of pharmacological development. Plant-based substances, together with their derivatives through analogues, play a significant character in the treatment of cancer by altering the tumor microenvironment and several signaling pathways. In this study, it was investigated whether the natural drugs, formononetin (FN) and sulforaphane (SFN), when combined, assess the efficacy of inhibiting cervical cancer cell proliferation by impeding the PI3K/Akt/mTOR signaling pathway in HeLa cells. The cells were treated with the combination of FN and SFN (FN + SFN) in various concentrations (0-50 µM) for 24 h and then analyzed for various experiments. The combination of FN + SFN-mediated cytotoxicity was analyzed by MTT assay. DCFH-DA staining was used to assess the ROS measurement, and apoptotic changes were studied by dual (AO/EtBr) staining assays. Protein expressions of cell survival, cell cycle, proliferation, and apoptosis protein were evaluated by flow cytometry and western blotting. Results showed that the cytotoxicity of FN and SFN was determined to be around 23.7 µM and 26.92 µM, respectively. Combining FN and SFN causes considerable cytotoxicity in HeLa cells, with an IC50 of 21.6 µM after 24-h incubation. Additionally, HeLa cells treated with FN and SFN together showed increased apoptotic signals and considerable ROS generation. Consequently, by preventing the production of PI3K, AKT, and mToR-mediated regulation of proliferation and cell cycle-regulating proteins, the combined use of FN + SFN has been regarded as a chemotherapeutic medication. Further research will need to be done shortly to determine how effectively the co-treatment promotes apoptosis to employ them economically.
Collapse
Affiliation(s)
- Ping Jiang
- Gynaecology and Obstetrics, Yantai Mountain Hospital, Yantai, 264005, China
| | - Wei Jiang
- Medical Department, Jinan Maternity and Child Care Hospital, Jinan, 250000, China
| | - Xiujin Li
- Delivery Room, Jinan Maternity and Child Care Hospital, Jinan, 250000, China
| | - Qiuling Zhu
- Delivery Room, Jinan Maternity and Child Care Hospital, Jinan, 250000, China.
| |
Collapse
|
2
|
Zheng K, Zhao J, Wang Q, Zhao Y, Yang H, Yang X, He L. Design and Evaluation of Ginkgolides Gastric Floating Controlled Release Tablets Based on Solid Supersaturated Self-nanoemulsifying. AAPS PharmSciTech 2023; 25:7. [PMID: 38147267 DOI: 10.1208/s12249-023-02717-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023] Open
Abstract
Ginkgolides are receptor antagonist of platelet activating factor with great clinical prospect, but its application is limited by its low solubility, short half-life and poor alkaline environment stability. It is difficult to solve these problems with a single drug delivery system. In this study, supersaturated self-nanoemulsifying gastric floating tablets of ginkgolides were developed through the combination of solid supersaturated self-nanoemulsifying drug delivery system (solid S-SNEDDS) and gastric retentive floating drug delivery system (GFDDS) to solve these problems of ginkgolides. Solid S-SNEDDS was prepared by D-optimal mixture design, normalization method and single factor experiment. The properties of solid-S-SNEDDS were studied by TEM, PXRD, FT-IR, SEM and in vitro drug release profile. Then, the optimal formulation of stomach floating tablet was obtained through single factor experiment and center composite design, followed by the study of in vitro release, model and mechanism of release, in vitro buoyancy and kinetics of erosion and swelling. PXRD and FT-IR showed that the drug in solid S-SNEDDS existed in an amorphous manner and formed hydrogen bond with excipients. The results showed that the cumulative release of GA and GB in the optimal tablets was 96.12% and 92.57% higher than the simple tablets within 12 h. The release mechanism of the tablet was skeleton erosion and drug diffusion. In 12 h, the optimal tablets can float stably in vitro and release the drug at a constant rate, with a cumulative release of more than 80%. In summary, the combination of SNEDDS and GFDDS is a promising means to solve the problems of ginkgolides.
Collapse
Affiliation(s)
- Kai Zheng
- Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, 110016, China
| | - Jing Zhao
- Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, 110016, China
| | - Qiuli Wang
- Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, 110016, China
| | - Yuyang Zhao
- Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, 110016, China
| | - Husheng Yang
- Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, 110016, China
| | - Xinggang Yang
- Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, 110016, China.
| | - Lian He
- Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Shenyang, 110042, China.
- Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Shenyang, 110042, China.
| |
Collapse
|
3
|
Development, characterisation, and in vitro anti-tumor effect of self-microemulsifying drug delivery system containing polyphyllin I. Drug Deliv Transl Res 2023; 13:356-370. [PMID: 35877046 DOI: 10.1007/s13346-022-01212-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2022] [Indexed: 01/01/2023]
Abstract
Polyphyllin I (PPI), an effective active ingredient in Paris polyphylla, has a diverse set of pharmacological properties. However, due to its poor solubility and oral absorption, its application and development are limited. In the study, we were committed to improving the solubility of PPI by developing a self-microemulsifying drug delivery system of PPI (PPI-SMEDDS), screening the best preparation process, and evaluating the quality and the in vivo pharmacokinetics of PPI, and PPI-SMEDDS following oral administration to rats were also studied. In addition, the pharmacological activities against human lung adenocarcinoma cell A549 in vitro were assessed. The best formulation had 15.89% ethyl oleate, 47.38% Cremophor RH40, and 36.73% 1,2 propylene glycol. The produced PPI-SMEDDS was clear and transparent, with an average particle size of 24.51 nm and a zeta potential of -17.54 ± 0.51 mV. In vitro, the cumulative release rate of PPI-SMEDDS was nearly 80% within 2 h. PPI-SMEDDS had a substantially greater area under the curve than PPI following oral treatment in rats, and the relative bioavailability of PPI in rats was 278.99%. More importantly, the anti-tumor effect of PPI-SMEDDS in vitro was significantly greater than that of PPI. These findings suggested that PPI-SMEDDS has the potential to improve the solubility, oral bioavailability of PPI, and anti-tumor effect, laying the groundwork for future research on the new PPI dosage form.
Collapse
|
4
|
Chen S, Wang Z, Liu L, Li Y, Ni X, Yuan H, Wang C. Redox homeostasis modulation using theranostic AIE nanoparticles results in positive-feedback drug accumulation and enhanced drug penetration to combat drug-resistant cancer. Mater Today Bio 2022; 16:100396. [PMID: 36060105 PMCID: PMC9434132 DOI: 10.1016/j.mtbio.2022.100396] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
Drug-resistant cancers usually have multiple barriers to compromise the effect of therapies, of which multidrug-resistance (MDR) phenotype as the intracellular barrier and dense tumor matrix as the extracellular barrier, significantly contribute to the poor anticancer performance of current drug delivery systems (DDS). Here in this study, we fabricated a novel aggregation-induced emission (AIE)-active polymer capable of self-assembling into ultrasmall nanoparticles (∼20 nm) with D-alpha Tocopheryl Polyethylene Glycol Succinate (TPGS), for dual-encapsulating of doxorubicin (Dox) and sulforaphane (SFN) (AT/Dox/SFN). It revealed that redox homeostasis modulation of MDR cells (MCF-7/Adr) using AT/Dox/SFN can trigger mitochondria damage and ATP deficiency, which reverse the MDR phenotype of MCF-7/Adr cells to afford enhanced cellular uptake of both drug and DDS in a positive-feedback manner. The enhanced cellular drug accumulation further initiates the “neighboring effect” for improved drug penetration. Using this strategy, the growth of in vivo MCF-7/Adr tumors can be effectively inhibited at a low dosage (1/5) of doxorubicin (Dox) as compared to free Dox. In summary, we offer a new approach to overcome both the intracellular and extracellular barriers of drug-resistant cancers and elucidate the potential action mechanisms, which are beneficial for better cancer management. Redox homeostasis modulation in MDR cancer cell results in positive-feedback drug accumulation and enhanced drug penetration. Mitochondria damage and neighboring effect is responsible for MDR reversal and enhanced drug penetration, respectively. AT/Dox/SFN effectively inhibits in vivo MCF-7/Adr tumors at a low dosage (1/5) of doxorubicin (Dox) as compared to free Dox.
Collapse
Affiliation(s)
- Shaoqing Chen
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu 213003, China
| | - Ziyu Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, China
| | - Li Liu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Yuting Li
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu 213003, China
- Corresponding author. Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China.
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang, China
- Corresponding author.
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
- Corresponding author.
| |
Collapse
|
5
|
Rodrigues Arruda B, Mendes MGA, Freitas PGCD, Reis AVF, Lima T, Crisóstomo LCCF, Nogueira KAB, Pessoa C, Petrilli R, Eloy JO. Nanocarriers for delivery of taxanes: A review on physicochemical and biological aspects. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
6
|
Molecular Pathways Related to Sulforaphane as Adjuvant Treatment: A Nanomedicine Perspective in Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101377. [PMID: 36295538 PMCID: PMC9610969 DOI: 10.3390/medicina58101377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Because cancer is a multifactorial disease, it is difficult to identify the specific agents responsible for the disease's progression and development, but lifestyle and diet have been shown to play a significant role. Diverse natural compounds are demonstrating efficacy in the development of novel cancer therapies, including sulforaphane (1-isothiocyanate-4-(methylsulfinyl)butane), a compound found in broccoli and other cruciferous vegetables that promotes key biological processes such as apoptosis, cell cycle arrest, autophagy, and suppression of key signalling pathways such as the PI3K/AKT/mTOR pathway in breast cancer cells. However, one of the primary challenges with sulforaphane treatment is its low solubility in water and oral bioavailability. As a consequence, several investigations were conducted using this component complexed in nanoparticles, which resulted in superior outcomes when combined with chemotherapy drugs. In this study, we discuss the properties and benefits of sulforaphane in cancer therapy, as well as its ability to form complexes with nanomolecules and chemotherapeutic agents that synergize the antitumour response in breast cancer cells.
Collapse
|
7
|
Dangre PV, Shinde SB, Surana SJ, Jain PG, Chalikwar SS. Development and exploration on flowability of solid self-nanoemulsifying drug delivery system of morin hydrate. ADV POWDER TECHNOL 2022. [DOI: 10.1016/j.apt.2022.103716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Chaturvedi S, Garg A. A comprehensive review on novel delivery approaches for exemestane. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
9
|
Tcibulnikova A, Zemlyakova E, Slezhkin V, Samusev I, Bryukhanov V, Khankaev A, Artamonov D. Spectroscopy of triplet-excited complexes of oxygen with spruce cone molecules extract from picea abies in AOT micelles under combined photoexcitation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
10
|
Li T, Shi W, Yao J, Hu J, Sun Q, Meng J, Wan J, Song H, Wang H. Combinatorial nanococktails via self-assembling lipid prodrugs for synergistically overcoming drug resistance and effective cancer therapy. Biomater Res 2022; 26:3. [PMID: 35101154 PMCID: PMC8805243 DOI: 10.1186/s40824-022-00249-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/10/2022] [Indexed: 02/08/2023] Open
Abstract
Background Combinatorial systemic chemotherapy is a powerful treatment paradigm against cancer, but it is fraught with problems due to the emergence of chemoresistance and additive systemic toxicity. In addition, coadministration of individual drugs suffers from uncontrollable pharmacokinetics and biodistribution, resulting in suboptimal combination synergy. Methods Toward the goal of addressing these unmet medical issues, we describe a unique strategy to integrate multiple structurally disparate drugs into a self-assembling nanococktail platform. Conjugation of a polyunsaturated fatty acid (e.g., linoleic acid) with two chemotherapies generated prodrug entities that were miscible with tunable drug ratios for aqueous self-assembly. In vitro and in vivo assays were performed to investigate the mechanism of combinatorial nanococktails in mitigating chemoresistance and the efficacy of nanotherapy. Results The coassembled nanoparticle cocktails were feasibly fabricated and further refined with an amphiphilic matrix to form a systemically injectable and PEGylated nanomedicine with minimal excipients. The drug ratio incorporated into the nanococktails was optimized and carefully examined in lung cancer cells to maximize therapeutic synergy. Mechanistically, subjugated resistance by nanococktail therapy was achieved through the altered cellular uptake pathway and compromised DNA repair via the ATM/Chk2/p53 cascade. In mice harboring cisplatin-resistant lung tumor xenografts, administration of the nanococktail outperformed free drug combinations in terms of antitumor efficacy and drug tolerability. Conclusion Overall, our study provides a facile and cost-effective approach for the generation of cytotoxic nanoparticles to synergistically treat chemoresistant cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s40824-022-00249-7.
Collapse
|
11
|
Kamal MM, Akter S, Al Hagbani T, Salawi A, Nazzal S. Sustained release of curcumin self-emulsifying drug delivery system (SEDDS) from solvent-cast Soluplus ® films. Pharm Dev Technol 2021; 26:1102-1109. [PMID: 34645368 DOI: 10.1080/10837450.2021.1993912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The objective of the present study was to investigate the feasibility of formulating and loading Curcumin SEDDS (Self-Emulsified Drug Delivery Systems) into films made from Soluplus® as the film-forming polymer. Films with up to 30% of Curcumin SEDDS were prepared by the solvent casting technique and analyzed for their mechanical and dissolution properties. A nine-run, two-factor, three-level factorial design was utilized to investigate the effect of SEDDS load (10, 20, and 30% w/w) and film thickness (10, 25, and 40 mils) on the tensile strength, elongation, and adhesiveness of the films. The dissolution profile of the films was also investigated by a USP Type 1 method. SEDDS loading was found to plasticize Soluplus® and to yield transparent films of good mechanical properties. Increasing SEDDS load, however, was found to reduce the tensile strength of the films, while increasing their adhesiveness and elongation. On the other hand, while an increase in film thickness was found to increase the tensile strength of the films, it reduced the elongation capacity of the films. Loading SEDDS into Soluplus® films was also found to sustain their release over 6 h, where a significant delay in release was found at lower SEDDS loads. This study demonstrated that Soluplus® can be used not only to formulate SEDDS into polymeric films but also to sustain their release over an extended time.
Collapse
Affiliation(s)
- Mohammad M Kamal
- Fresenius Kabi USA, Melrose Park, IL, USA.,School of Basic Pharmaceutical & Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Sharmin Akter
- Shaheed Ziaur Rahman Medical College and Hospital, Bogura, Bangladesh
| | - Turki Al Hagbani
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Ahmad Salawi
- College of Pharmacy, Jazan University, Gizan, Saudi Arabia
| | - Sami Nazzal
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX, USA
| |
Collapse
|
12
|
Buya AB, Beloqui A, Memvanga PB, Préat V. Self-Nano-Emulsifying Drug-Delivery Systems: From the Development to the Current Applications and Challenges in Oral Drug Delivery. Pharmaceutics 2020; 12:E1194. [PMID: 33317067 PMCID: PMC7764143 DOI: 10.3390/pharmaceutics12121194] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/31/2022] Open
Abstract
Approximately one third of newly discovered drug molecules show insufficient water solubility and therefore low oral bio-availability. Self-nano-emulsifying drug-delivery systems (SNEDDSs) are one of the emerging strategies developed to tackle the issues associated with their oral delivery. SNEDDSs are composed of an oil phase, surfactant, and cosurfactant or cosolvent. SNEDDSs characteristics, their ability to dissolve a drug, and in vivo considerations are determinant factors in the choice of SNEDDSs excipients. A SNEDDS formulation can be optimized through phase diagram approach or statistical design of experiments. The characterization of SNEDDSs includes multiple orthogonal methods required to fully control SNEDDS manufacture, stability, and biological fate. Encapsulating a drug in SNEDDSs can lead to increased solubilization, stability in the gastro-intestinal tract, and absorption, resulting in enhanced bio-availability. The transformation of liquid SNEDDSs into solid dosage forms has been shown to increase the stability and patient compliance. Supersaturated, mucus-permeating, and targeted SNEDDSs can be developed to increase efficacy and patient compliance. Self-emulsification approach has been successful in oral drug delivery. The present review gives an insight of SNEDDSs for the oral administration of both lipophilic and hydrophilic compounds from the experimental bench to marketed products.
Collapse
Affiliation(s)
- Aristote B. Buya
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Avenue Mounier 73, B1.73.12, 1200 Brussels, Belgium; (A.B.B.); (A.B.)
- Pharmaceutics and Phytopharmaceutical Drug Development Research Group, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI BP 212, Democratic Republic of the Congo;
| | - Ana Beloqui
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Avenue Mounier 73, B1.73.12, 1200 Brussels, Belgium; (A.B.B.); (A.B.)
| | - Patrick B. Memvanga
- Pharmaceutics and Phytopharmaceutical Drug Development Research Group, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa XI BP 212, Democratic Republic of the Congo;
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université Catholique de Louvain, Avenue Mounier 73, B1.73.12, 1200 Brussels, Belgium; (A.B.B.); (A.B.)
| |
Collapse
|
13
|
Inclusion Complex of Docetaxel with Sulfobutyl Ether β-Cyclodextrin: Preparation, In Vitro Cytotoxicity and In Vivo Safety. Polymers (Basel) 2020; 12:polym12102336. [PMID: 33066097 PMCID: PMC7601231 DOI: 10.3390/polym12102336] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 01/08/2023] Open
Abstract
Docetaxel (DTX), as a first-line anti-tumor drug, has been studied for decades for its diverse bioactivities. However, DTX presents poor solubility in water, low bioavailability and serious toxic side effects which has hindered its application in the clinic. To address these problems, docetaxel-sulfobutyl ether-β-cyclodextrin inclusion complex (DTX-SBE-β-CD) was prepared successfully by saturated aqueous solution method. Sulfobutyl ether β-cyclodetrin (SBE-β-CD) is used as delivery material. For this study, the inclusion complex of docetaxel with sulfobutyl ether β-cyclodetrin (DTX-SBE-β-CD) was prepared and optimized its properties to enhance the cytotoxicity of cancer cells. A large number of physical characterization results showed that DTX-SBE-β-CD inclusion complex was successfully prepared by saturated aqueous solution method. DTX-SBE-β-CD inclusion complex was optimized by Central Composite Design. DTX-SBE-β-CD had an inhibitory effect on the in vitro determination of MCF-7 and HepG2 cells by MTT assay. Pharmacokinetic studies were carried out on male Sprague–Dawley rats by tail injection, including the distribution, metabolism and elimination of DTX-SBE-β-CD in vivo. In the experimental study of inhibition of cancer cells, DTX and DTX-SBE-β-CD showed apparent concentration-dependent inhibitory actions on tumor cells and the inhibition of DTX-SBE-β-CD group was more obvious.
Collapse
|
14
|
Abid M, Naveed M, Azeem I, Faisal A, Faizan Nazar M, Yameen B. Colon specific enzyme responsive oligoester crosslinked dextran nanoparticles for controlled release of 5-fluorouracil. Int J Pharm 2020; 586:119605. [DOI: 10.1016/j.ijpharm.2020.119605] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022]
|
15
|
Advances in anti-breast cancer drugs and the application of nano-drug delivery systems in breast cancer therapy. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101662] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Development and characterization of curcumin-loaded solid self-emulsifying drug delivery system (SEDDS) by spray drying using Soluplus® as solid carrier. POWDER TECHNOL 2020. [DOI: 10.1016/j.powtec.2020.05.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
17
|
Sulforaphane as an anticancer molecule: mechanisms of action, synergistic effects, enhancement of drug safety, and delivery systems. Arch Pharm Res 2020; 43:371-384. [PMID: 32152852 DOI: 10.1007/s12272-020-01225-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/02/2020] [Indexed: 02/08/2023]
Abstract
Sulforaphane is an isothiocyanate compound that has been derived from cruciferous vegetables. It was shown in numerous studies to be active against multiple cancer types including pancreatic, prostate, breast, lung, cervical, and colorectal cancers. Sulforaphane exerts its therapeutics action by a variety of mechanisms, such as by detoxifying carcinogens and oxidants through blockage of phase I metabolic enzymes, and by arresting cell cycle in the G2/M and G1 phase to inhibit cell proliferation. The most striking observation was the ability of sulforaphane to potentiate the activity of several classes of anticancer agents including paclitaxel, docetaxel, and gemcitabine through additive and synergistic effects. Although a good number of reviews have reported on the mechanisms by which sulforaphane exerts its anticancer activity, a comprehensive review on the synergistic effect of sulforaphane and its delivery strategies is lacking. Therefore, the aim of the current review was to provide a summary of the studies that have been reported on the activity enhancement effect of sulforaphane in combination with other anticancer therapies. Also provided is a summary of the strategies that have been developed for the delivery of sulforaphane.
Collapse
|
18
|
Kamal MM, Nazzal S. Development and validation of a HPLC-UV method for the simultaneous detection and quantification of paclitaxel and sulforaphane in lipid based self-microemulsifying formulation. J Chromatogr Sci 2020; 57:931-938. [PMID: 31642896 DOI: 10.1093/chromsci/bmz068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/11/2019] [Accepted: 07/15/2019] [Indexed: 11/13/2022]
Abstract
Paclitaxel (PTX) and sulforaphane (SFN) are known anticancer molecules. Their activity was found to be potentiated when tested concurrently. Only recently, however, a novel SFN enabled PTX self-microemulsifying formulation (SMEDDS) was developed for their simultaneous delivery. This necessitated the development of an analytical method for the simultaneous detection and quantitation of PTX and SFN. In this study, a simple and sensitive isocratic high performance liquid chromatography-ultraviolet (HPLC-UV) analytical method was developed and validated per International Conference on Harmonization guidelines to satisfy this objective. Its application was demonstrated when quantifying the amount of PTX and SFN released from the SMEDDS in various dissolution media. The separation of the analytes was performed with the aid of a reversed phase C18 column at ambient temperature using a 60:40 mixture of acetonitrile and KH2PO4 buffer (pH 5.0) as the mobile phase. PTX and SFN peaks were detected at 202 nm with high resolution without interference from excipients. This method showed linearity within 2.5-100 μg/mL range with r2 > 0.999. The limit of detection and lower limit of quantitation were 0.1638 and 0.4964 μg/mL for PTX and 0.4419 and 1.3389 μg/mL for SFN, respectively. A total of 98-101% of the injected samples was recovered with RSD of 0.06-0.68% indicating the suitability of the method for the simultaneous detection and quantitation of the molecules in dissolution media.
Collapse
Affiliation(s)
- Mohammad M Kamal
- School of Basic Pharmaceutical & Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA.,Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sami Nazzal
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX 75235, USA
| |
Collapse
|
19
|
Rafiei H, Ashrafizadeh M, Ahmadi Z. MicroRNAs as novel targets of sulforaphane in cancer therapy: The beginning of a new tale? Phytother Res 2020; 34:721-728. [PMID: 31972874 DOI: 10.1002/ptr.6572] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/30/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Effective management and treatment of cancer depend on developing novel antitumor drugs with the capability of targeting various molecular pathways. Identification and subsequent targeting of these pathways are of importance in cancer therapy. MicroRNAs (miRNAs) are small noncoding RNA molecules responsible for post-transcriptional regulation of genes. Notably, miRNAs participate in a number of biological processes such as proliferation, apoptosis, differentiation, and cell cycle regulation. So, any impairment in the expression and function of miRNAs is associated with development of disorders, particularly cancer. Naturally occurring nutraceutical compounds have attracted much attention due to their great antitumor activity. Among them, sulforaphane isolated from Brassica oleracea (broccoli) is of interest due to its therapeutic and biological activities such as antidiabetic, antioxidant, anti-inflammatory, hepatoprotection, and cardiprotection. Sulforaphane has demonstrated great antitumor activity and is able to significantly inhibit proliferation, viability, migration, malignancy, and epithelial-to-mesenchymal transition of cancer cells. These antitumor effects have widely been investigated, and it appears that there is a need for a precise review to demonstrate the molecular pathway that sulforaphane follows to exert its antitumor activity. At the present review, we focus on the modulatory impact of sulforaphane on miRNAs and exhibit that how various miRNAs in different cancers are regulated by sulforaphane.
Collapse
Affiliation(s)
- Hossein Rafiei
- Department of Biology, Faculty of Sciences, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zahra Ahmadi
- Department of basic science, Islamic Azad university, Shoushtar Branch, Shoushtar, Iran
| |
Collapse
|
20
|
Shakhwar S, Darwish R, Kamal MM, Nazzal S, Pallerla S, Abu Fayyad A. Development and evaluation of paclitaxel nanoemulsion for cancer therapy. Pharm Dev Technol 2019; 25:510-516. [PMID: 31858867 DOI: 10.1080/10837450.2019.1706564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tocosol™ is a tocopherol-based paclitaxel (PTX) nanoemulsion consisting of α-tocopherol (α-T) isomer of vitamin E as a solubilizer and vitamin E TPGS as the primary emulsifier. Despite its positive attributes in early clinical studies, it failed the pivotal phase III clinical trials. The long-term goal of this work was to reformulate Tocosol™. In this study, Tocosol™ formulation was optimized by replacing the α-T isomer with the more pharmacological active isomer γ-tocotrienol (γ-T3), and the surfactant vitamin E TPGS was replaced with in-house designed PEGylated γ-T3 surfactant. The reformulated paclitaxel γ-T3/PEGylated γ-T3 -based nanoemulsion was significantly more active against pancreatic tumor cell lines than α-T/Vitamin E TPGS based formulation (IC50 = 0.5 μM and 1.1 μM, respectively). Furthermore, the reformulated product showed an average size of 220 ± 6 nm with surface charge equal to -42 ± 2 mV. The optimized product was physically and chemically stable over 6 months per ICH storage condition guidelines.
Collapse
Affiliation(s)
- Sara Shakhwar
- College of Pharmacy, Chicago State University, Chicago, IL, USA
| | - Rana Darwish
- College of Pharmacy, Chicago State University, Chicago, IL, USA
| | - Mohammad M Kamal
- College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Sami Nazzal
- School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX, USA
| | | | | |
Collapse
|
21
|
Melrose J. The Glucosinolates: A Sulphur Glucoside Family of Mustard Anti-Tumour and Antimicrobial Phytochemicals of Potential Therapeutic Application. Biomedicines 2019; 7:biomedicines7030062. [PMID: 31430999 PMCID: PMC6784281 DOI: 10.3390/biomedicines7030062] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 12/13/2022] Open
Abstract
This study reviewed aspects of the biology of two members of the glucosinolate family, namely sinigrin and glucoraphanin and their anti-tumour and antimicrobial properties. Sinigrin and glucoraphanin are converted by the β-sulphoglucosidase myrosinase or the gut microbiota into their bioactive forms, allyl isothiocyanate (AITC) and sulphoraphanin (SFN) which constitute part of a sophisticated defence system plants developed over several hundred million years of evolution to protect them from parasitic attack from aphids, ticks, bacteria or nematodes. Delivery of these components from consumption of cruciferous vegetables rich in the glucosinolates also delivers many other members of the glucosinolate family so the dietary AITCs and SFN do not act in isolation. In vitro experiments with purified AITC and SFN have demonstrated their therapeutic utility as antimicrobials against a range of clinically important bacteria and fungi. AITC and SFN are as potent as Vancomycin in the treatment of bacteria listed by the World Health Organisation as antibiotic-resistant “priority pathogens” and also act as anti-cancer agents through the induction of phase II antioxidant enzymes which inactivate potential carcinogens. Glucosinolates may be useful in the treatment of biofilms formed on medical implants and catheters by problematic pathogenic bacteria such as Pseudomonas aeruginosa and Staphylococcus aureus and are potent antimicrobials against a range of clinically important bacteria and fungi. The glucosinolates have also been applied in the prevention of bacterial and fungal spoilage of food products in advanced atmospheric packaging technology which improves the shelf-life of these products.
Collapse
Affiliation(s)
- James Melrose
- Honorary Senior Research Associate, Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia.
- Adjunct Professor, Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
- Sydney Medical School, Northern, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia.
| |
Collapse
|
22
|
Yang Z, Wang Y, Cheng J, Shan B, Wang Y, Wang R, Hou L. Solid self-microemulsifying drug delivery system of Sophoraflavanone G: Prescription optimization and pharmacokinetic evaluation. Eur J Pharm Sci 2019; 136:104953. [DOI: 10.1016/j.ejps.2019.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 05/08/2019] [Accepted: 06/04/2019] [Indexed: 01/02/2023]
|
23
|
Desai P, Thakkar A, Ann D, Wang J, Prabhu S. Loratadine self-microemulsifying drug delivery systems (SMEDDS) in combination with sulforaphane for the synergistic chemoprevention of pancreatic cancer. Drug Deliv Transl Res 2019; 9:641-651. [PMID: 30706304 PMCID: PMC6827432 DOI: 10.1007/s13346-019-00619-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pancreatic cancer (PC), currently the third leading cause of cancer-related deaths in the USA, is projected to become the second leading cause, behind lung cancer, by 2020. The increasing incidence, low survival rate, and limited treatment opportunities necessitate the use of alternative approaches such as chemoprevention, to tackle PC. In this study, we report significant synergistic chemoprevention efficacy for the first time from a low-dose combination of a classical antihistaminic drug, Loratadine (LOR) and a neutraceutical compound, Sulforaphane (SFN) using a self-microemulsifying drug delivery system (SMEDDS) formulation. The formulation was developed using Quality by Design approach (globule size, 95.13 ± 7.9 nm; PDI, 0.17 ± 0.04) and revealed significant (p < 0.05) enhancement in the in vitro dissolution profile confirming the enhanced solubility of BCS class II drug LOR with SMEDDS formulation. The LOR-SFN combination revealed ~ 40-fold reduction in IC50 concentration compared to LOR alone in MIA PaCa-2 and Panc-1 cell lines respectively, confirming the synergistic enhancement in chemoprevention. Further, the nanoformulation resulted in ~ 7-fold and ~ 11-fold reduction in IC50 values compared to LOR-SFN combination. Hence, our studies successfully demonstrate that a unique low-dose combination of LOR encapsulated within SMEDDs with SFN shows significantly enhanced chemopreventive efficacy of PC.
Collapse
Affiliation(s)
- Preshita Desai
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Arvind Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, 04103, USA
| | - David Ann
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Jeffrey Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Sunil Prabhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, 91766, USA.
| |
Collapse
|
24
|
Okamoto Y, Taguchi K, Sakuragi M, Imoto S, Yamasaki K, Otagiri M. Preparation, Characterization, and in Vitro/in Vivo Evaluation of Paclitaxel-Bound Albumin-Encapsulated Liposomes for the Treatment of Pancreatic Cancer. ACS OMEGA 2019; 4:8693-8700. [PMID: 31459959 PMCID: PMC6649292 DOI: 10.1021/acsomega.9b00537] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/09/2019] [Indexed: 05/28/2023]
Abstract
Paclitaxel (PTX)-loaded liposomes were developed with the goal of enhancing the effects of cancer treatment. Although loading substances into the lipid membrane of liposome cause some destabilization of the lipid membrane, PTX was nearly exclusively embedded in the lipid membrane of liposomes, due to its low water solubility. Hydrophobic drugs can be encapsulated into the inner core of bovine serum albumin (BSA)-encapsulated liposomes (BSA-liposome) via noncovalent binding to albumin. Since PTX is able to noncovalently bind to albumin, we attempted to prepare PTX-loaded BSA-liposome (PTX-BSA-liposome). The amount of PTX loaded in the BSA-liposome could be increased substantially by using ethanol, since ethanol increases PTX solubility in BSA solutions via prompting the binding PTX to BSA. On the basis of the results of transmission electron microscopy and small-angle X-ray scattering, PTX-BSA-liposome formed unilamellar vesicles that were spherical in shape and the PTX was encapsulated into the inner aqueous core of the liposome as a form of PTX-BSA complex. In addition, the PTX-BSA-liposome, as well as nab-PTX, showed cytotoxicity against human pancreatic cancer cells, AsPC-1 cells, in a PTX concentration-dependent manner. The in vivo antitumor effect of PTX-BSA-liposomes was also observed in a mouse model that had been subcutaneously inoculated with pancreatic cancer cells by virtue of its high accumulation at the tumor site via the enhanced permeability retention effect. These results suggest that PTX-BSA-liposomes have the potential for serving as a novel PTX preparation method for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yuko Okamoto
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Kazuaki Taguchi
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
- Faculty
of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Mina Sakuragi
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Shuhei Imoto
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Department of Nanoscience, and DDS Research Institute, Sojo University, Kumamoto 860-0082, Japan
| |
Collapse
|
25
|
Flores-Villaseñor SE, Peralta-Rodríguez RD, Padilla-Vaca F, Meléndez-Ortiz HI, Ramirez-Contreras JC, Franco B. Preparation of Peppermint Oil-Based Nanodevices Loaded with Paclitaxel: Cytotoxic and Apoptosis Studies in HeLa Cells. AAPS PharmSciTech 2019; 20:198. [PMID: 31127389 DOI: 10.1208/s12249-019-1399-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/17/2019] [Indexed: 01/08/2023] Open
Abstract
In this work, several normal, oil-in-water (o/w) microemulsions (MEs) were prepared using peppermint essential oil, jojoba oil, trans-anethole, and vitamin E as oil phases to test their capacity to load paclitaxel (PTX). Initially, pseudo-ternary partial phase diagrams were constructed in order to find the normal microemulsion region using d-α-tocopherol polyethylene glycol 1000 succinate (TPGS-1000) as surfactant and isobutanol (iso-BuOH) as co-surfactant. Selected ME formulations were loaded with PTX reaching concentrations of 0.6 mg mL-1 for the peppermint oil and trans-anethole MEs, while for the vitamin E and jojoba oil MEs, the maximum concentration was 0.3 mg mL-1. The PTX-loaded MEs were stable according to the results of heating-cooling cycles and mechanical force (centrifugation) test. Particularly, drug release profile for the PTX-loaded peppermint oil ME (MEPP) showed that ∼ 90% of drug was released in the first 48 h. Also, MEPP formulation showed 70% and 90% viability reduction on human cervical cancer (HeLa) cells after 24 and 48 h of exposure, respectively. In addition, HeLa cell apoptosis was confirmed by measuring caspase activity and DNA fragmentation. Results showed that the MEPP sample presented a major pro-apoptotic capability by comparing with the unloaded PTX ME sample.
Collapse
|
26
|
Wang Y, Zhou Y, Zheng Z, Li J, Yan Y, Wu W. Sulforaphane metabolites reduce resistance to paclitaxel via microtubule disruption. Cell Death Dis 2018; 9:1134. [PMID: 30429459 PMCID: PMC6235886 DOI: 10.1038/s41419-018-1174-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/16/2022]
Abstract
Long treatment with paclitaxel (PTX) might increase resistance and side-effects causing a failure in cancer chemotherapy. Here we uncovered that either sulforaphane-cysteine (SFN-Cys) or sulforaphane-N-acetyl-cysteine (SFN-NAC) induced apoptosis via phosphorylated ERK1/2-mediated upregulation of 26 S proteasome and Hsp70, and downregulation of βIII-tubulin, XIAP, Tau, Stathmin1 and α-tubulin causing microtubule disruption in human PTX-resistant non-small cell lung cancer (NSCLC) cells. Knockdown of either βIII-tubulin or α-tubulin via siRNA increased cell sensitivity to PTX, indicating that these two proteins help cells increase the resistance. Tissue microarray analysis showed that overexpression of βIII-tubulin correlated to NSCLC malignant grading. Immunofluorescence staining also showed that SFN metabolites induced a nest-like microtubule protein distribution with aggregation and disruption. Co-immunoprecipitation showed that SFN metabolites reduced the interaction between βIII-tubulin and Tau, and that between α-tubulin and XIAP. The combination of PTX with SFN metabolites decreased the resistance to PTX, and doses of both PTX and SFN metabolites, and enhanced apoptosis resulting from activated Caspase-3-caused microtubule degradation. Importantly, the effective dose of SFN metabolites combined with 20 nM PTX will be low to 4 μM. Thus, we might combine SFN metabolites with PTX for preclinical trial. Normally, more than 20 μM SFN metabolites only leading to apoptosis for SFN metabolites hindered their applications. These findings will help us develop a low-resistance and high-efficiency chemotherapy via PTX/SFN metabolites combination.
Collapse
Affiliation(s)
- Yalin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Zhongnan Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Juntao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Yuting Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China
| | - Wei Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China.
- Beijing Key Laboratory of Tumor Invasion and Metastasis Research, Capital Medical University, Beijing, P.R. China.
- Institute of Brain Tumor, Beijing Institute for Brain Disorders, Capital Medical University, No. 10, Xitoutiao, You An Men Wai Ave., Feng Tai District, Beijing, 100069, P.R. China.
| |
Collapse
|
27
|
Tan T, Wang Y, Wang H, Cao H, Wang Z, Wang J, Li J, Li Y, Zhang Z, Wang S. Apoferritin nanocages loading mertansine enable effective eradiation of cancer stem-like cells in vitro. Int J Pharm 2018; 553:201-209. [PMID: 30339945 DOI: 10.1016/j.ijpharm.2018.10.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/23/2018] [Accepted: 10/15/2018] [Indexed: 12/21/2022]
Abstract
Cancer stem-like cells (CSCs) are proposed to be responsible for tumor metastasis, resistance and relapse after therapy, but are unable to be eliminated by many current therapies. Herein, we report that the apoferritin nanocages loading cytotoxic mertansine (M-AFN) can significantly improve their uptake in CSCs-enriched tumorspheres and effectively eradicate CSCs in tumorspheres for anticancer therapy. M-AFN were uniformly nanocage structures with the mean diameter of 11.26 ± 2.58 nm and the loading capacity of 0.62%. In the CSCs-enriched tumorsphere model, M-AFN could be preferentially internalized by tumorsphere cells and the average half-inhibitory concentration (IC50) of M-AFN was obviously reduced by 5.46-fold when comparing to the parent 4T1 breast cancer cells. Moreover, both the already existing tumorspheres and the formation of secondary tumorspheres were drastically disrupted by M-AFN, but barely impacted by mertansine alone. The flow cytometer analysis showed the CSCs fractions in tumorspheres were considerably reduced by the M-AFN treatment. Therefore, the apoferritin nanocages represent an encouraging nanoplatform to eradicate CSCs for effective anticancer therapy.
Collapse
Affiliation(s)
- Tao Tan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Yuqi Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Hong Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haiqiang Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwan Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jing Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Siling Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| |
Collapse
|
28
|
Ediriweera MK, Tennekoon KH, Samarakoon SR. In vitro assays and techniques utilized in anticancer drug discovery. J Appl Toxicol 2018; 39:38-71. [DOI: 10.1002/jat.3658] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Meran Keshawa Ediriweera
- Institute of Biochemistry, Molecular Biology and Biotechnology; University of Colombo; Colombo 03 Sri Lanka
| | - Kamani Hemamala Tennekoon
- Institute of Biochemistry, Molecular Biology and Biotechnology; University of Colombo; Colombo 03 Sri Lanka
| | | |
Collapse
|
29
|
Ujhelyi Z, Vecsernyés M, Fehér P, Kósa D, Arany P, Nemes D, Sinka D, Vasvári G, Fenyvesi F, Váradi J, Bácskay I. Physico-chemical characterization of self-emulsifying drug delivery systems. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 27:81-86. [PMID: 30103867 DOI: 10.1016/j.ddtec.2018.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 01/21/2023]
Abstract
Self-emulsifying drug delivery systems (SEDDS) are regarded as a potential implement for oral delivery of water insoluble APIs to overcome their poor and irregular bioavailability. The correlation between the physicochemical parameters and the behavior of self-emulsifying drug delivery systems was established. The objective of this study was to summarize these physicochemical factors characterized SEDDS. Determination of self-emulsification process and ternary phase diagram are the basis of preparations. The position of APIs in SEDDS inclusion can be determined by dye solubilisation test. The end point of self-emulsification was controlled by turbimetric evaluation. Optimisation of droplet size and zeta potential are crucial parameters because they can influence i.e. the dissolution rate of APIs and the stability of SEDDS. Besides the basic methods in the characterization of SEDDS such as dispersibility tests, turbidimetric evaluation, viscosity tests, determinations with complex instruments such as photon correlation spectroscopy or dynamic light-scattering, electro kinetic potential measurement, non-destructive spectroscopic techniques (LFDS, FTIR, RS) and various microscopic techniques (SEM, PLM, EDS) has also been described.
Collapse
Affiliation(s)
- Zoltán Ujhelyi
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Miklós Vecsernyés
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Pálma Fehér
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Dóra Kósa
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Petra Arany
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Dániel Nemes
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Dávid Sinka
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Gábor Vasvári
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Ferenc Fenyvesi
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Judit Váradi
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Ildikó Bácskay
- University of Debrecen, Department of Pharmaceutical Technology, Nagyerdei krt. 98., H-4032 Debrecen, Hungary.
| |
Collapse
|
30
|
Kamal MM, Nazzal S. Development of a new class of sulforaphane-enabled self-emulsifying drug delivery systems (SFN-SEDDS) by high throughput screening: A case study with curcumin. Int J Pharm 2018; 539:147-156. [DOI: 10.1016/j.ijpharm.2018.01.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 10/17/2022]
|
31
|
Chirumbolo S, Bjørklund G. Sulforaphane and 5-fluorouracil synergistically inducing autophagy in breast cancer: A possible role for the Nrf2-Keap1-ARE signaling? Food Chem Toxicol 2018; 112:414-415. [PMID: 29305271 DOI: 10.1016/j.fct.2017.12.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, 37134, Verona Italy.
| | - Geir Bjørklund
- Council for Nutritional and Environmental Sciences, (CONEM), Mo i Rana, Norway
| |
Collapse
|