1
|
Tiwari R, Kolli M, Chauhan S, Yallapu MM. Tabletized Nanomedicine: From the Current Scenario to Developing Future Medicine. ACS NANO 2024; 18:11503-11524. [PMID: 38629397 DOI: 10.1021/acsnano.4c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The limitations of conventional therapeutic treatments prevailed in the development of nanotechnology-based medical formulations, termed nanomedicine. Nanomedicine is an advanced medicine that often consists of therapeutic agent(s) embedded in biodegradable or biocompatible nanomaterial-based formulations. Among nanomedicine approaches, tablet (oral) nanomedicine is still under development. In tabletized nanomedicine, the dynamic interplay between nanoformulations and the intricate milieu of the gastrointestinal tract simulates a pivotal role, particularly accentuating the influence exerted upon the luminal, mucosal, and epithelial cells. In this work, we document the perspectives and opportunities of nanoformulations toward the development of tabletized nanomedicine. This review also unveils the notion of integrating nanomedicine within a tablet formulation, which facilitates the controlled release of drugs, biomolecules, and agent(s) from the formulation to achieve a better therapeutic response. Finally, an attempt was made to explore current trends in nanomedicine technology such as bacteriophage, probiotic, and oligonucleotide tabletized nanomedicine and the combination of nanomedicine with imaging agents, i.e., nanotheranostics.
Collapse
Affiliation(s)
- Rahul Tiwari
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Meghana Kolli
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Sumeet Chauhan
- Department of Biology, College of Science, University of Texas Rio Grande Valley, Edinburg, Texas 78539, United States
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
2
|
Elhoseny SM, Saleh NM, Meshali MM. Self-Nanoemulsion Intrigues the Gold Phytopharmaceutical Chrysin: In Vitro Assessment and Intrinsic Analgesic Effect. AAPS PharmSciTech 2024; 25:54. [PMID: 38443653 DOI: 10.1208/s12249-024-02767-0] [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: 11/26/2023] [Accepted: 02/10/2024] [Indexed: 03/07/2024] Open
Abstract
Chrysin is a natural flavonoid with a wide range of bioactivities. Only a few investigations have assessed the analgesic activity of chrysin. The lipophilicity of chrysin reduces its aqueous solubility and bioavailability. Hence, self-nanoemulsifying drug delivery systems (SNEDDS) were designed to overcome this problem. Kollisolv GTA, Tween 80, and Transcutol HP were selected as oil, surfactant, and cosurfactant, respectively. SNEDDS A, B, and C were prepared, loaded with chrysin (0.1%w/w), and extensively evaluated. The optimized formula (B) encompasses 25% Kollisolv GTA, 18.75% Tween 80, and 56.25% Transcutol HP was further assessed. TEM, in vitro release, and biocompatibility towards the normal oral epithelial cell line (OEC) were estimated. Brain targeting and acetic acid-induced writhing in a mouse model were studied. After testing several adsorbents, powdered SNEDDS B was formulated and evaluated. The surfactant/cosurfactant (S/CoS) ratio of 1:3 w/w was appropriate for the preparation of SNEDDS. Formula B exhibited instant self-emulsification, spherical nanoscaled droplets of 155.4 ± 32.02 nm, and a zeta potential of - 12.5 ± 3.40 mV. The in vitro release proved the superiority of formula B over chrysin suspension (56.16 ± 10.23 and 9.26 ± 1.67%, respectively). The biocompatibility of formula B towards OEC was duplicated (5.69 ± 0.03 µg/mL). The nociceptive pain was mitigated by formula B more efficiently than chrysin suspension as the writhing numbers reduced from 8.33 ± 0.96 to 0 after 60 min of oral administration. Aerosil R972 was selected as an adsorbent, and its chemical compatibility was confirmed. In conclusion, our findings prove the therapeutic efficacy of chrysin self-nanoemulsion as a potential targeting platform to combat pain.
Collapse
Affiliation(s)
- Samar Mohamed Elhoseny
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Noha Mohamed Saleh
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mahasen Mohamed Meshali
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
3
|
Goo YT, Won YH, Hong SH, Choi JY, Sin GH, Kim CH, Jung HM, Choi YW. Optimization of a solidified micelle formulation for enhanced oral bioavailability of atorvastatin calcium using statistical experimental design. Pharm Dev Technol 2023; 28:479-491. [PMID: 37099663 DOI: 10.1080/10837450.2023.2208206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
To enhance the oral bioavailability of atorvastatin calcium (ATV), a novel solidified micelle (S-micelle) was developed. Two surfactants, Gelucire 48/16 (G48) and Tween 20 (T20), were employed for micelle formation, and two solid carriers (SC), Florite PS-10 (FLO) and Vivapur 105 (VP105), were selected as solid carriers. The S-micelle was optimized using a Box-Behnken design with three independent variables, including G48:T20 (X1, 1.8:1), SC:G48 + T20 (X2, 0.65:1), and FLO:VP105 (X3, 1.4:0.6), resulting in a droplet size (Y1) of 198.4 nm, dissolution efficiency at 15 min in the pH 1.2 medium (Y2) of 47.6%, Carr's index (Y3) of 16.9, and total quantity (Y4) of 562.5 mg. The optimized S-micelle resulted in good correlation showing percentage prediction values less than 10%. The optimized S-micelle formed a nanosized dispersion in the aqueous phase, with a higher dissolution rate than raw ATV and crushed Lipitor®. The optimized S-micelle improved the relative bioavailability of oral ATV (25 mg equivalent/kg) in rats by approximately 509% and 271% compared to raw ATV and crushed Lipitor®, respectively. In conclusion, the optimized S-micelle possesses great potential for the development of solidified formulations for improved oral absorption of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Yoon Tae Goo
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Yong-Hoon Won
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Sun Ho Hong
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Ji Yeh Choi
- York University, Department of Psychology, 4700 Keele St., Toronto, Ontario, Canada
| | - Gi Hyeong Sin
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Chang Hyun Kim
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Hyun Min Jung
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
4
|
Hsieh CM, Yang TL, Putri AD, Chen CT. Application of Design of Experiments in the Development of Self-Microemulsifying Drug Delivery Systems. Pharmaceuticals (Basel) 2023; 16:283. [PMID: 37259427 PMCID: PMC9958669 DOI: 10.3390/ph16020283] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 08/27/2023] Open
Abstract
Oral delivery has become the route of choice among all other types of drug administrations. However, typical chronic disease drugs are often poorly water-soluble, have low dissolution rates, and undergo first-pass metabolism, ultimately leading to low bioavailability and lack of efficacy. The lipid-based formulation offers tremendous benefits of using versatile excipients and has great compatibility with all types of dosage forms. Self-microemulsifying drug delivery system (SMEDDS) promotes drug self-emulsification in a combination of oil, surfactant, and co-surfactant, thereby facilitating better drug solubility and absorption. The feasible preparation of SMEDDS creates a promising strategy to improve the drawbacks of lipophilic drugs administered orally. Selecting a decent mixing among these components is, therefore, of importance for successful SMEDDS. Quality by Design (QbD) brings a systematic approach to drug development, and it offers promise to significantly improve the manufacturing quality performance of SMEDDS. Furthermore, it could be benefited efficiently by conducting pre-formulation studies integrated with the statistical design of experiment (DoE). In this review, we highlight the recent findings for the development of microemulsions and SMEDDS by using DoE methods to optimize the formulations for drugs in different excipients with controllable ratios. A brief overview of DoE concepts is discussed, along with its technical benefits in improving SMEDDS formulations.
Collapse
Affiliation(s)
- Chien-Ming Hsieh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Ting-Lun Yang
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan
| | - Athika Darumas Putri
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Department of Pharmaceutical Chemistry, Semarang College of Pharmaceutical Sciences (STIFAR), Semarang City 50192, Indonesia
| | - Chin-Tin Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
5
|
Sohn JS, Choi JS. A study on the improved dissolution and permeability of ticagrelor with sodium oleate in a ternary system. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
6
|
Jung M, Jin M, Jeon WJ, Lee H, Kim H, Won JH, Yoo H, Bai HW, Han SC, Suh H, Kang KU, Lee HK, Cho CW. Development of a long-acting tablet with ticagrelor high-loaded nanostructured lipid carriers. Drug Deliv Transl Res 2022; 13:1212-1227. [PMID: 35794353 DOI: 10.1007/s13346-022-01205-7] [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: 06/22/2022] [Indexed: 11/25/2022]
Abstract
Ticagrelor (TCG), an antiplatelet agent, has low solubility and permeability; thus, there are many trials to apply the pharmaceutical technology for the enhancement of TCG solubility and permeability. Herein, we have developed the TCG high-loaded nanostructured lipid carrier (HL-NLC) and solidified the HL-NLC to develop the oral tablet. The HL-NLC was successfully fabricated and optimized with a particle size of 164.5 nm, a PDI of 0.199, an encapsulation efficiency of 98.5%, and a drug loading of 16.4%. For the solidification of HL-NLC (S-HL-NLC), the adsorbent was determined based on the physical properties of the S-HL-NLC, such as bulk density, tap density, angle of repose, Hausner ratio, Carr's index, and drug content. Florite R was chosen because of its excellent adsorption capacity, excellent physical properties, and solubility of the powder after manufacturing. Using an S-HL-NLC, the S-HL-NLC tablet with HPMC 4 K was prepared, which is showed a released extent of more than 90% at 24 h. Thus, we have developed the sustained release tablet containing the TCG-loaded HL-NLC. Moreover, the formulations have exhibited no cytotoxicity against Caco-2 cells and improved the cellular uptake of TCG. In pharmacokinetic study, compared with raw TCG, the bioavailability of HL-NLC and S-HL-NLC was increased by 293% and 323%, respectively. In conclusion, we successfully developed the TCG high-loaded NLC tablet, that exhibited a sustained release profile and enhanced oral bioavailability.
Collapse
Affiliation(s)
- Minwoo Jung
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - Minki Jin
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - Woo-Jin Jeon
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - HaeSoo Lee
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - Haeun Kim
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - Jong-Hee Won
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - Hyelim Yoo
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea
| | - Hyoung-Woo Bai
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Institute of Toxicology (KIT), Jeollabuk-do, Jeongeup, 53212, South Korea.,Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Su-Cheol Han
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Institute of Toxicology (KIT), Jeollabuk-do, Jeongeup, 53212, South Korea
| | - Hearan Suh
- Postera Health Science Inc, Han River Misa 1st at Hyundai Knowledge Industry Center 550, Misa-daero, Hanam-si, 1005, Gyeonggi-do, South Korea
| | - Kyoung Un Kang
- Postera Health Science Inc, Han River Misa 1st at Hyundai Knowledge Industry Center 550, Misa-daero, Hanam-si, 1005, Gyeonggi-do, South Korea
| | - Hong-Ki Lee
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Institute of Toxicology (KIT), Jeollabuk-do, Jeongeup, 53212, South Korea.
| | - Cheong-Weon Cho
- College of Pharmacy, Chungnam National University, 99, Daehak-ro, Daejeon, 34134, South Korea.
| |
Collapse
|
7
|
Zhang W, Sun Y, Wei S, Wei B, Xu X, Tang Y. Untargeted metabolomics reveals the mechanism of quercetin enhancing the bioavailability of ticagrelor. Biomed Chromatogr 2021; 35:e5206. [PMID: 34185878 DOI: 10.1002/bmc.5206] [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: 04/01/2021] [Revised: 06/16/2021] [Accepted: 06/24/2021] [Indexed: 11/07/2022]
Abstract
Ticagrelor is a first-line clinical drug for the treatment of acute coronary syndrome, but its oral bioavailability is relatively low. Flavonoids (polyphenol compounds commonly found in plant foods) seriously affect human metabolism and health. This study compared the effects of quercetin, luteolin and catechin on the pharmacokinetic parameters of ticagrelor and found that quercetin can significantly increase the Cmax and area under the curve from time zero to 36 h (AUC0-36 ) of ticagrelor, that is, quercetin can enhance the bioavailability of ticagrelor, but luteolin and catechin cannot. The difference between the ticagrelor group and the combination of quercetin and ticagrelor was analyzed through untargeted metabolomics methods and multivariate data analysis, which identified changes in the levels of seven metabolites (deoxycholic acid, taurocholic acid, glycocholic acid, glycoursodeoxycholic acid, tryptophan, phenylalanine and kynurenine). Based on the changes of these metabolites, we found that the metabolic pathways of phenylalanine, tyrosine and tryptophan and the biosynthetic pathway of bile acids were changed. A metabolomics study revealed that quercetin improves the oral bioavailability of ticagrelor and that this might rely on changing the metabolic pathways of phenylalanine, tyrosine and tryptophan and the biosynthetic pathway of bile acids. The research results at the metabolic level provide us with a strong basis and direction for further exploring the mechanism underlying quercetin's ability to enhance the bioavailability of ticagrelor, and this may be useful for finding new agents that enhance the bioavailability.
Collapse
Affiliation(s)
- Weijie Zhang
- Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China.,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Co-innovation Center of Henan Province for New drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yaxin Sun
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Co-innovation Center of Henan Province for New drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Shuangyan Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Co-innovation Center of Henan Province for New drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Bo Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Co-innovation Center of Henan Province for New drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Co-innovation Center of Henan Province for New drug R & D and Preclinical Safety; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Youcai Tang
- Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| |
Collapse
|
8
|
Goo YT, Sa CK, Choi JY, Kim MS, Kim CH, Kim HK, Choi YW. Development of a Solid Supersaturable Micelle of Revaprazan for Improved Dissolution and Oral Bioavailability Using Box-Behnken Design. Int J Nanomedicine 2021; 16:1245-1259. [PMID: 33633449 PMCID: PMC7901570 DOI: 10.2147/ijn.s298450] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/23/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To enhance the oral bioavailability of revaprazan (RVP), a novel solid, supersaturable micelle (SSuM) was developed. METHODS Surfactants and solid carriers were screened based on a solubility and a flowability test, respectively. Supersaturating agents, including Poloxamer 407 (P407), were screened. The SSuM was optimized using a Box-Behnken design with three independent variables, including Gelucire 44/14:Brij L4 (G44/BL4; X1) and the amounts of Florite PS-10 (FLO; X2) and Vivapur 105 (VP105; X3), and three response variables, ie, dissolution efficiency at 30 min (Y1), dissolution enhancing capacity (Y2), and Carr's index (Y3). The solid state property was evaluated, and a dissolution test was conducted. RVP, Revanex®, solid micelle (P407-free from the composition of SSuM), and SSuM were orally administrated to rats (RVP 20 mg equivalent/kg) for in vivo pharmacokinetic study. RESULTS G44 and BL4 showed great solubility, with a critical micelle concentration range of 119.2-333.0 μg/mL. P407 had an excellent supersaturating effect. FLO and VP105 were selected as solid carriers, with a critical solidifying ratio (g/mL) of 0.30 and 0.91, respectively. With optimized values of X1 (-0.41), X2 (0.31), and X3 (-0.78), RVP (200 mg)-containing SSuM consisting of G44 (253.8 mg), BL4 (106.2 mg), FLO (99.3 mg), VP105 (199.8 mg), and P407 (40 mg) was developed, resulting in Y1 (40.3%), Y2 (0.008), and Y3 (12.3%). RVP existed in an amorphous state in the optimized SSuM, and the SSuM formed a nanosized dispersion in the aqueous phase, with approximately 71.7% dissolution at 2 h. The optimized SSuM improved the relative bioavailability of RVP in rats by approximately 478%, 276%, and 161% compared to raw RVP, Revanex®, and solid micelle, respectively. CONCLUSION The optimized SSuM has great potential for the development of solidified formulations of poorly water-soluble drugs with improved oral absorption.
Collapse
Affiliation(s)
- Yoon Tae Goo
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Cheol-Ki Sa
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Ji Yeh Choi
- Department of Psychology, York University, Toronto, Ontario, Canada
| | - Min Song Kim
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Chang Hyun Kim
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyeon Kyun Kim
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Li R, Wang Y, Yang Q, Lai B, Zhou X, Feng M. Enhanced Stability of the Pharmacologically Active Lactone Form of 10-Hydroxycamptothecin by Self-Microemulsifying Drug Delivery Systems. AAPS PharmSciTech 2020; 21:324. [PMID: 33200259 DOI: 10.1208/s12249-020-01860-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/24/2020] [Indexed: 12/25/2022] Open
Abstract
10-Hydroxycamptothecin (HCPT) is a DNA inhibitor of topoisomerase I and exerts antitumor activities against various types of cancer. However, reversible conversion from a pharmacologically active lactone form to an inactive carboxylate form of HCPT and poor water solubility hamper its clinical applications. To overcome these shortcomings, we designed a fine self-microemulsifying drug delivery system (SMEDDS) for HCPT to effectively protect HCPT in its active lactone form as well as improving dissolution rates. A formulation of HCPT-SMEDDS that contained ethyl oleate, D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), and polyethylene glycol 400 (PEG400) was optimized by using the central composite design and response surface methodology. Following 1:100 aqueous dilution of the optimized HCPT-SMEDDS, the droplet size of resulting microemulsions was 25.6 ± 0.7 nm, and the zeta potential was - 15.2 ± 0.4 mV. The optimized HCPT-SMEDDS appeared to stabilize the lactone moiety of HCPT with 73.6% being present in the pharmacologically active lactone forms in simulated intestinal fluid, but only 45.7% for free HCPT. Furthermore, the physically stable formulation showed the active lactone form predominated in HCPT-SMEDDS (> 95%) for 6 months under the accelerated storage condition. Meanwhile, the optimized SMEDDS formulation also significantly improved dissolution rates and membrane permeability of the lactone form of HCPT. Therefore, HCPT-SMEDDS involved designing for the ease of manufacture, and provided a potent oral dosage form for preserving its active lactone form as well as enhancing the dissolution rate.
Collapse
|
10
|
Elshafeey AH, Zayed R, Shukr MH, Elsayed I. Sucrose acetate isobutyrate based nanovesicles: A promising platform for drug delivery and bioavailability enhancement. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
11
|
Development and Evaluation of Docetaxel-Phospholipid Complex Loaded Self-Microemulsifying Drug Delivery System: Optimization and In Vitro/Ex Vivo Studies. Pharmaceutics 2020; 12:pharmaceutics12060544. [PMID: 32545452 PMCID: PMC7357111 DOI: 10.3390/pharmaceutics12060544] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/18/2023] Open
Abstract
Docetaxel (DTX) has clinical efficacy in the treatment of breast cancer, but it is difficult to develop a product for oral administration, due to low solubility and permeability. This study focused on preparing a self-microemulsifying drug delivery system (SME) loaded with DTX-phospholipid complex (DTX@PLC), to improve the dissolution and gastrointestinal (GI) permeability of DTX. A dual technique combining the phospholipid complexation and SME formulation described as improving upon the disadvantages of DTX has been proposed. We hypothesized that the complexation of DTX with phospholipids can improve the lipophilicity of DTX, thereby increasing the affinity of the drug to the cell lipid membrane, and simultaneously improving permeability through the GI barrier. Meanwhile, DTX@PLC-loaded SME (DTX@PLC-SME) increases the dissolution and surface area of DTX by forming a microemulsion in the intestinal fluid, providing sufficient opportunity for the drug to contact the GI membrane. First, we prepared DTX@PLC-SME by combining dual technologies, which are advantages for oral absorption. Next, we optimized DTX@PLC-SME with nanosized droplets (117.1 nm), low precipitation (8.9%), and high solubility (33.0 mg/g), which formed a homogeneous microemulsion in the aqueous phase. Dissolution and cellular uptake studies demonstrated that DTX@PLC-SME showed 5.6-fold higher dissolution and 2.3-fold higher DTX uptake in Caco-2 cells than raw material. In addition, an ex vivo gut sac study confirmed that DTX@PLC-SME improved GI permeability of DTX by 2.6-fold compared to raw material. These results suggested that DTX@PLC-SME can significantly overcome the disadvantages of anticancer agents, such as low solubility and permeability.
Collapse
|
12
|
Goo YT, Park SY, Chae BR, Yoon HY, Kim CH, Choi JY, Song SH, Choi YW. Optimization of solid self-dispersing micelle for enhancing dissolution and oral bioavailability of valsartan using Box-Behnken design. Int J Pharm 2020; 585:119483. [PMID: 32485217 DOI: 10.1016/j.ijpharm.2020.119483] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/11/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
A novel solid self-dispersing micelle (S-SDM) was developed to enhance the oral bioavailability of valsartan (VST) and to reduce the total mass of solidified supersaturable self-microemulsifying drug delivery system (S-SuSMEDDS), composed of Capmul MCM, Tween 80 (T80), Gelucire 44/14 (G44), Poloxamer 407, Florite PS-10 (FLO), and low-substituted hydroxypropyl cellulose B1 (HPC). Excluding oil component from S-SuSMEDDS, S-SDM was optimized using a Box-Behnken design with three independent variables: X1 (T80/G44, 0.63), X2 (FLO/HPC, 0.41), and X3 (solid carrier, 177.6 mg); and three response factors: Y1 (droplet size, 191.9 nm), Y2 (dissolution efficiency at 15 min, 55.0%), and Y3 (angle of repose, 32.4°). The desirability function was 0.636, showing an excellent agreement between the predicted and experimental values. With approximately 75% weight of S-SuSMEDDS, no distinct crystallinity of VST was observed in S-SDM, resulting in critical micelle concentration value of 32 μg/mL. Optimized S-SDM showed an approximate 4-fold improved dissolution (pH 1.2, 500 mL) compared with raw VST. Following oral administration in rats, optimized S-SDM improved relative bioavailability by approximately 235%, 216%, and 127% versus raw VST, Diovan® (commercial reference), and S-SuSMEDDS, respectively. Thus, optimized S-SDM could be a selectable candidate for developing water-insoluble drugs in reduced quantity.
Collapse
Affiliation(s)
- Yoon Tae Goo
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Sun Young Park
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Bo Ram Chae
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Ho Yub Yoon
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Chang Hyun Kim
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Ji Yeh Choi
- Department of Psychology, York University, 4700 Keele St, Toronto, Ontario, Canada.
| | - Seh Hyon Song
- College of Pharmacy, Kyungsung University, 309 Suyeong-ro Nam-gu, Busan 48434, Republic of Korea.
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, 84 Heuksuk-ro Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
13
|
Mandić J, Pirnat V, Luštrik M, German Ilić I, Vrečer F, Gašperlin M, Zvonar Pobirk A. Solidification of SMEDDS by fluid bed granulation and manufacturing of fast drug release tablets. Int J Pharm 2020; 583:119377. [PMID: 32339633 DOI: 10.1016/j.ijpharm.2020.119377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/05/2020] [Accepted: 04/23/2020] [Indexed: 11/29/2022]
Abstract
Solidification of self-microemulsifying drug delivery systems (SMEDDS) is a rising experimental field with important potential for pharmaceutical industry, however fluid-bed granulation with SMEDDS is yet an unexplored solidification technique. The aim of the study was to solidify carvedilol-loaded SMEDDS utilizing fluid bed granulation process and to investigate how the formulation variables (type of solid carrier, optimization of granulation dispersion) and fluid-bed granulation process variables can be optimized in order to achieve suitable agglomeration process, high drug loading and appropriate product characteristics. Obtained granulates exhibited complete drug release, comparable to liquid SMEDDS and superior to crystalline carvedilol, nevertheless compromise between large SMEDDS loading and appropriate flow properties of the granules has to be made. Representative granulates with highest drug loading were further compressed into tablets. It was shown that the optimal excipient selection of compression mixture and compression force can lead to fast carvedilol release even from the tablets. Selfmicroemulsifying properties were not impaired neither after the solidification process and nor after the compression of solid SMEDDS into tablets. This suggests that fluid-bed granulation with SMEDDS offers a perspective alternative for solidification of the SMEDDS, enabling preservation of self-microemulsifying properties, acceptable drug loading and complete drug release.
Collapse
Affiliation(s)
- Jelena Mandić
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia; Krka, d.d., Novo mesto, Šmarješka cesta 6, 8000 Novo mesto, Slovenia
| | - Vesna Pirnat
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Matevž Luštrik
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Ilija German Ilić
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Franc Vrečer
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia; Krka, d.d., Novo mesto, Šmarješka cesta 6, 8000 Novo mesto, Slovenia
| | - Mirjana Gašperlin
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Alenka Zvonar Pobirk
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
14
|
Na YG, Byeon JJ, Kim MK, Han MG, Cho CW, Baek JS, Lee HK, Shin YG. Pharmacokinetic/Pharmacodynamic Modeling To Predict the Antiplatelet Effect of the Ticagrelor-Loaded Self-Microemulsifying Drug Delivery System in Rats. Mol Pharm 2020; 17:1079-1089. [PMID: 32053381 DOI: 10.1021/acs.molpharmaceut.9b00964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ticagrelor (TCG) has been used as an antiplatelet agent for acute coronary syndrome patients. The aim of this research was to establish a population pharmacokinetic/pharmacodynamic (PK/PD) model of TCG and to apply the model for predicting the PD response of the TCG-loaded self-microemulsifying drug delivery system (TCG-SME) in rats. Pure TCG and TCG-SME (2, 5, and 10 mg/kg of TCG) were orally administered to male Sprague-Dawley rats. Plasma samples were collected at scheduled time-points and then analyzed for TCG plasma concentrations and antiplatelet effects. The inhibition of platelet aggregation of TCG was measured as a PD response. The PK profiles of pure TCG and TCG-SME could be well-explained with a two-compartment PK model. The accuracy of the PK model was assessed with a goodness-of-fit plot and conditional weight residual error (CWRES). Also, the visual predictive check was investigated based on the predictions. A population PK/PD model for pure TCG was established as an indirect response Emax model linked to the two-compartment PK model of pure TCG. The PK/PD model proposed a suitable fitting to link the plasma concentration of TCG simultaneously with platelet aggregation. Based on the PK data of TCG-SME, as well as the established PK/PD model of pure TCG, the PD profiles of TCG-SME were simulated. TCG-SME was more effective in inducing the antiplatelet effect than pure TCG at equivalent doses of TCG. The accuracy of the simulation was verified by comparing the simulated PD profile with the profile observed in rats. The observations were close to the model simulations. In addition, the values of CWRES were almost within ±2. In conclusion, the PK/PD modeling approach can provide a way for predicting mathematically the PD responses from PK profiles of other TCG formulations and a conceptual prediction for future clinical assessment.
Collapse
Affiliation(s)
- Young-Guk Na
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jin-Ju Byeon
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Min-Ki Kim
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Min-Gu Han
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Cheong-Weon Cho
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jong-Suep Baek
- Department of Herbal Medicine Resource, Kangwon National University, 346 Hwangjo-gil, Dogye-eup, Samcheok-si, Gangwon-do 25949, Republic of Korea
| | - Hong-Ki Lee
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Young G Shin
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| |
Collapse
|
15
|
Na YG, Pham TMA, Byeon JJ, Kim MK, Han MG, Baek JS, Lee HK, Cho CW. Development and evaluation of TPGS/PVA-based nanosuspension for enhancing dissolution and oral bioavailability of ticagrelor. Int J Pharm 2020; 581:119287. [PMID: 32243963 DOI: 10.1016/j.ijpharm.2020.119287] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/21/2020] [Accepted: 03/28/2020] [Indexed: 12/28/2022]
Abstract
In this study, we developed ticagrelor-dispersed nanosuspension (TCG-NSP) to enhance the dissolution and oral bioavailability of ticagrelor (TCG) through a statistical design approach. TCG, a reversible P2Y12 receptor antagonist, is classified as a biopharmaceutics classification system (BCS) class IV drug with low solubility and permeability, resulting in low oral bioavailability. Nanosuspension (NSP) is an efficient pharmaceutical technique for overcoming the disadvantages. First, we optimized TCG-NSP consisting of D-α-Tocopherol polyethylene glycol 1000 succinate (TPGS) and polyvinyl alcohol (PVA), which exhibited homogeneously dispersed TCG particle (233 nm) and low precipitation (3%). Characterization studies demonstrated that TCG-NSP provided amorphous TCG particles and supersaturation effect, resulting in higher dissolution than a commercial product. In addition, everted gut sac and pharmacokinetic studies confirmed that TCG-NSP improved the gastrointestinal permeation of TCG by 2.8-fold compared to commercial product, thereby enhancing the oral bioavailability (2.2-fold). These results suggested that TCG-NSP could be successfully used as an efficient pharmaceutical formulation to achieve the enhanced dissolution and oral bioavailability of TCG.
Collapse
Affiliation(s)
- Young-Guk Na
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Thi Mai Anh Pham
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jin-Ju Byeon
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Min-Ki Kim
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Min-Gu Han
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jong-Suep Baek
- Department of Herbal Medicine Resource, Kangwon National University, 346 Hwangjo-gil, Dogye-eup, Samcheok-si, Gangwon-do 25949, Republic of Korea
| | - Hong-Ki Lee
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| | - Cheong-Weon Cho
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|