1
|
Yao Y, Xu Z, Ding H, Yang S, Chen B, Zhou M, Zhu Y, Yang A, Yan X, Liang C, Kou X, Chen B, Huang W, Li Y. Carrier-free nanoparticles-new strategy of improving druggability of natural products. J Nanobiotechnology 2025; 23:108. [PMID: 39953594 PMCID: PMC11827262 DOI: 10.1186/s12951-025-03146-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
There are abundant natural products resources and extensive clinical use experience in China. However, the active components of natural products generally have problems such as poor water solubility and low bioavailability, which limit their druggability. Carrier-free nanoparticles, such as nanocrystals, self-assembled nanoparticles, and extracellular vesicles derived from both animal and plant sources, have great application potential in improving the safety and efficacy of drugs due to their simple and flexible preparation methods, high drug loading capacity and delivery efficiency, as well as long half-life in blood circulation. It has been widely used in biomedical fields such as anti-tumor, anti-bacterial, anti-inflammatory and anti-oxidation. Therefore, based on the natural products that have been used in clinic, this review focuses on the advantages of carrier-free nanoparticles in delivering active compounds, in order to improve the delivery process of natural products in vivo and improve their draggability.
Collapse
Affiliation(s)
- Yaqi Yao
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhenna Xu
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haoran Ding
- College of Pharmacy, Shandong Xiandai University, Jinan, 250104, China
| | - Shenshen Yang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bohan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mengjiao Zhou
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yehan Zhu
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Aihong Yang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xingxu Yan
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Chenrui Liang
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaodi Kou
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Bo Chen
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yubo Li
- National Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
2
|
Garbiec E, Rosiak N, Sip S, Zalewski P, Cielecka-Piontek J. Curcumin Solubility and Bioactivity Enhancement Through Amorphization with Tryptophan via Supercritical Fluid Technology. Int J Mol Sci 2025; 26:855. [PMID: 39859569 PMCID: PMC11766122 DOI: 10.3390/ijms26020855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Curcumin, a compound known for its antioxidant and neuroprotective properties, faces challenges due to its low water solubility, which can limit its effectiveness. One effective method to address this issue is through amorphization. Incorporating curcumin into a polymeric matrix to form amorphous solid dispersions is a common approach. Another strategy involves co-amorphous systems, where low-molecular-weight components act as co-formers. A recent innovative approach combines these strategies. This study used tryptophan as a co-former and prepared systems using supercritical fluid technology. The amorphous nature of two systems was confirmed through X-ray powder diffraction: one with 10% curcumin and a polymer, and another with 10% curcumin, a polymer, and tryptophan. Fourier-transform infrared analysis demonstrated molecular interactions among all components in the systems. Scanning electron microscopy revealed that the amorphization process significantly modified the morphology of the powder particles. The ternary system with tryptophan notably increased curcumin solubility by over 300-fold. The amorphous form of curcumin in both systems exhibited significantly higher dissolution rates compared to its crystalline form. The system with tryptophan showed more than a threefold improvement in permeability according to the PAMPA test. The enhanced solubility led to over a sixfold increase in antioxidant activity and a 25-fold improvement in the inhibition of the enzyme butyrylcholinesterase.
Collapse
Affiliation(s)
| | | | | | | | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland; (E.G.); (N.R.); (S.S.); (P.Z.)
| |
Collapse
|
3
|
Khatri B, Thakkar V, Dalwadi S, Shah A, Rana H, Shah P, Gandhi T, Prajapati B. Preparation and In-Vitro Characterization of Solid Lipid Nanoparticles Containing Artemisinin and Curcumin. Pharm Nanotechnol 2025; 13:199-211. [PMID: 39039683 DOI: 10.2174/0122117385296893240626061552] [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: 01/29/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Malaria remains a formidable public health obstacle across Africa, Southeast Asia, and portions of South America, exacerbated by resistance to antimalarial medications, such as artemisinin-based combinations. The combination of curcumin and artemisinin shows promise due to its potential for dose reduction, reduced toxicity, synergistic effects, and suitability for drug delivery improvement. OBJECTIVES This research aims to enhance the solubility and dissolution rates of curcumin and artemisinin by employing Solid Lipid Nanoparticles (SLNs). Oral delivery of both drugs faces challenges due to their poor water solubility, inefficient absorption, and rapid metabolism and elimination. METHODS The study focuses on formulating and optimizing Solid Lipid Nanoparticles (SLNs) encapsulating artemisinin (ART) and curcumin (CUR). SLNs were developed using the hot homogenization method, incorporating ultrasonication. Drug-excipient compatibility was evaluated using Differential Scanning Calorimetry (DSC). Lipid and surfactant screening was performed to select suitable components. A 3² full factorial design was utilized to investigate the influence of lipid and surfactant concentrations on key parameters, such as entrapment efficiency (%EE) and cumulative drug release (%CDR). Additionally, evaluations of %EE, drug loading, particle size, zeta potential, and in-vitro drug release were conducted. RESULTS Successful development of artemisinin and curcumin SLNs was achieved using a full factorial design, demonstrating controlled drug release and high entrapment efficiency. The optimized nanoparticles exhibited a size of 114.7nm, uniformity (PDI: 0.261), and a zeta potential of -9.24 mV. Artemisinin and curcumin showed %EE values of 79.1% and 74.5%, respectively, with cumulative drug release of 85.1% and 80.9%, respectively. The full factorial design indicated that increased lipid concentration improved %EE, while higher surfactant concentration enhanced drug release and %EE. Stability studies of the optimized batch revealed no alterations in physical or chemical characteristics. CONCLUSION The study successfully developed Solid Lipid Nanoparticles (SLNs) for artemisinin and curcumin, achieving controlled drug release, high entrapment efficiency, and desired particle size and uniformity. This advancement holds promise for enhancing drug delivery of herbal formulations.
Collapse
Affiliation(s)
- Bhagyashri Khatri
- Department of Pharmaceutics, Anand Pharmacy College, Anand, Gujarat, India
| | - Vaishali Thakkar
- Department of Pharmaceutics, Anand Pharmacy College, Anand, Gujarat, India
| | - Saloni Dalwadi
- Department of Pharmaceutics, Anand Pharmacy College, Anand, Gujarat, India
| | - Avani Shah
- Department of Pharmaceutics, Anand Pharmacy College, Anand, Gujarat, India
| | - Hardik Rana
- Department of Pharmaceutics, Anand Pharmacy College, Anand, Gujarat, India
| | - Purvi Shah
- Department of Quality Assurance, Parul Institute of Pharmacy and Research, Parul University, Vadodara, Gujarat, India
| | - Tejal Gandhi
- Department of Pharmacology, Anand Pharmacy College, Anand, India
| | | |
Collapse
|
4
|
Wang H, Zhao P, Ma R, Jia J, Fu Q. Drug-drug co-amorphous systems: An emerging formulation strategy for poorly water-soluble drugs. Drug Discov Today 2024; 29:103883. [PMID: 38219970 DOI: 10.1016/j.drudis.2024.103883] [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: 10/24/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Overcoming the poor water solubility of small-molecule drugs is a major challenge in the development of clinical pharmaceuticals. Amorphization of crystalline drugs is a highly effective strategy to improve their aqueous solubility. However, amorphous drugs are thermodynamically unstable and likely to crystallize during manufacturing and storage. Recently, drug-drug co-amorphous systems have emerged as a novel strategy to not only enable enhanced dissolution and physical stability of the individual drugs within the system but also to provide a strategy for combination therapy of the same or different clinical indications. This review serves to highlight advances in the methods used to manufacture and characterize drug-drug co-amorphous systems, summarize drug-drug co-amorphous applications reported in recent decades, and provide an outlook on future possibilities and perspectives.
Collapse
Affiliation(s)
- Hongge Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Peixu Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Ruilong Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jirun Jia
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
5
|
Li J, Wang X, Yu D, Zhoujin Y, Wang K. Molecular complexes of drug combinations: A review of cocrystals, salts, coamorphous systems and amorphous solid dispersions. Int J Pharm 2023; 648:123555. [PMID: 37890646 DOI: 10.1016/j.ijpharm.2023.123555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
As the advancements in the medical technology and healthcare develop through the years, combinational therapy has evolved to be an important treatment modality in many disease settings, including cancer, cardiovascular disease and infectious diseases. In an effort to alleviate "pill burden" and improve patient compliance, fixed dose combinations (FDCs) have been developed to be used as effective therapeutics. Among all FDCs, the category of drug-drug molecular complexes has been proven an efficient methodology in designing and treating diseases, with many drugs being approved. Among all drug-drug molecular complexes, drug-drug cocrystals, salts, coamorphous systems and solid dispersions have been successfully developed and many have been approved by the FDA. In this review, we dwell deeply into the molecular mechanisms behind the different types of drug-drug molecular complexes, including the key functional groups involved in the intermolecular interactions, the applications of each category of molecular complexes, as well as the advantages and challenges thereof. This comprehensive review provides useful insights into the practical design and manufacture of drug-drug molecular complexes and points out the future direction for the development of new advantageous combinational therapies that benefit more patients.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Xiyan Wang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, United States
| | - Yunping Zhoujin
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Kunlin Wang
- BeBetter Med Inc., Guangzhou, 510663, PR China; College of Pharmacy, Jinan University, Guangzhou, 510006, PR China.
| |
Collapse
|
6
|
Samie A, Alavian H, Vafaei-Pour Z, Mohammadpour AH, Jafarian AH, Danesh NM, Abnous K, Taghdisi SM. Accelerated Wound Healing with a Diminutive Scar through Cocrystal Engineered Curcumin. Mol Pharm 2023; 20:5090-5107. [PMID: 37624646 DOI: 10.1021/acs.molpharmaceut.3c00398] [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] [Indexed: 08/27/2023]
Abstract
Pharmaceutical cocrystals ( Regulatory Classification of Pharmaceutical Co-Crystals Guidance for Industry; Food and Drug Administration, 2018) are crystalline solids produced through supramolecular chemistry to modulate the physicochemical properties of active pharmaceutical ingredients (APIs). Despite their extensive development in interdisciplinary sciences, this is a pioneering study on the efficacy of pharmaceutical cocrystals in wound healing and scar reducing. Curcumin-pyrogallol cocrystal (CUR-PYR) was accordingly cherry-picked since its superior physicochemical properties adequately compensate for limitative drawbacks of curcumin (CUR). CUR-PYR has been synthesized by a liquid-assisted grinding (LAG) method and characterized via FT-IR, DSC, and PXRD analyses. In vitro antibacterial study indicated that CUR-PYR cocrystal, CUR+PYR physical mixture (PM), and PYR are more effective against both Gram-negative (Pseudomonas aeruginosa and Escherichia coli) and Gram-positive (Staphylococcus aureus and Bacillus subtilis) bacteria in comparison with CUR. In vitro results also demonstrated that the viability of HDF and NIH-3T3 cells treated with CUR-PYR were improved more than those received CUR which is attributed to the effect of PYR in the form of cocrystal. The wound healing process has been monitored through a 15 day in vivo experiment on 75 male rats stratified into six groups: five groups treated by CUR-PYR+Vaseline (CUR-PYR.ung), CUR+PYR+Vaseline (CUR+PYR.ung), CUR+Vaseline (CUR.ung), PYR+Vaseline (PYR.ung), and Vaseline (VAS) ointments and a negative control group of 0.9% sodium chloride solution (NS). It was revealed that the wounds under CUR-PYR.ung treatment closed by day 12 postsurgery, while the wounds in other groups failed to reach the complete closure end point until the end of the experiment. Surprisingly, a diminutive scar (3.89 ± 0.97% of initial wound size) was observed in the CUR-PYR.ung treated wounds by day 15 after injury, followed by corresponding values for PYR.ung (12.08 ± 2.75%), CUR+PYR.ung (13.89 ± 5.02%), CUR.ung (16.24 ± 6.39%), VAS (18.97 ± 6.89%), and NS (20.33 ± 5.77%). Besides, investigating histopathological parameters including inflammation, granulation tissue, re-epithelialization, and collagen deposition signified outstandingly higher ability of CUR-PYR cocrystal in wound healing than either of its two constituents separately or their simple PM. It was concluded that desired solubility of the prepared cocrystal was essentially responsible for accelerating wound closure and promoting tissue regeneration which yielded minimal scarring. This prototype research suggests a promising application of pharmaceutical cocrystals for the purpose of wound healing.
Collapse
Affiliation(s)
- Ali Samie
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Hoda Alavian
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Zeinab Vafaei-Pour
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Amir Hooshang Mohammadpour
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Science, Mashhad 9177948954, Iran
| | - Amir Hossein Jafarian
- Cancer and Molecular Research Center, Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Noor Mohammad Danesh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Khalil Abnous
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
7
|
Dubey T, Sonawane SK, Mannava MKC, Nangia AK, Chandrashekar M, Chinnathambi S. The inhibitory effect of Curcumin-Artemisinin co-amorphous on Tau aggregation and Tau phosphorylation. Colloids Surf B Biointerfaces 2023; 221:112970. [DOI: 10.1016/j.colsurfb.2022.112970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 11/08/2022]
|
8
|
Polymeric solid dispersion Vs co-amorphous technology: A critical comparison. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103980] [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]
|
9
|
Shi Q, Wang Y, Moinuddin SM, Feng X, Ahsan F. Co-amorphous Drug Delivery Systems: a Review of Physical Stability, In Vitro and In Vivo Performance. AAPS PharmSciTech 2022; 23:259. [PMID: 36123515 DOI: 10.1208/s12249-022-02421-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Over the past few decades, co-amorphous solids have been used as a promising approach for delivering poorly water-soluble drugs. Co-amorphous solids, comprising pharmacologically relevant drug substances or excipients, improve physical stability, solubility, dissolution, and bioavailability compared with single amorphous ingredients. In this review, we have summarized recent advances in physical stability and in vitro and in vivo performances of co-amorphous solids. We have highlighted the role of molar ratio, molecular interaction, and mobility that affects the physical stability of co-amorphous solids. This review delves deep as to how co-amorphous solids affect the physicochemical properties in vitro and in vivo. We also described the challenges to the formulation of co-amorphous solids. A better understanding of the mechanisms of the physical stability, in vitro and in vivo performance of co-amorphous solids, and proper selection of the co-former is likely to expedite the development of robust co-amorphous-based pharmaceutical formulations and can address the challenges associated with the delivery of poorly soluble drugs.
Collapse
Affiliation(s)
- Qin Shi
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224005, China.
| | - Yanan Wang
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224005, China
| | - Sakib M Moinuddin
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California, 95757, USA.,East Bay Institute For Research & Education (EBIRE), 10535 Hospital Way, Mather, California, 95655, USA
| | - Xiaodong Feng
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California, 95757, USA
| | - Fakhrul Ahsan
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California, 95757, USA. .,East Bay Institute For Research & Education (EBIRE), 10535 Hospital Way, Mather, California, 95655, USA.
| |
Collapse
|
10
|
Shi Q, Li F, Yeh S, Moinuddin SM, Xin J, Xu J, Chen H, Ling B. Recent Advances in Enhancement of Dissolution and Supersaturation of Poorly Water-Soluble Drug in Amorphous Pharmaceutical Solids: A Review. AAPS PharmSciTech 2021; 23:16. [PMID: 34893936 DOI: 10.1208/s12249-021-02137-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Amorphization is one of the most effective pharmaceutical approaches to enhance the dissolution and oral bioavailability of poorly water-soluble drugs. In recent years, amorphous formulations have been experiencing rapid development both in theoretical and practical application. Based on using different types of stabilizing agents, amorphous formulations can be mainly classified as polymer-based amorphous solid dispersion, coamorphous formulation, mesoporous silica-based amorphous formulation, etc. This paper summarizes recent advances in the dissolution and supersaturation of these amorphous formulations. Moreover, we also highlight the roles of stabilizing agents such as polymers, low molecular weight co-formers, and mesoporous silica. Maintaining supersaturation in solution is a key factor for the enhancement of dissolution profile and oral bioavailability, and thus, the strategies and challenges for maintaining supersaturation are also discussed. With an in-depth understanding of the inherent mechanisms of dissolution behaviors, the design of amorphous pharmaceutical formulations will become more scientific and reasonable, leading to vigorous development of commercial amorphous drug products.
Collapse
|
11
|
Araya-Sibaja AM, Salazar-López NJ, Wilhelm Romero K, Vega-Baudrit JR, Domínguez-Avila JA, Velázquez Contreras CA, Robles-Zepeda RE, Navarro-Hoyos M, González-Aguilar GA. Use of nanosystems to improve the anticancer effects of curcumin. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:1047-1062. [PMID: 34621615 PMCID: PMC8450944 DOI: 10.3762/bjnano.12.78] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/23/2021] [Indexed: 05/08/2023]
Abstract
Curcumin (CUR) is a phenolic compound that is safe for human consumption. It exhibits chemopreventive, antiproliferative, antiangiogenic, and antimetastatic effects. However, these benefits can be hampered due to the lipophilic nature, rapid metabolism, low bioavailability, and fast elimination of the molecule. Considering this, the present work reviews the use of CUR-based nanosystems as anticancer agents, including conventional nanosystems (i.e., liposomes, nanoemulsions, nanocrystals, nanosuspensions, polymeric nanoparticles) and nanosystems that respond to external stimuli (i.e., magnetic nanoparticles and photodynamic therapy). Previous studies showed that the effects of CUR were improved when loaded into nanosystems as compared to the free compound, as well as synergist effects when it is co-administrated alongside with other molecules. In order to maximize the beneficial health effects of CUR, critical factors need to be strictly controlled, such as particle size, morphology, and interaction between the encapsulating material and CUR. In addition, there is an area of study to be explored in the development of CUR-based smart materials for nanomedical applications. Imaging-guided drug delivery of CUR-based nanosystems may also directly target specific cells, thereby increasing the therapeutic and chemopreventive efficacy of this versatile compound.
Collapse
Affiliation(s)
- Andrea M Araya-Sibaja
- Laboratorio Nacional de Nanotecnología LANOTEC-CeNAT-CONARE, 1174-1200, Pavas, San José, Costa Rica
- Universidad Técnica Nacional, 1902-4050, Alajuela, Costa Rica
| | - Norma J Salazar-López
- Laboratorio de Antioxidantes y Alimentos Funcionales, Centro de Investigación en Alimentación y Desarrollo (CIAD), A.C., Hermosillo, Sonora 83304, México
- Universidad Autónoma de Baja California, Facultad de Medicina de Mexicali, Lic. en Nutrición, Dr. Humberto Torres Sanginés S/N, Centro Cívico, Mexicali, Baja California 21000, México
| | - Krissia Wilhelm Romero
- Laboratorio Nacional de Nanotecnología LANOTEC-CeNAT-CONARE, 1174-1200, Pavas, San José, Costa Rica
- Laboratorio BioDESS, Escuela de Química, Universidad de Costa Rica, San Pedro de Montes de Oca 2060, San José, Costa Rica
| | - José R Vega-Baudrit
- Laboratorio Nacional de Nanotecnología LANOTEC-CeNAT-CONARE, 1174-1200, Pavas, San José, Costa Rica
- Laboratorio de Investigación y Tecnología de Polímeros POLIUNA, Escuela de Química, Universidad Nacional de Costa Rica, Heredia 86-3000, Costa Rica
| | - J Abraham Domínguez-Avila
- Cátedras CONACYT-Centro de Investigación en Alimentación y Desarrollo A. C., Hermosillo, Sonora 83304, México
| | - Carlos A Velázquez Contreras
- Unidad Regional Centro, Departamento de Ciencias Químico-Biológicas y de la Salud, Universidad de Sonora, Hermosillo, Sonora 83000, México
| | - Ramón E Robles-Zepeda
- Unidad Regional Centro, Departamento de Ciencias Químico-Biológicas y de la Salud, Universidad de Sonora, Hermosillo, Sonora 83000, México
| | - Mirtha Navarro-Hoyos
- Laboratorio BioDESS, Escuela de Química, Universidad de Costa Rica, San Pedro de Montes de Oca 2060, San José, Costa Rica
| | - Gustavo A González-Aguilar
- Laboratorio de Antioxidantes y Alimentos Funcionales, Centro de Investigación en Alimentación y Desarrollo (CIAD), A.C., Hermosillo, Sonora 83304, México
| |
Collapse
|
12
|
Tseng WC, Chen CY, Chern CY, Wang CA, Lee WC, Chi YC, Cheng SF, Kuo YT, Chiu YC, Tseng ST, Lin PY, Liou SJ, Li YC, Chen CC. Targeting HR Repair as a Synthetic Lethal Approach to Increase DNA Damage Sensitivity by a RAD52 Inhibitor in BRCA2-Deficient Cancer Cells. Int J Mol Sci 2021; 22:4422. [PMID: 33922657 PMCID: PMC8122931 DOI: 10.3390/ijms22094422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/01/2023] Open
Abstract
BRCA mutation, one of the most common types of mutations in breast and ovarian cancer, has been suggested to be synthetically lethal with depletion of RAD52. Pharmacologically inhibiting RAD52 specifically eradicates BRCA-deficient cancer cells. In this study, we demonstrated that curcumin, a plant polyphenol, sensitizes BRCA2-deficient cells to CPT-11 by impairing RAD52 recombinase in MCF7 cells. More specifically, in MCF7-siBRCA2 cells, curcumin reduced homologous recombination, resulting in tumor growth suppression. Furthermore, a BRCA2-deficient cell line, Capan1, became resistant to CPT-11 when BRCA2 was reintroduced. In vivo, xenograft model studies showed that curcumin combined with CPT-11 reduced the growth of BRCA2-knockout MCF7 tumors but not MCF7 tumors. In conclusion, our data indicate that curcumin, which has RAD52 inhibitor activity, is a promising candidate for sensitizing BRCA2-deficient cells to DNA damage-based cancer therapies.
Collapse
Affiliation(s)
- Wei-Che Tseng
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- Graduate Institute of Health Industry Technology and Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Ching-Yuh Chern
- Department of Applied Chemistry, National Chiayi University, Chiayi 600, Taiwan; (C.-Y.C.); (Y.-C.L.)
| | - Chu-An Wang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Wen-Chih Lee
- Translational Research Program in Pediatric Orthopedics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Ying-Chih Chi
- Cryo-EM Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Shu-Fang Cheng
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Yi-Tsen Kuo
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Ya-Chen Chiu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Shih-Ting Tseng
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Pei-Ya Lin
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Shou-Jhen Liou
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
| | - Yi-Chen Li
- Department of Applied Chemistry, National Chiayi University, Chiayi 600, Taiwan; (C.-Y.C.); (Y.-C.L.)
| | - Chin-Chuan Chen
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan; (W.-C.T.); (S.-F.C.); (Y.-T.K.); (Y.-C.C.); (S.-T.T.); (P.-Y.L.); (S.-J.L.)
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| |
Collapse
|
13
|
Co-Amorphous Drug Formulations in Numbers: Recent Advances in Co-Amorphous Drug Formulations with Focus on Co-Formability, Molar Ratio, Preparation Methods, Physical Stability, In Vitro and In Vivo Performance, and New Formulation Strategies. Pharmaceutics 2021; 13:pharmaceutics13030389. [PMID: 33804159 PMCID: PMC7999207 DOI: 10.3390/pharmaceutics13030389] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/20/2022] Open
Abstract
Co-amorphous drug delivery systems (CAMS) are characterized by the combination of two or more (initially crystalline) low molecular weight components that form a homogeneous single-phase amorphous system. Over the past decades, CAMS have been widely investigated as a promising approach to address the challenge of low water solubility of many active pharmaceutical ingredients. Most of the studies on CAMS were performed on a case-by-case basis, and only a few systematic studies are available. A quantitative analysis of the literature on CAMS under certain aspects highlights not only which aspects have been of great interest, but also which future developments are necessary to expand this research field. This review provides a comprehensive updated overview on the current published work on CAMS using a quantitative approach, focusing on three critical quality attributes of CAMS, i.e., co-formability, physical stability, and dissolution performance. Specifically, co-formability, molar ratio of drug and co-former, preparation methods, physical stability, and in vitro and in vivo performance were covered. For each aspect, a quantitative assessment on the current status was performed, allowing both recent advances and remaining research gaps to be identified. Furthermore, novel research aspects such as the design of ternary CAMS are discussed.
Collapse
|
14
|
Pantwalawalkar J, More H, Bhange D, Patil U, Jadhav N. Novel curcumin ascorbic acid cocrystal for improved solubility. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Mohapatra TK, Moharana AK, Swain RP, Subudhi B. Coamorphisation of acetyl salicylic acid and curcumin for enhancing dissolution, anti-inflammatory effect and minimizing gastro toxicity. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
16
|
Abstract
Eutectic, co-amorphous, cocrystal, and physical mixtures of curcumin with basic amino acids are prepared and characterized by PXRD, DSC, NMR, FT-IR, and SEM; solubility and dissolution improvement achieved in 40% ethanol–water system.
Collapse
Affiliation(s)
- Anilkumar Gunnam
- School of Chemistry
- University of Hyderabad
- Hyderabad 500 046
- India
| | | |
Collapse
|
17
|
Rai SK, Baidya D, Nangia AK. Salts, solvates and hydrates of the multi-kinase inhibitor drug pazopanib with hydroxybenzoic acids. CrystEngComm 2021. [DOI: 10.1039/d1ce00785h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eight cocrystal-salts of the multi-kinase drug pazopanib with hydroxybenzoic acids are sustained by the strong, ionic aminopyridinium⋯carboxylate heterosynthon of N–H⋯O hydrogen bonds between the carboxylic acid donor and amino-pyrimidine acceptor.
Collapse
Affiliation(s)
- Sunil K. Rai
- Division of Organic Chemistry, CSIR – National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India
| | - Debjani Baidya
- Division of Organic Chemistry, CSIR – National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India
| | - Ashwini K. Nangia
- Division of Organic Chemistry, CSIR – National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India
- School of Chemistry, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Central University P.O., Hyderabad 500 046, India
| |
Collapse
|
18
|
Tomar D, Singh PK, Hoque S, Modani S, Sriram A, Kumar R, Madan J, Khatri D, Dua K. Amorphous systems for delivery of nutraceuticals: challenges opportunities. Crit Rev Food Sci Nutr 2020; 62:1204-1221. [PMID: 33103462 DOI: 10.1080/10408398.2020.1836607] [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] [Indexed: 01/28/2023]
Abstract
Amorphous solid products have recently gained a lot of attention as key solutions to improve the solubility and bioavailability of poorly soluble nutraceuticals. A pure amorphous drug is a high-energy form; physically/chemically unstable and so easily gets recrystallized into the less soluble crystalline form limiting solubility and bioavailability issues. Amorphous solid dispersion and co-amorphous are new formulation approach that stabilized unstable amorphous form through different mechanisms such as preventing mobility, high glass transition temperature and molecular interaction. Nutraceuticals have been received the utmost importance due to their health benefits. However, most of these compounds have been associated with poor oral bioavailability due to poor solubility, high lipophilicity, high melting point, poor permeability, degradability and rapid metabolism in the gastrointestinal tract (GIT) which limits its health benefits. This review provides us a systematic application of amorphous systems to the delivery of poorly soluble nutraceuticals, with the aim of overcoming their pharmacokinetic limitations and improved pharmacological potential. In particular, it describes the challenges associated with delivery of oral nutraceuticals, various methods involved in the preparation and characterization of amorphous systems and permeability enhancement of nutraceuticals are in detail.
Collapse
Affiliation(s)
- Devendrasingh Tomar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pankaj K Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sajidul Hoque
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sheela Modani
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Anitha Sriram
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rahul Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dharmendra Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health (GSH), The University of Technology Sydney, Ultimo, Australia
| |
Collapse
|
19
|
Zheng D, Ruan YT, Yin ZP, Zhang QF. A Comparison of Solubility, Stability, and Bioavailability between Astilbin and Neoastilbin Isolated from Smilax glabra Rhizoma. Molecules 2020; 25:molecules25204728. [PMID: 33076319 PMCID: PMC7587598 DOI: 10.3390/molecules25204728] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/05/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022] Open
Abstract
Astilbin and neoastilbin are two flavonoid stereoisomers. In the present study, their solubility, stability, and bioavailability were compared in a rat. The results revealed that the water solubility of astilbin and neoastilbin was 132.72 μg/mL and 217.16 μg/mL, respectively. The oil–water distribution coefficient (log P) of astilbin and neoastilbin in simulated gastric fluid (SGF) was 1.57 and 1.39, and in simulated intestinal fluid (SIF) was 1.09 and 0.98, respectively. In SIF, about 78.6% astilbin remained after 4 h of incubation at 37 °C, while this value was 88.3% for neoastilbin. Most of the degraded astilbin and neoastilbin were isomerized into their cis-trans-isomer, namely neoisoastilbin and isoastilbin, respectively, and the decomposed parts were rare. For bioavailability comparison in a rat, an HPLC method for trace amounts of astilbin and neoastilbin determination in plasma was developed, and the pretreatment of plasma was optimized. A pharmacokinetic study showed that the absolute bioavailability of astilbin and neoastilbin in a rat showed no significant difference with values of 0.30% and 0.28%, respectively.
Collapse
|
20
|
Sathisaran I, Devidas Bhatia D, Vishvanath Dalvi S. New curcumin-trimesic acid cocrystal and anti-invasion activity of curcumin multicomponent solids against 3D tumor models. Int J Pharm 2020; 587:119667. [PMID: 32702448 DOI: 10.1016/j.ijpharm.2020.119667] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/28/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022]
Abstract
Curcumin (CUR) is a Biopharmaceutics Classification System (BCS) class IV drug with poor aqueous solubility and low permeability. The dissolution of CUR can be enhanced through the cocrystallization approach. In this work, we report a new cocrystal phase of CUR with trimesic acid (TMA) with the enhanced dissolution of CUR. Cytotoxicity and cell invasion assays were conducted on (2D) monolayers and three-dimensional (3D) tumor models of triple-negative breast cancer (TNBC) cells, MDA-MB-231 using the new CUR-TMA cocrystal phase along with different CUR solid forms prepared in our previous works. The cytotoxicity and internalization assays conducted on 2D monolayers indicated that all CUR multicomponent solid forms except Curcumin-Folic Acid Dihydrate (CUR-FAD) (1:1) coamorphous solid exhibited enhanced bioavailability than unprocessed CUR. Cell invasion assay conducted on 3D tumor spheroid models showed that Curcumin-Hydroxyquinol (CUR-HXQ) cocrystal completely inhibited cell invasion whereas CUR-FAD (1:1) coamorphous solid induced enhanced invasion of cells from spheroid models.
Collapse
Affiliation(s)
- Indumathi Sathisaran
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Dhiraj Devidas Bhatia
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Sameer Vishvanath Dalvi
- Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
21
|
Han J, Wei Y, Lu Y, Wang R, Zhang J, Gao Y, Qian S. Co-amorphous systems for the delivery of poorly water-soluble drugs: recent advances and an update. Expert Opin Drug Deliv 2020; 17:1411-1435. [DOI: 10.1080/17425247.2020.1796631] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jiawei Han
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuanfeng Wei
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yan Lu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Runze Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Jianjun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuan Gao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Shuai Qian
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
22
|
Ueda H, Peter Bøtker J, Edinger M, Löbmann K, Grohganz H, Müllertz A, Rades T, Østergaard J. Formulation of co-amorphous systems from naproxen and naproxen sodium and in situ monitoring of physicochemical state changes during dissolution testing by Raman spectroscopy. Int J Pharm 2020; 587:119662. [PMID: 32682958 DOI: 10.1016/j.ijpharm.2020.119662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 11/29/2022]
Abstract
Co-amorphous systems comprising low-molecular weight drugs and co-formers constitute an interesting approach to optimize pharmaceutical performance of drugs with low aqueous solubility. Within the different types of co-amorphous systems, the combination of a drug with its own salt may be an attractive formulation option due the absence of any inactive co-formers. The aim of this study was to investigate the possibility of forming a co-amorphous system from naproxen (NAP) and its sodium salt (NAP(Na)). Ball milling of NAP and NAP(Na) at equal molar ratio resulted in the formation of a co-amorphous system whilst NAP and NAP(Na) alone were crystalline following both, ball milling and melt quenching. Infrared spectroscopy and physical stability testing revealed that intermolecular interactions were able to maintain the ball milled NAP-NAP(Na) system amorphous for 2 months at 40 °C. Surprisingly, the dissolution rate of co-amorphous NAP-NAP(Na) was only intermediate between those of crystalline NAP and crystalline NAP(Na). In situ Raman spectroscopic measurements indicated an initial phase separation of the co-amorphous form to NAP and NAP(Na) followed by dissociation of sodium from NAP(Na) and crystallization to NAP. These findings contribute to the design of co-amorphous formulations with the combination of a drug and its own salt.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Laboratory for Medicinal Chemistry Research, Physical Chemistry, Shionogi & Co., Ltd., Osaka, Japan
| | - Johan Peter Bøtker
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Edinger
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Korbinian Löbmann
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Holger Grohganz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jesper Østergaard
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Purification of Curcumin from Ternary Extract-Similar Mixtures of Curcuminoids in a Single Crystallization Step. CRYSTALS 2020. [DOI: 10.3390/cryst10030206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Crystallization-based separation of curcumin from ternary mixtures of curcuminoids having compositions comparable to commercial extracts was studied experimentally. Based on solubility and supersolubility data of both, pure curcumin and curcumin in presence of the two major impurities demethoxycurcumin (DMC) and bis(demethoxy)curcumin (BDMC), seeded cooling crystallization procedures were derived using acetone, acetonitrile and 50/50 (wt/wt) mixtures of acetone/2-propanol and acetone/acetonitrile as solvents. Starting from initial curcumin contents of 67–75% in the curcuminoid mixtures single step crystallization processes provided crystalline curcumin free of BDMC at residual DMC contents of 0.6–9.9%. Curcumin at highest purity of 99.4% was obtained from a 50/50 (wt/wt) acetone/2-propanol solution in a single crystallization step. It is demonstrated that the total product yield can be significantly enhanced via addition of water, 2-propanol and acetonitrile as anti-solvents at the end of a cooling crystallization process.
Collapse
|
24
|
Mao QQ, Xu XY, Shang A, Gan RY, Wu DT, Atanasov AG, Li HB. Phytochemicals for the Prevention and Treatment of Gastric Cancer: Effects and Mechanisms. Int J Mol Sci 2020; 21:E570. [PMID: 31963129 PMCID: PMC7014214 DOI: 10.3390/ijms21020570] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer is the fifth most common cancer, and the third most prevalent cause of cancer-related deaths in the world. Voluminous evidence has demonstrated that phytochemicals play a critical role in the prevention and management of gastric cancer. Most epidemiological investigations indicate that the increased intake of phytochemicals could reduce the risk of gastric cancer. Experimental studies have elucidated the mechanisms of action, including inhibiting cancer cell proliferation, inducing apoptosis and autophagy, and suppressing angiogenesis as well as cancer cell metastasis. These mechanisms have also been related to the inhibition of Helicobacter pylori and the modulation of gut microbiota. In addition, the intake of phytochemicals could enhance the efficacy of anticancer chemotherapeutics. Moreover, clinical studies have illustrated that phytochemicals have the potential for the prevention and the management of gastric cancer in humans. To provide an updated understanding of relationships between phytochemicals and gastric cancer, this review summarizes the effects of phytochemicals on gastric cancer, highlighting the underlying mechanisms. This review could be helpful for guiding the public in preventing gastric cancer through phytochemicals, as well as in developing functional food and drugs for the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Qian-Qian Mao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Q.-Q.M.); (X.-Y.X.); (A.S.)
| | - Xiao-Yu Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Q.-Q.M.); (X.-Y.X.); (A.S.)
| | - Ao Shang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Q.-Q.M.); (X.-Y.X.); (A.S.)
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu 610213, China
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ding-Tao Wu
- Institute of Food Processing and Safety, College of Food Science, Sichuan Agricultural University, Ya’an 625014, China;
| | - Atanas G. Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland;
- Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev str., 1113 Sofia, Bulgaria
- Department of Pharmacognosy, University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Q.-Q.M.); (X.-Y.X.); (A.S.)
| |
Collapse
|
25
|
Lodagekar A, Chavan RB, Mannava MKC, Yadav B, Chella N, Nangia AK, Shastri NR. Co amorphous valsartan nifedipine system: Preparation, characterization, in vitro and in vivo evaluation. Eur J Pharm Sci 2019; 139:105048. [PMID: 31446077 DOI: 10.1016/j.ejps.2019.105048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 01/30/2023]
Abstract
Co amorphous systems are supersaturated drug delivery systems which offer a basic platform for delivery of multicomponent adducts (combination of more than one active pharmaceutical ingredient (API)) and/or as a fixed dose combination therapy, in addition to their potential to improve the apparent solubility, dissolution rate and ultimately bioavailability of poorly water soluble APIs. In the present work, a new drug-drug co amorphous system namely valsartan-nifedipine was prepared by quench cooling technique. Prepared co amorphous system was characterized for its solid state behavior with the help of Fourier Transform Infrared spectroscopy (FTIR), Differential Scanning Calorimetry (DSC) and Powder X Ray Diffractometry (PXRD). The optimized co amorphous system was stable for 1 month when exposed to accelerated stability condition (40 ± 2 °C and 75 ± 5% RH). The improved stability of amorphous nifedipine in co amorphous system was attributed to improved miscibility and intra and intermolecular non-covalent interactions mainly due to presence of hydrogen bonding between valsartan and nifedipine which was studied by FTIR analysis. Co amorphous systems were evaluated by mainly in vitro dissolution and in vivo benefit. In vitro dissolution study showed nearly 5.66 folds and 1.61 folds improvement which was translated to 3.63 and 2.19 times enhancement in vivo Cmax for nifedipine and valsartan respectively.
Collapse
Affiliation(s)
- Anurag Lodagekar
- Solid State Pharmaceutical Research Group (SSPRG), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rahul B Chavan
- Solid State Pharmaceutical Research Group (SSPRG), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - M K Chaitanya Mannava
- School of Chemistry, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli Central University P.O, Hyderabad 500046, India
| | - Balvant Yadav
- Solid State Pharmaceutical Research Group (SSPRG), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Naveen Chella
- Solid State Pharmaceutical Research Group (SSPRG), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Ashwini K Nangia
- School of Chemistry, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli Central University P.O, Hyderabad 500046, India; CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India
| | - Nalini R Shastri
- Solid State Pharmaceutical Research Group (SSPRG), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
26
|
Liu X, Cao J, Huang G, Zhao Q, Shen J. Biological Activities of Artemisinin Derivatives Beyond Malaria. Curr Top Med Chem 2019; 19:205-222. [DOI: 10.2174/1568026619666190122144217] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/26/2022]
Abstract
Artemisinin is isolated from Artemisia annua L. with peroxide-containing sesquiterpene lactone structure. Because of its unique structural characteristics and promising anticancer, antivirus activities, it has recently received increasing attention. The aim of this review is to summarize recent discoveries of artemisinin's novel derivatives with new pharmaceutical effects beyond malaria with a focus on its antitumor and antivirus activity, as well as potential results of combination therapy with other clinical drugs.
Collapse
Affiliation(s)
- Xiaoyan Liu
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianguo Cao
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, 201418, China
| | - Guozheng Huang
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, 201418, China
| | - Qingjie Zhao
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingshan Shen
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
27
|
Shi Q, Moinuddin SM, Cai T. Advances in coamorphous drug delivery systems. Acta Pharm Sin B 2019; 9:19-35. [PMID: 30766775 PMCID: PMC6361732 DOI: 10.1016/j.apsb.2018.08.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/11/2018] [Accepted: 08/12/2018] [Indexed: 01/18/2023] Open
Abstract
In recent years, the coamorphous drug delivery system has been established as a promising formulation approach for delivering poorly water-soluble drugs. The coamorphous solid is a single-phase system containing an active pharmaceutical ingredient (API) and other low molecular weight molecules that might be pharmacologically relevant APIs or excipients. These formulations exhibit considerable advantages over neat crystalline or amorphous material, including improved physical stability, dissolution profiles, and potentially enhanced therapeutic efficacy. This review provides a comprehensive overview of coamorphous drug delivery systems from the perspectives of preparation, physicochemical characteristics, physical stability, in vitro and in vivo performance. Furthermore, the challenges and strategies in developing robust coamorphous drug products of high quality and performance are briefly discussed.
Collapse
Key Words
- API, active pharmaceutical ingredient;
- AUC, area under plasma concentrations-time curve
- BCS, bio-pharmaceutics classification systems
- Bioavailability
- Characterization
- Cmax, maximum plasma concentration
- Coamorphous
- Css, plasma concentration at steady state
- DSC, differential scanning calorimetry
- DVS, dynamic vapor sorption
- Dc, relative degree of crystallization
- Dissolution
- FT-IR, fourier transform infrared spectroscopy
- HME, hot melt extrusion
- HPLC, high performance liquid chromatography
- IDR, intrinsic dissolution rate
- LFRS, low-frequency Raman spectroscopy
- LLPS, liquid—liquid phase separation
- MTDSC, modulated temperature differential scanning calorimetry
- NMR, nuclear magnetic resonance
- P-gp, P-glycoprotein
- PXRD, powder X-ray diffraction
- Physical stability
- Preparation
- RH, relative humidity
- SEM, scanning electron microscope
- TGA, thermogravimetric analysis
- Tg, glass transition temperature
- Tmax, time of maximum plasma concentration
- UV, ultraviolet spectroscopy
Collapse
Affiliation(s)
| | | | - Ting Cai
- Corresponding author. Tel.: +86 25 83271123.
| |
Collapse
|
28
|
Sathisaran I, Dalvi SV. Engineering Cocrystals of PoorlyWater-Soluble Drugs to Enhance Dissolution in Aqueous Medium. Pharmaceutics 2018; 10:E108. [PMID: 30065221 PMCID: PMC6161265 DOI: 10.3390/pharmaceutics10030108] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 01/17/2023] Open
Abstract
Biopharmaceutics Classification System (BCS) Class II and IV drugs suffer from poor aqueous solubility and hence low bioavailability. Most of these drugs are hydrophobic and cannot be developed into a pharmaceutical formulation due to their poor aqueous solubility. One of the ways to enhance the aqueous solubility of poorlywater-soluble drugs is to use the principles of crystal engineering to formulate cocrystals of these molecules with water-soluble molecules (which are generally called coformers). Many researchers have shown that the cocrystals significantly enhance the aqueous solubility of poorly water-soluble drugs. In this review, we present a consolidated account of reports available in the literature related to the cocrystallization of poorly water-soluble drugs. The current practice to formulate new drug cocrystals with enhanced solubility involves a lot of empiricism. Therefore, in this work, attempts have been made to understand a general framework involved in successful (and unsuccessful) cocrystallization events which can yield different solid forms such as cocrystals, cocrystal polymorphs, cocrystal hydrates/solvates, salts, coamorphous solids, eutectics and solid solutions. The rationale behind screening suitable coformers for cocrystallization has been explained based on the rules of five i.e., hydrogen bonding, halogen bonding (and in general non-covalent bonding), length of carbon chain, molecular recognition points and coformer aqueous solubility. Different techniques to screen coformers for effective cocrystallization and methods to synthesize cocrystals have been discussed. Recent advances in technologies for continuous and solvent-free production of cocrystals have also been discussed. Furthermore, mechanisms involved in solubilization of these solid forms and the parameters influencing dissolution and stability of specific solid forms have been discussed. Overall, this review provides a consolidated account of the rationale for design of cocrystals, past efforts, recent developments and future perspectives for cocrystallization research which will be extremely useful for researchers working in pharmaceutical formulation development.
Collapse
Affiliation(s)
- Indumathi Sathisaran
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Sameer Vishvanath Dalvi
- Department of Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
29
|
Suresh K, Nangia A. Curcumin: pharmaceutical solids as a platform to improve solubility and bioavailability. CrystEngComm 2018. [DOI: 10.1039/c8ce00469b] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The remarkable improvements in the pharmacokinetics and high bioavailability of curcumin polymorphs, amorphous, cocrystals, eutectics, and coamorphous solids are discussed. The importance of pharmaceutical solids in the advanced formulation development of herbal and bioactive molecule curcumin is presented.
Collapse
Affiliation(s)
- Kuthuru Suresh
- School of Chemistry
- University of Hyderabad
- Hyderabad 500 046
- India
| | - Ashwini Nangia
- School of Chemistry
- University of Hyderabad
- Hyderabad 500 046
- India
- CSIR-National Chemical Laboratory
| |
Collapse
|