1
|
Sana SS, Chandel AKS, Raorane CJ, Aly Aly Saad M, Kim SC, Raj V, Sangkil Lee. Recent advances in nano and micro formulations of Ginsenoside to enhance their therapeutic efficacy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:156007. [PMID: 39276537 DOI: 10.1016/j.phymed.2024.156007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND AND AIMS Ginsenosides, the main component of Panax ginseng, have long been recognized for their therapeutic benefits and are thought to have neuroprotective, antidiabetic, anti-depressant, antioxidant, anti-cancer, and anti-stress properties. However, due to their low water solubility, low biomembrane permeability, gastrointestinal dysfunction, and total metabolism in the body, ginsenosides have a poor absorption profile that has hindered the therapeutic potential of these organic molecules. METHODS Initially, we broadly illuminated the several techniques of extraction of Ginsenosides using Panax quinquefolius and Panax ginseng. Subsequently, we focused on different delivery methods to improve the stability, permeability, and solubility of natural chemicals, which raises the bioavailability of ginsenoside. Lastly, we explained significance of a variety of nano and microscale delivery systems, including liposomes, ethosomes, transfersomes, metal/metal oxide systems, micro/nanoemulsions, polymeric micro/nanoparticles (NPs), liposomes, transfersomes, and micelles to increase the bioavailability of ginsenosides. RESULTS The utilization of micro/nanoscale delivery methods, such as liposome-based delivery, polymer micro/nanoparticle distribution, and micro/nanoemulsion, to increase the bioavailability of ginsenosides has recently advanced, and we have emphasized these advances in this study. Furthermore, the disadvantages of ginsenosides were also discussed, including the challenges associated with putting these delivery systems into practice in clinical settings and suggestions for further research. CONCLUSION In summary, ginsenosides-based administration has several benefits that make it a potentially useful substance for a range of therapeutic purposes.
Collapse
Affiliation(s)
- Siva Sankar Sana
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | | | | | - Mohamed Aly Aly Saad
- Department of Electrical and Computer Engineering, Georgia Tech Shenzhen Institute (GTSI), Shenzhen, Guangdong 518052, China
| | - Seong-Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Nguyen DT, Kim MH, Baek MJ, Kang NW, Kim DD. Preparation and evaluation of proliposomes formulation for enhancing the oral bioavailability of ginsenosides. J Ginseng Res 2024; 48:417-424. [PMID: 39036737 PMCID: PMC11259707 DOI: 10.1016/j.jgr.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/22/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Background This research main objective was to evaluate a proliposomes (PLs) formulation for the enhancement of oral bioavailability of ginsenosides, using ginsenoside Rg3 (Rg3) as a marker. Methods A novel PLs formulation was prepared using a modified evaporation-on-matrix method. Soy phosphatidylcholine, Rg3-enriched extract, poloxamer 188 (Lutrol® F 68) and sorbitol were mixed and dissolved using a aqueous ethanolic solution, followed by the removal of ethanol and lyophilization. The characterization of Rg3-PLs formulations was performed by powder X-ray diffractometry (PXRD), transmission electron microscopy (TEM) and in vitro release. The enhancement of oral bioavailability was investigated and analyzed by non-compartmental parameters after oral administration of the formulations. Results PXRD of Rg3-PLs indicated that Rg3 was transformed from crystalline into its amorphous form during the preparation process. The Rg3-encapsulated liposomes with vesicular-shaped morphology were generated after the reconstitution by gentle hand-shaking in water; they had a mean diameter of approximately 350 nm, a negative zeta potential (-28.6 mV) and a high entrapment efficiency (97.3%). The results of the in vitro release study exhibited that significantly more amount of Rg3 was released from the PLs formulation in comparison with that from the suspension of Rg3-enriched extract (control group). The pharmacokinetic parameters after oral administration of PLs formulation in rats showed an approximately 11.8-fold increase in the bioavailability of Rg3, compared to that of the control group. Conclusion The developed PLs formulation could be a favorable delivery system to improve the oral bioavailability of ginsenosides, including Rg3.
Collapse
Affiliation(s)
- Duy-Thuc Nguyen
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Min-Hwan Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Min-Jun Baek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Nae-Won Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Lin W, Yan Y, Huang Q, Zheng D. MDMX in Cancer: A Partner of p53 and a p53-Independent Effector. Biologics 2024; 18:61-78. [PMID: 38318098 PMCID: PMC10839028 DOI: 10.2147/btt.s436629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
The p53 tumor suppressor protein plays an important role in physiological and pathological processes. MDM2 and its homolog MDMX are the most important negative regulators of p53. Many studies have shown that MDMX promotes the growth of cancer cells by influencing the regulation of the downstream target gene of tumor suppressor p53. Studies have found that inhibiting the MDMX-p53 interaction can effectively restore the tumor suppressor activity of p53. MDMX has growth-promoting activities without p53 or in the presence of mutant p53. Therefore, it is extremely important to study the function of MDMX in tumorigenesis, progression and prognosis. This article mainly reviews the current research progress and mechanism on MDMX function, summarizes known MDMX inhibitors and provides new ideas for the development of more specific and effective MDMX inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Wu Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Yuxiang Yan
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Qingling Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People’s Republic of China
| |
Collapse
|
4
|
Ben-Eltriki M, Shankar G, Tomlinson Guns ES, Deb S. Pharmacokinetics and pharmacodynamics of Rh2 and aPPD ginsenosides in prostate cancer: a drug interaction perspective. Cancer Chemother Pharmacol 2023; 92:419-437. [PMID: 37709921 DOI: 10.1007/s00280-023-04583-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Ginsenoside Rh2 and its aglycon (aPPD) are one of the major metabolites from Panax ginseng. Preclinical studies suggest that Rh2 and aPPD have antitumor effects in prostate cancer (PCa). Our aims in this review are (1) to describe the pharmacokinetic (PK) properties of Rh2 and aPPD ginsenosides; 2) to provide an overview of the preclinical findings on the use of Rh2 and aPPD in the treatment of PCa; and (3) to highlight the mechanisms of its PK and pharmacodynamic (PD) drug interactions. Increasing evidence points to the potential efficacy of Rh2 or aPPD for PCa treatment. Based on the laboratory studies, Rh2 or aPPD combinations revealed an additive or synergistic interaction or enhanced sensitivity of anticancer drugs toward PCa. This review reveals that enhanced anticancer activities were demonstrated in preclinical studies through interactions of Rh2 and/or aPPD with the proteins related to PK (e.g., cytochrome P450 enzymes, transporters) or PD of the other anticancer drugs or PCa signaling pathways. In conclusion, combining Rh2 or aPPD with anti-prostate cancer drugs leads to PK or PD interactions which could facilitate either therapeutically beneficial or toxic effects.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC, Canada.
- Community Pharmacist, Vancouver Area, BC, Canada.
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Gehana Shankar
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| |
Collapse
|
5
|
Hu QR, Hong H, Zhang ZH, Feng H, Luo T, Li J, Deng ZY, Chen F. Methods on improvements of the poor oral bioavailability of ginsenosides: Pre-processing, structural modification, drug combination, and micro- or nano- delivery system. J Ginseng Res 2023; 47:694-705. [PMID: 38107396 PMCID: PMC10721471 DOI: 10.1016/j.jgr.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 12/19/2023] Open
Abstract
Panax ginseng Meyer is a traditional Chinese medicine that is widely used as tonic in Asia. The main pharmacologically active components of ginseng are the dammarane-type ginsenosides, which have been shown to have anti-cancer, anti-inflammatory, immunoregulatory, neuroprotective, and metabolic regulatory activities. Moreover, some of ginsenosides (eg, Rh2 and Rg3) have been developed into nutraceuticals. However, the utilization of ginsenosides in clinic is restrictive due to poor permeability in cells and low bioavailability in human body. Obviously, the dammarane skeleton and glycosyls of ginsenosides are responsible for these limitations. Therefore, improving the oral bioavailability of ginsenosides has become a pressing issue. Here, based on the structures of ginsenosides, we summarized the understanding of the factors affecting the oral bioavailability of ginsenosides, introduced the methods to enhance the oral bioavailability and proposed the future perspectives on improving the oral bioavailability of ginsenosides.
Collapse
Affiliation(s)
- Qi-rui Hu
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Huan Hong
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Zhi-hong Zhang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Hua Feng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Ze-yuan Deng
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Fang Chen
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
Balusamy SR, Perumalsamy H, Huq MA, Yoon TH, Mijakovic I, Thangavelu L, Yang DC, Rahimi S. A comprehensive and systemic review of ginseng-based nanomaterials: Synthesis, targeted delivery, and biomedical applications. Med Res Rev 2023; 43:1374-1410. [PMID: 36939049 DOI: 10.1002/med.21953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 11/22/2022] [Accepted: 02/26/2023] [Indexed: 03/21/2023]
Abstract
Among 17 Panax species identified across the world, Panax ginseng (Korean ginseng), Panax quinquefolius (American ginseng), and Panax notoginseng (Chinese ginseng) are highly recognized for the presence of bioactive compound, ginsenosides and their pharmacological effects. P. ginseng is widely used for synthesis of different types of nanoparticles compared to P. quinquefolius and P. notoginseng. The use of nano-ginseng could increase the oral bioavailability, membrane permeability, and thus provide effective delivery of ginsenosides to the target sites through transport system. In this review, we explore the synthesis of ginseng nanoparticles using plant extracts from various organs, microbes, and polymers, as well as their biomedical applications. Furthermore, we highlight transporters involved in transport of ginsenoside nanoparticles to the target sites. Size, zeta potential, temperature, and pH are also discussed as the critical parameters affecting the quality of ginseng nanoparticles synthesis.
Collapse
Affiliation(s)
- Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Seoul, Gwangjin-gu, Republic of Korea
| | - Haribalan Perumalsamy
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
- Institute for Next Generation Material Design, Hanyang University, Seoul, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Md Amdadul Huq
- Department of Food and Nutrition, Chung Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Tae Hyun Yoon
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
- Institute for Next Generation Material Design, Hanyang University, Seoul, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Ivan Mijakovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamilnadu, India
| | - Deok Chun Yang
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Shadi Rahimi
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
7
|
Ivanenkov YA, Kukushkin ME, Beloglazkina AA, Shafikov RR, Barashkin AA, Ayginin AA, Serebryakova MS, Majouga AG, Skvortsov DA, Tafeenko VA, Beloglazkina EK. Synthesis and Biological Evaluation of Novel Dispiro-Indolinones with Anticancer Activity. Molecules 2023; 28:molecules28031325. [PMID: 36770991 PMCID: PMC9919490 DOI: 10.3390/molecules28031325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Novel variously substituted thiohydantoin-based dispiro-indolinones were prepared using a regio- and diastereoselective synthetic route from 5-arylidene-2-thiohydantoins, isatines, and sarcosine. The obtained molecules were subsequently evaluated in vitro against the cancer cell lines LNCaP, PC3, HCTwt, and HCT(-/-). Several compounds demonstrated a relatively high cytotoxic activity vs. LNCaP cells (IC50 = 1.2-3.5 µM) and a reasonable selectivity index (SI = 3-10). Confocal microscopy revealed that the conjugate of propargyl-substituted dispiro-indolinone with the fluorescent dye Sulfo-Cy5-azide was mainly localized in the cytoplasm of HEK293 cells. P388-inoculated mice and HCT116-xenograft BALB/c nude mice were used to evaluate the anticancer activity of compound 29 in vivo. Particularly, the TGRI value for the P388 model was 93% at the final control timepoint. No mortality was registered among the population up to day 31 of the study. In the HCT116 xenograft model, the compound (170 mg/kg, i.p., o.d., 10 days) provided a T/C ratio close to 60% on day 8 after the treatment was completed. The therapeutic index-estimated as LD50/ED50-for compound 29 in mice was ≥2.5. Molecular docking studies were carried out to predict the possible binding modes of the examined molecules towards MDM2 as the feasible biological target. However, such a mechanism was not confirmed by Western blot data and, apparently, the synthesized compounds have a different mechanism of cytotoxic action.
Collapse
Affiliation(s)
- Yan A. Ivanenkov
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute (VNIIA), 22. ul. Sushchevskaya, 127055 Moscow, Russia
| | - Maxim E. Kukushkin
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | | | - Radik R. Shafikov
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, GSP-7, Ulitsa Mklukho-Maklaya 16/10, 17997 Moscow, Russia
- A. N. Belozersky Research Institute of Physico-Chemical Biology MSU, Leninskye Gory, House 1, Building 40, 119992 Moscow, Russia
| | - Alexander A. Barashkin
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Andrey A. Ayginin
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Marina S. Serebryakova
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Alexander G. Majouga
- College of New Materials and Nanotechnologies, National University of Science and Technology MISiS, 119049 Moscow, Russia
| | - Dmitry A. Skvortsov
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Viktor A. Tafeenko
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Elena K. Beloglazkina
- Chemistry Department, Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
- Correspondence:
| |
Collapse
|
8
|
Murugesan M, Mathiyalagan R, Boopathi V, Kong BM, Choi SK, Lee CS, Yang DC, Kang SC, Thambi T. Production of Minor Ginsenoside CK from Major Ginsenosides by Biotransformation and Its Advances in Targeted Delivery to Tumor Tissues Using Nanoformulations. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12193427. [PMID: 36234555 PMCID: PMC9565578 DOI: 10.3390/nano12193427] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/24/2022] [Accepted: 09/25/2022] [Indexed: 05/13/2023]
Abstract
For over 2000 years, ginseng (roots of Panax ginseng C.A. Meyer) has been used as a traditional herbal medicine. Ginsenosides are bioactive compounds present in ginseng responsible for the pharmacological effects and curing various acute diseases as well as chronic diseases including cardiovascular disease, cancer and diabetes. Structurally, ginsenosides consist of a hydrophobic aglycone moiety fused with one to four hydrophilic glycoside moieties. Based on the position of sugar units and their abundance, ginsenosides are classified into major and minor ginsenosides. Despite the great potential of ginsenosides, major ginsenosides are poorly absorbed in the blood circulation, resulting in poor bioavailability. Interestingly, owing to their small molecular weight, minor ginsenosides exhibit good permeability across cell membranes and bioavailability. However, extremely small quantities of minor ginsenosides extracted from ginseng plants cannot fulfill the requirement of scientific and clinical studies. Therefore, the production of minor ginsenosides in mass production is a topic of interest. In addition, their poor solubility and lack of targetability to tumor tissues limits their application in cancer therapy. In this review, various methods used for the transformation of major ginsenosides to minor ginsenoside compound K (CK) are summarized. For the production of CK, various transformation methods apply to major ginsenosides. The challenges present in these transformations and future research directions for producing bulk quantities of minor ginsenosides are discussed. Furthermore, attention is also paid to the utilization of nanoformulation technology to improve the bioavailability of minor ginsenoside CK.
Collapse
Affiliation(s)
- Mohanapriya Murugesan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
| | - Vinothini Boopathi
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
| | - Byoung Man Kong
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
| | - Sung-Keun Choi
- Daedong Korea Ginseng Co., Ltd., 86, Gunbuk-ro, Gunbuk-myeon, Geumsan-gun 32718, Chungcheongnam-do, Korea
| | - Chang-Soon Lee
- Daedong Korea Ginseng Co., Ltd., 86, Gunbuk-ro, Gunbuk-myeon, Geumsan-gun 32718, Chungcheongnam-do, Korea
| | - Deok Chun Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
| | - Se Chan Kang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
- Correspondence: (S.C.K.); (T.T.)
| | - Thavasyappan Thambi
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Korea
- Correspondence: (S.C.K.); (T.T.)
| |
Collapse
|
9
|
Zhang D, Liu L, Wang J, Zhang H, Zhang Z, Xing G, Wang X, Liu M. Drug-loaded PEG-PLGA nanoparticles for cancer treatment. Front Pharmacol 2022; 13:990505. [PMID: 36059964 PMCID: PMC9437283 DOI: 10.3389/fphar.2022.990505] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022] Open
Abstract
Nanoparticles based on single-component synthetic polymers, such as poly (lactic acid-co-glycolic acid) (PLGA), have been extensively studied for antitumor drug delivery and adjuvant therapy due to their ability to encapsulate and release drugs, as well as passively target tumors. Amphiphilic block co-polymers, such as polyethylene glycol (PEG)-PLGA, have also been used to prepare multifunctional nanodrug delivery systems with prolonged circulation time and greater bioavailability that can encapsulate a wider variety of drugs, including small molecules, gene-targeting drugs, traditional Chinese medicine (TCM) and multi-target enzyme inhibitors, enhancing their antitumor effect and safety. In addition, the surface of PEG-PLGA nanoparticles has been modified with various ligands to achieve active targeting and selective accumulation of antitumor drugs in tumor cells. Modification with two ligands has also been applied with good antitumor effects, while the use of imaging agents and pH-responsive or magnetic materials has paved the way for the application of such nanoparticles in clinical diagnosis. In this work, we provide an overview of the synthesis and application of PEG-PLGA nanoparticles in cancer treatment and we discuss the recent advances in ligand modification for active tumor targeting.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Pharmaceutical Department of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hong Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Gang Xing
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xuan Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Xuan Wang, ; Minghua Liu,
| | - Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- *Correspondence: Xuan Wang, ; Minghua Liu,
| |
Collapse
|
10
|
Zhao Y, Li J, Chen J, Ye M, Jin X. Functional roles of E3 ubiquitin ligases in prostate cancer. J Mol Med (Berl) 2022; 100:1125-1144. [PMID: 35816219 DOI: 10.1007/s00109-022-02229-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/16/2022]
Abstract
Prostate cancer (PCa) is a malignant epithelial tumor of the prostate gland with a high male cancer incidence. Numerous studies indicate that abnormal function of ubiquitin-proteasome system (UPS) is associated with the progression and metastasis of PCa. E3 ubiquitin ligases, key components of UPS, determine the specificity of substrates, and substantial advances of E3 ubiquitin ligases have been reached recently. Herein, we introduce the structures and functions of E3 ubiquitin ligases and summarize the mechanisms of E3 ubiquitin ligases-related PCa signaling pathways. In addition, some progresses in the development of inhibitors targeting E3 ubiquitin ligases are also included.
Collapse
Affiliation(s)
- Yiting Zhao
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China.,Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Jinyun Li
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China
| | - Jun Chen
- Department of Chemoradiotherapy, the Affiliated People's Hospital of Ningbo University, Ningbo, 315040, China
| | - Meng Ye
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China
| | - Xiaofeng Jin
- Department of Oncology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China. .,Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
11
|
Yu D, Qi S, Guan X, Yu W, Yu X, Cai M, Li Q, Wang W, Zhang W, Qin JJ. Inhibition of STAT3 Signaling Pathway by Terphenyllin Suppresses Growth and Metastasis of Gastric Cancer. Front Pharmacol 2022; 13:870367. [PMID: 35401187 PMCID: PMC8993145 DOI: 10.3389/fphar.2022.870367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer is a common type of malignant tumor with a relatively poor prognosis and presents a serious threat to global health. Signal Transducer and Activator of Transcription-3 (STAT3) has been strongly implicated in many cancers, and its constitutive activation promotes growth, angiogenesis, inflammation, and immune evasion. Therefore, considerable efforts have been put into developing effective and safe STAT3 inhibitors. In this study, we performed a virtual screening by molecular docking and found that terphenyllin, a marine-derived natural product, directly interacted with STAT3. We further found that terphenyllin inhibited the phosphorylation and activation of STAT3 and decreased the protein levels of STAT3-dependent target genes, including c-Myc and Cyclin D1. Subsequently, we demonstrated that terphenyllin exerted its potent anticancer efficacy against gastric cancer in vitro and in vivo. Terphenyllin concentration-dependently inhibited growth, proliferation, and colony formation and induced cell cycle arrest and apoptosis of gastric cancer cells in vitro. Moreover, terphenyllin treatment suppressed the tumor growth and metastasis in a gastric cancer orthotopic mouse model without notable toxicity in vivo. Taken together, our results indicated that terphenyllin exerts its anticancer activity by inhibiting the STAT3 signaling pathway and may serve as a potent STAT3 inhibitor for gastric cancer treatment.
Collapse
Affiliation(s)
- Dehua Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Simin Qi
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Wenkai Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xuefei Yu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Maohua Cai
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qinglin Li
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Weiyi Wang
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Weidong Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
12
|
Wang F, Shan Q, Chang X, Li Z, Gui S. Paeonol-loaded PLGA nanoparticles as an oral drug delivery system: Design, optimization and evaluation. Int J Pharm 2021; 602:120617. [PMID: 33887394 DOI: 10.1016/j.ijpharm.2021.120617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/29/2021] [Accepted: 04/15/2021] [Indexed: 01/09/2023]
Abstract
Herein, we report a novel type of NPs by loading paeonol (Pae) into PLGA NPs, to enhance drug stability and oral bioavailability. The paeonol (Pae)-loaded polylactic-co-Gly-colic acid (PLGA) nanoparticles (Pae-PLGA-NPs) were prepared by nanoprecipitation method. The resultant NPs were in spherical shape with an average particle size around 237.7 ± 4.92 nm, and the PDI and zeta potential were 0.110 ± 0.01 and -25.33 ± 1.37 mV, respectively. The encapsulation efficiency (EE) and drug loading (DL) of the Pae-PLGA-NPs were 86.26 ± 1.12 and 12.74 ± 0.37% respectively. The in vitro drug release, in vivo pharmacokinetics and in situ single-pass intestinal perfusion (SPIPs) of Pae-PLGA-NPs was investigated. In vivo, the AUC(0-t), C max, MRT(0-t), and T1/2z of the Pae-PLGA-NPs group were 3.79-, 1.89-, 1.40- and 1.49-fold greater than those of the Pae suspension group, respectively. The in situ single-pass intestinal perfusion of NPs results showed the Ka values in the duodenum, jejunum, ileum and colon were 1.12-, 1.40-, 1.52- and 2.21-fold higher than those of Pae solution, respectively. Moreover, the Papp values of the ileum and colon were 1.27- and 1.31-fold higher than those of the solution group. Such findings suggested the Pae-PLGA-NPs can significantly improve the intestinal absorption characteristics, and have a beneficial effect on oral administration as a nanometer-sized carrier.
Collapse
Affiliation(s)
| | | | - Xiangwei Chang
- Anhui University of Chinese Medicine, Hefei, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics Anhui Education Department (AUCM), China
| | - Zhenbao Li
- Anhui University of Chinese Medicine, Hefei, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics Anhui Education Department (AUCM), China
| | - Shuangying Gui
- Anhui University of Chinese Medicine, Hefei, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics Anhui Education Department (AUCM), China; Anhui Province Key Laboratory of Pharmaceutical Technology and Application (Anhui University of Chinese Medicine), Hefei, China.
| |
Collapse
|
13
|
Luong Huynh D, Nguyen NH, Nguyen CT. Pharmacological properties of ginsenosides in inflammation-derived cancers. Mol Cell Biochem 2021; 476:3329-3340. [PMID: 33900512 DOI: 10.1007/s11010-021-04162-w] [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] [Received: 01/21/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Ginseng is commonly used as an herbal medicine for improvement of life quality. It is also used as a supplemental medication with anti-cancer drugs to enhance chemotherapy efficacy and shows some beneficial effects. Ginsenosides, also known as saponins, are the major active pharmacological compounds found in ginseng and have been extensively using in treatment of not only cancers but also the other inflammatory diseases such as atherosclerosis, diabetes, acute lung injury, cardiovascular, and infectious diseases. The anti-cancer activities of ginsengs and ginsenosides in different types of cancers have been well studied experimentally and clinically. The major anti-cancer mechanisms of ginseng compounds include inhibition of angiogenesis and metastasis as well as induction of cell cycle arrest and apoptosis. Herein, we review and summarize the current knowledge on the pharmacological effects of ginsengs and ginseng-derived compounds in the treatment of cancers. Moreover, the molecular and cellular mechanism(s) by which ginsengs and ginsenosides modulate the immune response in cancer diseases as well as ginsengs-drugs interaction are also discussed.
Collapse
Affiliation(s)
- Do Luong Huynh
- Institute of Research and Development, Duy Tan University, Da Nang, 550000, Vietnam
| | - Nguyen Hoai Nguyen
- Faculty of Biotechnology, Ho Chi Minh City Open University, 97 Vo Van Tan Street, District 3, Ho Chi Minh City, Vietnam
| | - Cuong Thach Nguyen
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
14
|
Zhao J, Duan Z, Ma X, Liu Y, Fan D. Recent advances in systemic and local delivery of ginsenosides using nanoparticles and nanofibers. Chin J Chem Eng 2021. [DOI: 10.1016/j.cjche.2020.11.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
15
|
Xu B, Yuan L, Hu Y, Xu Z, Qin JJ, Cheng XD. Synthesis, Characterization, Cellular Uptake, and In Vitro Anticancer Activity of Fullerenol-Doxorubicin Conjugates. Front Pharmacol 2021; 11:598155. [PMID: 33568999 PMCID: PMC7868567 DOI: 10.3389/fphar.2020.598155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Doxorubicin (DOX) is one of the most commonly used chemotherapeutic agents for treating human cancer. However, its clinical use has been limited by DOX-induced cardiotoxicity as well as other side effects. In the present study, we designed and synthesized the fullerenol (FU)-DOX conjugates and folic acid (FA)-grafted FU-DOX conjugates for improving the selectivity and activity of DOX in cancer cells. We further characterized the physicochemical properties and examined the release kinetics, cellular uptake, and in vitro anticancer activities of FU-DOX and FA-FU-DOX. The results showed that FU-DOX and FA-FU-DOX had a mean diameter of <200 nm and a low polydispersity. Both FU-DOX and FA-FU-DOX exhibited pH sensitivity and their DOX release rates were higher at pH 5.9 vs. pH 7.4. The cellular uptake studies indicated that FU conjugation enhanced the intracellular accumulation of DOX in human hepatocellular carcinoma (HCC) cell lines (BEL-7402 and HepG2) and the immortalized normal human hepatocytes (L02). The conjugation of FA to FU-DOX further promoted the drug internalization in an FR-dependent manner and enhanced the cytotoxicity against HCC cells. In conclusion, the newly prepared FA-FU-DOX conjugates can optimize the safety and efficacy profile of DOX.
Collapse
Affiliation(s)
- Beihua Xu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Hu
- School of Pharmaceutical Sciences, Zhejiang Pharmaceutical College, Ningbo, China
| | - Zhiyuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
16
|
Kermanizadeh A, Jacobsen NR, Murphy F, Powell L, Parry L, Zhang H, Møller P. A Review of the Current State of Nanomedicines for Targeting and Treatment of Cancers: Achievements and Future Challenges. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Fiona Murphy
- Heriot Watt University School of Engineering and Physical Sciences Edinburgh EH14 4AS UK
| | - Leagh Powell
- Heriot Watt University School of Engineering and Physical Sciences Edinburgh EH14 4AS UK
| | - Lee Parry
- Cardiff University European Cancer Stem Cell Research Institute, School of Biosciences Cardiff CF24 4HQ UK
| | - Haiyuan Zhang
- Changchun Institute of Applied Chemistry Laboratory of Chemical Biology Changchun 130022 China
| | - Peter Møller
- University of Copenhagen Department of Public Health Copenhagen DK1014 Denmark
| |
Collapse
|
17
|
Qi W, Yan X, Xu X, Song B, Sun L, Zhao D, Sun L. The effects of cytarabine combined with ginsenoside compound K synergistically induce DNA damage in acute myeloid leukemia cells. Biomed Pharmacother 2020; 132:110812. [PMID: 33059263 DOI: 10.1016/j.biopha.2020.110812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
AML is a kind of hematological malignant tumor that urgently requires different treatment options in order to increase the cure rate and survival rate. Cytarabine (ara-C) is currently the main drug used to treat AML patients and is usually combined with different chemotherapeutic agents. However, due to resistance to ara-C, a new combination is needed to reduce ara-C resistance and improve treatment outcome. As is known to all, ginseng is a traditional Chinese herb; compound K is the principal metabolic product of ginsenoside which also has anti-cancer activity in some cancer cells, while the mechanism is unclear. In our previous study, we found that compound K inhibited AML cell viability and induced apoptosis, and compound K combined with ara-C synergistically induced AML cell proliferation arrest. Thus, we sought to investigate the reason for this by focusing on the mitochondrial dysfunction and DNA damage. In this paper, our results provide a foundation for the clinical evaluation of concomitant administration of compound K and ara-C in order to reduce the resistance to ara-C and improve AML treatment.
Collapse
Affiliation(s)
- Wenxiu Qi
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiuci Yan
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaohao Xu
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bailin Song
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liping Sun
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Daqing Zhao
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China; Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun, Jilin, China.
| |
Collapse
|
18
|
Xu Y, Li X, Gong W, Huang HB, Zhu BW, Hu JN. Construction of Ginsenoside Nanoparticles with pH/Reduction Dual Response for Enhancement of Their Cytotoxicity Toward HepG2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8545-8556. [PMID: 32686932 DOI: 10.1021/acs.jafc.0c03698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The aim of this study is to construct a pH- and reduction-responsive nanodrug delivery system to effectively deliver a ginsenoside (Rh2) and enhance its cytotoxicity against human hepatocarcinoma cells (HepG2). Here, pullulan polysaccharide was grafted by urocanic acid and α-lipoic acid (α-LA) to obtain a copolymer, α-LA-conjugated N-urocanyl pullulan (LA-URPA), which was expected to have pH and redox dual response. Then, the copolymer LA-URPA was used to encapsulate ginsenoside Rh2 to form Rh2 nanoparticles (Rh2 NPs). The results showed that Rh2 NPs exhibited an average size of 119.87 nm with a uniform spherical morphology. Of note, Rh2 NPs showed a high encapsulation efficiency of 86.00%. Moreover, Rh2 NPs possessed excellent pH/reduction dual-responsive drug release under acidic conditions (pH 5.5) and glutathione (GSH) stimulation with a low drug leakage of 14.8% within 96 h. Furthermore, Rh2 NPs with pH/reduction dual response had higher cytotoxicity than Rh2 after incubation with HepG2 cells for 72 h, indicating that Rh2 NPs had a longer circulation time. After the treatment with Rh2 NPs, the excessive increase of reactive oxygen species and the decrease of superoxide dismutase, glutathione (GSH), and mitochondrial membrane potential suggested that the mitochondrial pathway mediated by oxidative stress played a role in this Rh2 NP-induced apoptosis. In conclusion, this study provides a new strategy for improving the application of ginsenoside Rh2 in the food and pharmaceutical fields.
Collapse
Affiliation(s)
- Yu Xu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Xiang Li
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Wei Gong
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Hai-Bo Huang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Bei-Wei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Jiang-Ning Hu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
19
|
Yu DH, Xu ZY, Mo S, Yuan L, Cheng XD, Qin JJ. Targeting MDMX for Cancer Therapy: Rationale, Strategies, and Challenges. Front Oncol 2020; 10:1389. [PMID: 32850448 PMCID: PMC7419686 DOI: 10.3389/fonc.2020.01389] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
The oncogene MDMX, also known as MDM4 is a critical negative regulator of the tumor suppressor p53 and has been implicated in the initiation and progression of human cancers. Increasing evidence indicates that MDMX is often amplified and highly expressed in human cancers, promotes cancer cell growth, and inhibits apoptosis by dampening p53-mediated transcription of its target genes. Inhibiting MDMX-p53 interaction has been found to be effective for restoring the tumor suppressor activity of p53. Therefore, MDMX is becoming one of the most promising molecular targets for developing anticancer therapeutics. In the present review, we mainly focus on the current MDMX-targeting strategies and known MDMX inhibitors, as well as their mechanisms of action and in vitro and in vivo anticancer activities. We also propose other potential targeting strategies for developing more specific and effective MDMX inhibitors for cancer therapy.
Collapse
Affiliation(s)
- De-Hua Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhi-Yuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shaowei Mo
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
20
|
The effects of ginsenosides on platelet aggregation and vascular intima in the treatment of cardiovascular diseases: From molecular mechanisms to clinical applications. Pharmacol Res 2020; 159:105031. [PMID: 32562816 DOI: 10.1016/j.phrs.2020.105031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
Thrombosis initiated by abnormal platelet aggregation is a pivotal pathological event that precedes most cases of cardiovascular diseases (CVD). Recently, growing evidence indicates that platelet could be a potential target for CVD prevention. However, as the conventional antithrombotic management strategy, applications of current antiplatelet agents are somewhat limited by their various side effects, such as bleeding risk and drug resistance. Hence, efforts have been made to search for agents as complementary therapies. Ginsenoside, the principal active component extracted from Panax ginseng, has gained much attention for its regulations on multiple crucial events of platelet aggregation. From structural characteristics to clinical applications, this review anatomized the intrinsic structure-function relationship of antiplatelet potency of ginsenosides, and the involved signal pathways were specifically summarized. Additionally, the emphasis was placed on clinical studies that investigate the antithrombotic efficacy of ginsenosides in the treatment of CVD. Further, a broad overview of approaches for improving the bioavailability of ginsenosides was concluded. Limitations and prospects of current studies were also discussed. This study may provide some new insights into the systematic understanding of ginsenosides in CVD treatment and lay a foundation for future research.
Collapse
|
21
|
Kim D, Park M, Haleem I, Lee Y, Koo J, Na YC, Song G, Lee J. Natural Product Ginsenoside 20(S)-25-Methoxyl-Dammarane-3β, 12β, 20-Triol in Cancer Treatment: A Review of the Pharmacological Mechanisms and Pharmacokinetics. Front Pharmacol 2020; 11:521. [PMID: 32425780 PMCID: PMC7212460 DOI: 10.3389/fphar.2020.00521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Panax ginseng has been used as an herbal medicine for thousands of years. Most of its pharmacological effects are attributed to its constituent ginsenosides, including 20(S)-25-methoxyl-dammarane-3β, 12β, 20-triol (20(S)-25-OCH3-PPD), which is one of the protopanaxadiol type ginsenosides. It has been found to exhibit anticancer effects by interacting with multiple pharmacological pathways, such as the Wnt/β-catenin, MDM2, ERK/MAPK, and STAT3 signaling pathways. However, its therapeutic potential could be limited by its low bioavailability mainly due to its low aqueous solubility. Thus, several studies have been conducted on its pharmacokinetics and its delivery systems, so as to increase its oral bioavailability. In this review, comprehensive information on its varying pharmacological pathways in cancer, as well as its pharmacokinetic behavior and pharmaceutical strategies, is provided. This information would be useful in the understanding of its diverse mechanisms and pharmacokinetics as an anticancer drug, leading to the design of superior 20(S)-25-OCH3-PPD-containing formulations that maximize its therapeutic potential.
Collapse
Affiliation(s)
- Dohyun Kim
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Minwoo Park
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Iqra Haleem
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Younghong Lee
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Jain Koo
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Young Chae Na
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Gidong Song
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| |
Collapse
|
22
|
Zhang J, Wang W, Zhou Y, Yang J, Xu J, Xu Z, Xu B, Yan L, Cheng XD, Li M, Qin JJ. Terphenyllin Suppresses Orthotopic Pancreatic Tumor Growth and Prevents Metastasis in Mice. Front Pharmacol 2020; 11:457. [PMID: 32322210 PMCID: PMC7157903 DOI: 10.3389/fphar.2020.00457] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer (PC) is an aggressive and fatal disease with high incidences of metastasis and recurrence. However, there are no effective treatment options for the majority of PC patients, especially for those with locally advanced tumors and metastatic diseases. Therefore, it is urgently needed to develop safe and effective anti-PC therapeutic agents. We have recently identified a novel marine-derived natural product terphenyllin with potent anti-PC activity. The present study was designed to investigate the efficacy and mechanisms of action of terphenyllin in several human PC cell lines and an orthotopic PC mouse model. The results showed that terphenyllin significantly inhibited the viability of all PC cell lines with minimal effects on a normal human pancreatic cell line (HPNE). We next demonstrated the effects of terphenyllin on colony formation, apoptosis, migration, and invasion in both Panc1 and HPAC cell lines in a concentration-dependent manner. Terphenyllin also suppressed the tumor growth and metastasis in the Panc1 orthotopic mouse model. We further showed the profound effects of terphenyllin on the expression of apoptosis-associated proteins, including Bax, Bad, Puma, BimL, Bcl-2, phos-Bcl-2 (Ser70), Bcl-xL, caspase 7, and PARP, which contributed to its anti-PC activity. In summary, terphenyllin suppressed the PC cell growth and metastasis in vitro and in vivo and may be developed as an anti-PC therapeutic agent in the future.
Collapse
Affiliation(s)
- Jia Zhang
- Shanxi Province Academy of Traditional Chinese Medicine, Taiyuan, China
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weiyi Wang
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Yang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingli Xu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Beihua Xu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Minghua Li
- Shanxi Province Academy of Traditional Chinese Medicine, Taiyuan, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
23
|
Qi SM, Cheng G, Cheng XD, Xu Z, Xu B, Zhang WD, Qin JJ. Targeting USP7-Mediated Deubiquitination of MDM2/MDMX-p53 Pathway for Cancer Therapy: Are We There Yet? Front Cell Dev Biol 2020; 8:233. [PMID: 32300595 PMCID: PMC7142254 DOI: 10.3389/fcell.2020.00233] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/19/2020] [Indexed: 01/13/2023] Open
Abstract
The p53 tumor suppressor protein and its major negative regulators MDM2 and MDMX oncoproteins form the MDM2/MDMX-p53 circuitry, which plays critical roles in regulating cancer cell growth, proliferation, cell cycle progression, apoptosis, senescence, angiogenesis, and immune response. Recent studies have shown that the stabilities of p53, MDM2, and MDMX are tightly controlled by the ubiquitin-proteasome system. Ubiquitin specific protease 7 (USP7), one of the most studied deubiquitinating enzymes plays a crucial role in protecting MDM2 and MDMX from ubiquitination-mediated proteasomal degradation. USP7 is overexpressed in human cancers and contributes to cancer initiation and progression. USP7 inhibition promotes the degradation of MDM2 and MDMX, activates the p53 signaling, and causes cell cycle arrest and apoptosis, making USP7 a potential target for cancer therapy. Several small-molecule inhibitors of USP7 have been developed and shown promising efficacy in preclinical settings. In the present review, we focus on recent advances in the understanding of the USP7-MDM2/MDMX-p53 network in human cancers as well as the discovery and development of USP7 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Si-Min Qi
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Gang Cheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhiyuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Beihua Xu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, Shanghai, China.,Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
24
|
Guo D, Xu P, Chen D, Wang L, Zhu Y, Zuo Y, Chen B. Daunorubicin-Loaded CdTe QDs Conjugated with Anti-CD123 mAbs: A Novel Delivery System for Myelodysplastic Syndromes Treatment. Int J Nanomedicine 2020; 15:521-536. [PMID: 32021192 PMCID: PMC6988587 DOI: 10.2147/ijn.s233395] [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: 10/04/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION The myelodysplastic syndromes (MDS) are a very heterogeneous group of myeloid disorders characterized by peripheral blood cytopenias and increase risk of transformation to acute myeloid leukemia (AML). Daunorubicin (DNR) is an indispensable drug for the treatment of MDS and AML. However, its side effects including cardiac toxicity and bone marrow suppression severely limit clinical application. Many researches reported high expression of CD123 antigen on high-risk MDS cells, so we constructed a novel drug delivery system comprising daunorubicin-loaded CdTe QDs conjugated with anti-CD123 mAbs (DNR-CdTe-CD123) to develop targeted combination chemotherapy for MDS. METHODS CdTe conjugated antiCD123 through amide bond, co-loaded with DNR with electrostatic bonding. Then, we determined characterization and release rate of DNR-CdTe-CD123. The therapeutic effect and side effect of drug delivery system were evaluated through in vitro and in vivo experiments. RESULTS CdTe showed appropriate diameter and good dispersibility and DNR was loaded into CdTes with high encapsulation efficiency and drug loading. The maximum drug loading and encapsulation efficiency were 42.08 ± 0.64% and 74.52 ± 1.81%, respectively, at DNR concentration of 0.2mg/mL and anti-CD123 mAbs volume of 5ul (100ug/mL). Flow cytometry (FCM) showed that CD123 antigen was highly expressed on MUTZ-1 cells, and its expression rate was 72.89 ± 10.67%. In vitro experiments showed that the inhibition rate and apoptosis rate of MUTZ-1 cells treated with DNR-CdTe-CD123 were higher than those in the other groups (P<0.05). Compared with the other groups, the level of apoptosis-related protein (P53, cleaved caspase-9, Bax and cleaved caspase-3) were upregulated in DNR-CdTe-CD123 group (P<0.05). In vivo experiments, DNR-CdTe-CD123 can effectively inhibit the tumor growth of MDS-bearing nude mice and reduce the side effects of DNR on myocardial cells. CONCLUSION The system of DNR-CdTe-CD123 enhances the therapeutic effects and reduce the side effects of DNR, thus providing a novel platform for MDS treatment.
Collapse
Affiliation(s)
- Dan Guo
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu226001, People’s Republic of China
| | - Peipei Xu
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Dangui Chen
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Lili Wang
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Yudi Zhu
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Yifan Zuo
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu210093, People’s Republic of China
| |
Collapse
|
25
|
Peng B, He R, Xu Q, Yang Y, Hu Q, Hou H, Liu X, Li J. Ginsenoside 20(S)-protopanaxadiol inhibits triple-negative breast cancer metastasis in vivo by targeting EGFR-mediated MAPK pathway. Pharmacol Res 2019; 142:1-13. [DOI: 10.1016/j.phrs.2019.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 01/30/2023]
|
26
|
Kamel MEF, Mohammad HMF, Maurice C, Hagras MM. Ginseng Nanoparticles Protect Against Methotrexate-Induced Testicular Toxicity in Rats. EGYPTIAN JOURNAL OF BASIC AND CLINICAL PHARMACOLOGY 2019. [DOI: 10.32527/2019/101397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
27
|
Kim YJ, Perumalsamy H, Markus J, Balusamy SR, Wang C, Ho Kang S, Lee S, Park SY, Kim S, Castro-Aceituno V, Kim SH, Yang DC. Development of Lactobacillus kimchicus DCY51T-mediated gold nanoparticles for delivery of ginsenoside compound K: in vitro photothermal effects and apoptosis detection in cancer cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:30-44. [DOI: 10.1080/21691401.2018.1541900] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Yeon-Ju Kim
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Haribalan Perumalsamy
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Josua Markus
- Graduate School of Biotechnology and Ginseng Bank, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | | | - Chao Wang
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Seong Ho Kang
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Seungah Lee
- Department of Applied Chemistry and Institute of Natural Sciences, College of Applied Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Sang Yong Park
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Sung Kim
- Center for Global Converging Humanities, Kyung Hee University, Yongin-si, Republic of Korea
| | - Verónica Castro-Aceituno
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Seung Hyun Kim
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| | - Deok Chun Yang
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
- Graduate School of Biotechnology and Ginseng Bank, College of Life Science, Kyung Hee University, Yongin-si, Republic of Korea
| |
Collapse
|
28
|
Pharmacokinetic interaction of calcitriol with 20(S)-protopanaxadiol in mice: Determined by LC/MS analysis. Eur J Pharm Sci 2019; 130:173-180. [PMID: 30654110 DOI: 10.1016/j.ejps.2019.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/23/2018] [Accepted: 01/11/2019] [Indexed: 12/31/2022]
Abstract
The physiological and anti-cancer functions of vitamin D3 are accomplished primarily via 1α,25-dihydroxyvitamin D3 (calcitriol), whereas 20(S)-protopanaxadiol (aPPD) is a ginsenoside, which is isolated from Panax ginseng, with potential anti-cancer benefits. In the present study, we report a pharmacokinetic (PK) herb-nutrient interaction between calcitriol and aPPD in mice. A liquid chromatography mass spectrometry (LC/MS) method was developed using 4-phenyl-1,2,4-triazoline-3,5-dione derivatizing agent and we subsequently used the method to quantitate calcitriol in mouse serum. The limit of quantitation was 0.01 ng/ml which is approximately 100 fold lower than the previously reported assay from our laboratory. Calcitriol PK parameters were determined in non-tumor-bearing or C4-2 human prostate tumor-bearing nude mice following oral co-administration of calcitriol either alone or in combination with aPPD. Mice were pretreated with oral aPPD (70 mg/kg) or vehicle control twice daily for seven consecutive days, followed by a single oral dose of 4 μg/kg calcitriol alone or in combination with aPPD. Our PK results demonstrated that co-administration of calcitriol with aPPD (following pre-treatment with vehicle for seven days) resulted in a 35% increase in the area under the curve (AUC0-24 h) and a 41% increase in the maximum serum concentration (Cmax) compared to the calcitriol only group. aPPD therefore significantly increased calcitriol serum exposure. We also saw a reduction in the time required to reach Cmax. In contrast, calcitriol PK in mice co-administered with calcitriol and aPPD as well as those pretreated seven consecutive days with aPPD was no different than that determined for the mice that received vehicle for seven days as pre-treatment. Co-administration of calcitriol with aPPD therefore could increase health benefits of vitamin D3, however any increased risk of hypercalcemia, resulting from this combination approach, requires further investigation. Lastly, we surmise that a cytochrome P450 inhibition-based mechanism may contribute to the observed PK interaction.
Collapse
|
29
|
Kermanizadeh A, Powell LG, Stone V, Møller P. Nanodelivery systems and stabilized solid-drug nanoparticles for orally administered medicine: current landscape. Int J Nanomedicine 2018; 13:7575-7605. [PMID: 30510419 PMCID: PMC6248225 DOI: 10.2147/ijn.s177418] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanoparticles as a means of targeted delivery of therapeutics and imaging agents could greatly enhance the transport of biologically active contents to specific target tissues, while avoiding or reducing potentially undesired side effects. Generally speaking, the oral route of administration is associated with good patient compliance, as it is convenient, economical, noninvasive, and does not require special training. Here, we review the progress of the utilization of nanodelivery-system carriers or stabilized solid-drug nanoparticles following oral administration, with particular attention on toxicological data. Mechanisms of cytotoxicity are discussed and the problem of extrapolating knowledge to human scenarios highlighted. Additionally, issues associated with administration of drugs via the oral route are underlined, while strategies utilized to overcome these are highlighted. This review aims to offer a balanced overview of strategies currently being used in the application of nanosize constructs for oral medical applications.
Collapse
Affiliation(s)
- Ali Kermanizadeh
- NanoSafety Research Group, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK, .,Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark,
| | - Leagh G Powell
- NanoSafety Research Group, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK,
| | - Vicki Stone
- NanoSafety Research Group, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK,
| | - Peter Møller
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark,
| |
Collapse
|
30
|
Pan W, Xue B, Yang C, Miao L, Zhou L, Chen Q, Cai Q, Liu Y, Liu D, He H, Zhang Y, Yin T, Tang X. Biopharmaceutical characters and bioavailability improving strategies of ginsenosides. Fitoterapia 2018; 129:272-282. [PMID: 29883635 DOI: 10.1016/j.fitote.2018.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/29/2018] [Accepted: 06/03/2018] [Indexed: 02/06/2023]
Abstract
Deglycosylation is the most important gastrointestinal metabolism in which ginsenosides are split off from glycosyl moieties by the enzymes secreted from intestinal microflora, and two possible metabolic pathways of protopanaxdiol-type ginsenosides (PPD-type ginsenosides) and protopanaxtriol-type ginsenosides (PPT-type ginsenosides) have been concluded. The former is deglycosylated at C-3 and/or C-20, and transformed to protopanaxdiol (PPD). By comparison, the latter is deglycosylated at C-6 and/or C-20, and eventually transformed to protopanaxtriol (PPT) instead. The pharmacokinetic behavior of PPD-type ginsenosides and PPT-type ginsenosides is different, mainly in a faster absorption and elimination rate of PPT-type ginsenosides, but almost all of ginsenosides have a low oral bioavailability, which is relevant to the properties, the stability in the gastrointestinal tract, membrane permeability and the intestinal and hepatic first-pass effect of ginsenosides. Fortunately, its bioavailability can be improved by means of pharmaceutical strategies, including nanoparticles, liposomes, emulsions, micelles, etc. These drug delivery systems can significantly increase the bioavailability of ginsenosides, as well as controlling or targeting drug release. Ginsenosides are widely used in the treatment of various diseases, the most famous one is the Shen Yi capsule, which is the world's first clinical application of tumor neovascularization inhibitors. Hence, this article aims to draw people's attention on ocotillol-type ginsenosides, which have prominent anti-Alzheimer's disease activity, but have been overlooked previously, such as its representative compound-Pseudoginsenoside F11(PF11), and then provide a reference for the druggability and further developments of ocotillol-type ginsenosides by utilizing the homogeneous structure between dammarane-type ginsenosides and ocotillol-type ginsenosides.
Collapse
Affiliation(s)
- Wenli Pan
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Binli Xue
- Shaanxi Blood Center, Zhuque Street 407, Xi'an 710061, Shaanxi Province, PR China
| | - Chulei Yang
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Linlin Miao
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Lingli Zhou
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Qiuyue Chen
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Qing Cai
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Yi Liu
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Dongchun Liu
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Haibing He
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Yu Zhang
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Tian Yin
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China
| | - Xing Tang
- Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, Liaoning Province, PR China.
| |
Collapse
|
31
|
Han X, Song J, Lian LH, Yao YL, Shao DY, Fan Y, Hou LS, Wang G, Zheng S, Wu YL, Nan JX. Ginsenoside 25-OCH 3-PPD Promotes Activity of LXRs To Ameliorate P2X7R-Mediated NLRP3 Inflammasome in the Development of Hepatic Fibrosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:7023-7035. [PMID: 29929367 DOI: 10.1021/acs.jafc.8b01982] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ginseng is widely used in energy drinks, dietary supplements, and herbal medicines, and its pharmacological actions are related with energy metabolism. As an important modulating energy metabolism pathway, liver X receptors (LXRs) can promote the resolving of hepatic fibrosis and inflammation. The present study aims to evaluate the regulation of 25-OCH3-PPD, a ginsenoside isolated from Panax ginseng, against hepatic fibrosis and inflammation in thioacetamide (TAA)-stimulated mice by activating the LXRs pathway. 25-OCH3-PPD decreases serum ALT/AST levels and improves the histological pathology of liver in TAA-induced mice; attenuates transcripts of pro-fibrogenic markers associated with hepatic stellate cell activation; attenuates the levels of pro-Inflammatory cytokines and blocks apoptosis happened in liver; inhibits NLRP3 inflammasome by affecting P2X7R activation; and regulates PI3K/Akt and LKB1/AMPK-SIRT1. 25-OCH3-PPD also facilitates LX25Rs and FXR activities decreased by TAA stimulation. 25-OCH3-PPD also decreases α-SMA via regulation of LXRs and P2X7R-NLRP3 in vitro. Our data suggest the possibility that 25-OCH3-PPD promotes activity of LXRs to ameliorate P2X7R-mediated NLRP3 inflammasome in the development of hepatic fibrosis.
Collapse
Affiliation(s)
- Xin Han
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Jian Song
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Li-Hua Lian
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - You-Li Yao
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Dan-Yang Shao
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Ying Fan
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Li-Shuang Hou
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Ge Wang
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Shuang Zheng
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Yan-Ling Wu
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
| | - Ji-Xing Nan
- Key Laboratory for Natural Resource of ChangBai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin Province 133002 , China
- Clinical Research Center , Affiliated Hospital of Yanbian University , Yanji , Jilin Province 133002 , China
| |
Collapse
|
32
|
He MH, Chen L, Zheng T, Tu Y, He Q, Fu HL, Lin JC, Zhang W, Shu G, He L, Yuan ZX. Potential Applications of Nanotechnology in Urological Cancer. Front Pharmacol 2018; 9:745. [PMID: 30038573 PMCID: PMC6046453 DOI: 10.3389/fphar.2018.00745] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/19/2018] [Indexed: 01/16/2023] Open
Abstract
Nowadays, the potential scope of nanotechnology in uro-oncology (cancers of the prostate, bladder, and kidney) is broad, ranging from drug delivery, prevention, and diagnosis to treatment. Novel drug delivery methods using magnetic nanoparticles, gold nanoparticles, and polymeric nanoparticles have been investigated in prostate cancer. Additionally, renal cancer treatment may be profoundly influenced by applications of nanotechnology principles. Various nanoparticle-based strategies for kidney cancer therapy have been proposed. Partly due to the dilution of drug concentrations by urine production, causing inadequate drug delivery to tumor cells in the treatment of bladder cancer, various multifunctional bladder-targeted nanoparticles have been developed to enhance therapeutic efficiency. In each of these cancer research fields, nanotechnology has shown several advantages over widely used traditional methods. Different types of nanoparticles improve the solubility of poorly soluble drugs, and multifunctional nanoparticles have good specificity toward prostate, renal, and bladder cancer. Moreover, nanotechnology can also combine with other novel technologies to further enhance effectivity. As our understanding of nanotechnologies grows, additional opportunities to improve the diagnosis and treatment of urological cancer are excepted to arise. In this review, we focus on nanotechnologies with potential applications in urological cancer therapy and highlight clinical areas that would benefit from nanoparticle therapy.
Collapse
Affiliation(s)
- Ming-Hui He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li Chen
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ting Zheng
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yu Tu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qian He
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ju-Chun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wei Zhang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
33
|
Kim H, Lee JH, Kim JE, Kim YS, Ryu CH, Lee HJ, Kim HM, Jeon H, Won HJ, Lee JY, Lee J. Micro-/nano-sized delivery systems of ginsenosides for improved systemic bioavailability. J Ginseng Res 2018; 42:361-369. [PMID: 29983618 PMCID: PMC6026383 DOI: 10.1016/j.jgr.2017.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 02/04/2023] Open
Abstract
Ginsenosides, dammarane-type triterpene saponins obtained from ginseng, have been used as a natural medicine for many years in the Orient due to their various pharmacological activities. However, the therapeutic potential of ginsenosides has been largely limited by the low bioavailability of the natural products caused mainly by low aqueous solubility, poor biomembrane permeability, instability in the gastrointestinal tract, and extensive metabolism in the body. To enhance the bioavailability of ginsenosides, diverse micro-/nano-sized delivery systems such as emulsions, polymeric particles, and vesicular systems have been investigated. The delivery systems improved the bioavailability of ginsenosides by enhancing solubility, permeability, and stability of the natural products. This mini-review aims to provide comprehensive information on the micro-/nano-sized delivery systems for increasing the bioavailability of ginsenosides, which may be helpful for designing better delivery systems to maximize the versatile therapeutic potential of ginsenosides.
Collapse
Affiliation(s)
- Hyeongmin Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Jong Hyuk Lee
- Department of Pharmaceutical Engineering, College of Life and Health Sciences, Hoseo University, Asan, Republic of Korea
| | - Jee Eun Kim
- Graduate School of Pharmaceutical Management, Chung-Ang University, Seoul, Republic of Korea
| | - Young Su Kim
- Graduate School of Pharmaceutical Management, Chung-Ang University, Seoul, Republic of Korea
| | - Choong Ho Ryu
- Graduate School of Pharmaceutical Management, Chung-Ang University, Seoul, Republic of Korea
| | - Hong Joo Lee
- Graduate School of Pharmaceutical Management, Chung-Ang University, Seoul, Republic of Korea
| | - Hye Min Kim
- Graduate School of Pharmaceutical Management, Chung-Ang University, Seoul, Republic of Korea
| | - Hyojin Jeon
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Hyo-Joong Won
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Ji-Yun Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
- Graduate School of Pharmaceutical Management, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
34
|
Jogi H, Maheshwari R, Raval N, Kuche K, Tambe V, Mak KK, Pichika MR, Tekade RK. Carbon nanotubes in the delivery of anticancer herbal drugs. Nanomedicine (Lond) 2018; 13:1187-1220. [DOI: 10.2217/nnm-2017-0397] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer is estimated to be a significant health problem of the 21st century. The situation gets even tougher when it comes to its treatment using chemotherapy employing synthetic anticancer molecules with numerous side effects. Recently, there has been a paradigm shift toward the adoption of herbal drugs for the treatment of cancer. In this context, a suitable delivery system is principally warranted to deliver these herbal biomolecules specifically at the tumorous site. To achieve this goal, carbon nanotubes (CNTs) have been widely explored to deliver anticancer herbal molecules with improved therapeutic efficacy and safety. This review uniquely expounds the biopharmaceutical, clinical and safety aspects of different anticancer herbal drugs delivered through CNTs with a cross-talk on their outcomes. This review will serve as a one-stop-shop for the readers on various anticancer herbal drugs delivered through CNTs as a futuristic delivery device.
Collapse
Affiliation(s)
- Hardi Jogi
- National Institute of Pharmaceutical Education & Research (NIPER) – Ahmedabad, Opposite Air Force Station Palaj, Gandhinagar, Gujarat, 382355 India
| | - Rahul Maheshwari
- National Institute of Pharmaceutical Education & Research (NIPER) – Ahmedabad, Opposite Air Force Station Palaj, Gandhinagar, Gujarat, 382355 India
| | - Nidhi Raval
- National Institute of Pharmaceutical Education & Research (NIPER) – Ahmedabad, Opposite Air Force Station Palaj, Gandhinagar, Gujarat, 382355 India
| | - Kaushik Kuche
- National Institute of Pharmaceutical Education & Research (NIPER) – Ahmedabad, Opposite Air Force Station Palaj, Gandhinagar, Gujarat, 382355 India
| | - Vishakha Tambe
- National Institute of Pharmaceutical Education & Research (NIPER) – Ahmedabad, Opposite Air Force Station Palaj, Gandhinagar, Gujarat, 382355 India
| | - Kit-Kay Mak
- School of Postgraduate Studies & Research, International Medical University, Kuala Lumpur, Malaysia
| | - Mallikarjuna Rao Pichika
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education & Research (NIPER) – Ahmedabad, Opposite Air Force Station Palaj, Gandhinagar, Gujarat, 382355 India
| |
Collapse
|
35
|
20(S)-protopanaxadiol regio-selectively targets androgen receptor: anticancer effects in castration-resistant prostate tumors. Oncotarget 2018; 9:20965-20978. [PMID: 29765513 PMCID: PMC5940378 DOI: 10.18632/oncotarget.24695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/24/2018] [Indexed: 01/21/2023] Open
Abstract
We have explored the effects of 20(S)-protopanaxadiol (aPPD), a naturally derived ginsenoside, against androgen receptor (AR) positive castration resistant prostate cancer (CRPC) xenograft tumors and have examined its interactions with AR. In silico docking studies for aPPD binding to AR, alongside transactivation bioassays and in vivo efficacy studies were carried out in the castration-resistant C4-2 xenograft model. Immunohistochemical (IHC) and Western blot analyses followed by evaluation of AR, apoptotic, cell cycle and proliferative markers in excised tumors was performed. The growth of established CRPC tumors was inhibited by 53% with aPPD and a corresponding decrease in serum PSA was seen compared to controls. The IHC data revealed that Ki-67 was significantly lower for aPPD treated tumors and was associated with elevated p21 and cleaved caspase-3 expression, compared to vehicle treatment. Furthermore, aPPD decreased AR protein expression in xenograft tumors, while significantly upregulating p27 and Bax protein levels. In vitro data supporting this suggests that aPPD binds to and significantly inhibits the N-terminal or the DNA binding domains of AR. The AR androgen binding site docking score for androgen (dihydrotestosterone) was −11.1, while that of aPPD was −7.1. The novel findings described herein indicate aPPD potently inhibits PCa in vivo partly via inhibition of a site on the AR N-terminal domain. This manifested as cell cycle arrest and concurrent induction of apoptosis via an increase in Bax, cleaved-caspase-3, p27 and p21 expression.
Collapse
|
36
|
Qin JJ, Li X, Wang W, Zi X, Zhang R. Targeting the NFAT1-MDM2-MDMX Network Inhibits the Proliferation and Invasion of Prostate Cancer Cells, Independent of p53 and Androgen. Front Pharmacol 2017; 8:917. [PMID: 29311926 PMCID: PMC5735069 DOI: 10.3389/fphar.2017.00917] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/31/2022] Open
Abstract
The MDM2 and MDMX oncogenes are overexpressed in various types of human cancer and are highly associated with the initiation, progression, metastasis and chemotherapeutic resistance of these diseases, including prostate cancer. The present study was designed to test a natural MDM2 inhibitor, Inulanolide A (InuA), for anti-prostate cancer activity and to determine the underlying mechanism(s) of action. InuA directly bound to the RING domains of both MDM2 and MDMX with high affinity and specificity and disrupted MDM2-MDMX binding, markedly enhancing MDM2 protein degradation. We further discovered that InuA bound to the DNA binding domain of NFAT1, resulting in marked inhibition of MDM2 transcription. InuA inhibited the proliferation, migration, and invasion of prostate cancer cells, regardless of their p53 status and AR responsiveness. Double knockdown of MDM2 and NFAT1 also revealed that the expression of both of these molecules is important for InuA’s inhibitory effects on the proliferation and invasion of prostate cancer cells. In summary, InuA represents a novel class of bifunctional MDM2 inhibitors, and should be further investigated as a candidate lead compound for prostate cancer prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Xin Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States.,Center for Drug Discovery, University of Houston, Houston, TX, United States
| | - Xiaolin Zi
- Department of Urology, University of California, Irvine, Irvine, CA, United States.,Department of Pharmacology, University of California, Irvine, Irvine, CA, United States
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States.,Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States.,Center for Drug Discovery, University of Houston, Houston, TX, United States
| |
Collapse
|
37
|
Xia T, Wang J, Wang Y, Wang Y, Cai J, Wang M, Chen Q, Song J, Yu Z, Huang W, Fang J. Inhibition of autophagy potentiates anticancer property of 20(S)-ginsenoside Rh2 by promoting mitochondria-dependent apoptosis in human acute lymphoblastic leukaemia cells. Oncotarget 2017; 7:27336-49. [PMID: 27027340 PMCID: PMC5053654 DOI: 10.18632/oncotarget.8285] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/01/2016] [Indexed: 01/03/2023] Open
Abstract
Acute lymphoblastic leukaemia (ALL) is the most prevalent childhood malignancy. Although most children with ALL are cured, there is still a group of patients for which therapy fails owing to severe toxicities and drug resistance. Ginsenoside Rh2 (GRh2), a major bioactive component isolated from Panax ginseng, has been shown to have a therapeutic effect on some tumors. However, the molecular mechanisms of cell death induced by 20(S)-GRh2 in ALL cells remains unclear. In this study, we showed that 20(S)-GRh2 inhibited the cell growth and induced mitochondria-dependent apoptosis and autophagy. But it has no cytotoxic effect on human normal blood cells. Furthermore, autophagy plays a protective role in 20(S)-GRh2-induced apoptosis in ALL cell lines and human primary ALL cells. We demonstrated that either genetic or pharmacologic inhibition of autophagy could be more effective in reducing viability and enhancing 20(S)-GRh2-induced toxicity than 20(S)-GRh2 treatment alone. In addition, inhibition of autophagy could aggravate mitochondrial ROS generation and mitochondrial damage, and then accelerate mitochondria-dependent apoptosis. Taken together, these results suggest that inhibition of autophagy can sensitize ALL cells towards 20(S)-GRh2. The appropriate inhibition of autophagy could provide a powerful strategy to increase the potency of 20(S)-GRh2 as a novel anticancer agent for ALL therapy.
Collapse
Affiliation(s)
- Ting Xia
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P.R. China
| | - Jiancheng Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Yingnan Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Yuanyuan Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Jianye Cai
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Min Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P.R. China
| | - Qidan Chen
- Department of Chemistry and Pharmacy, Zhuhai College, Jilin University, Zhuhai, P.R. China
| | - Jia Song
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P.R. China
| | - Ziqi Yu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P.R. China
| | - Wei Huang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P.R. China
| | - Jianpei Fang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P.R. China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, P.R. China
| |
Collapse
|
38
|
Almoustafa HA, Alshawsh MA, Chik Z. Technical aspects of preparing PEG-PLGA nanoparticles as carrier for chemotherapeutic agents by nanoprecipitation method. Int J Pharm 2017; 533:275-284. [DOI: 10.1016/j.ijpharm.2017.09.054] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/17/2017] [Accepted: 09/18/2017] [Indexed: 11/15/2022]
|
39
|
20(S)-25-methoxyl-dammarane-3β, 12β, 20-triol negatively regulates activation of STAT3 and ERK pathways and exhibits anti-cancer effects in HepG2 cells. Apoptosis 2017; 22:1404-1418. [DOI: 10.1007/s10495-017-1416-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
40
|
Qin JJ, Wang W, Zhang R. Experimental Therapy of Advanced Breast Cancer: Targeting NFAT1-MDM2-p53 Pathway. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:195-216. [PMID: 29096894 PMCID: PMC6663080 DOI: 10.1016/bs.pmbts.2017.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advanced breast cancer, especially advanced triple-negative breast cancer, is typically more aggressive and more difficult to treat than other breast cancer phenotypes. There is currently no curable option for breast cancer patients with advanced diseases, highlighting the urgent need for novel treatment strategies. We have recently discovered that the nuclear factor of activated T cells 1 (NFAT1) activates the murine double minute 2 (MDM2) oncogene. Both MDM2 and NFAT1 are overexpressed and constitutively activated in breast cancer, particularly in advanced breast cancer, and contribute to its initiation, progression, and metastasis. MDM2 regulates cancer cell proliferation, cell cycle progression, apoptosis, migration, and invasion through both p53-dependent and -independent mechanisms. We have proposed to target the NFAT1-MDM2-p53 pathway for the treatment of human cancers, especially breast cancer. We have recently identified NFAT1 and MDM2 dual inhibitors that have shown excellent in vitro and in vivo activities against breast cancer, including triple-negative breast cancer. Herein, we summarize recent advances made in the understanding of the oncogenic functions of MDM2 and NFAT1 in breast cancer, as well as current targeting strategies and representative inhibitors. We also propose several strategies for inhibiting the NFAT1-MDM2-p53 pathway, which could be useful for developing more specific and effective inhibitors for breast cancer therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- University of Houston, Houston, TX, United States; Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Wei Wang
- University of Houston, Houston, TX, United States; Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Ruiwen Zhang
- University of Houston, Houston, TX, United States; Texas Tech University Health Sciences Center, Amarillo, TX, United States.
| |
Collapse
|
41
|
Xue B, Wang W, Qin JJ, Nijampatnam B, Murugesan S, Kozlovskaya V, Zhang R, Velu SE, Kharlampieva E. Highly efficient delivery of potent anticancer iminoquinone derivative by multilayer hydrogel cubes. Acta Biomater 2017; 58:386-398. [PMID: 28583901 PMCID: PMC5736006 DOI: 10.1016/j.actbio.2017.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/15/2017] [Accepted: 06/02/2017] [Indexed: 01/04/2023]
Abstract
We report a novel delivery platform for a highly potent anticancer drug, 7-(benzylamino)-3,4-dihydro-pyrrolo[4,3,2-de]quinolin-8(1H)-one (BA-TPQ), using pH- and redox-sensitive poly(methacrylic acid) (PMAA) hydrogel cubes of micrometer size as the encapsulating matrix. The hydrogels are obtained upon cross-linking PMAA with cystamine in PMAA/poly(N-vinylpyrrolidone) multilayers assembled within mesoporous sacrificial templates. The BA-TPQ-loaded hydrogels maintain their cubical shape and pH-sensitivity after lyophilization, which is advantageous for long-term storage. Conversely, the particles degrade in vitro in the presence of glutathione (5mM) providing 80% drug release within 24h. Encapsulating BA-TPQ into hydrogels significantly increases its transport via Caco-2 cell monolayers used as a model for oral delivery where the apparent permeability of BA-TPQ-hydrogel cubes was∼2-fold higher than that of BA-TPQ. BA-TPQ-hydrogel cubes exhibit better anticancer activity against HepG2 (IC50=0.52µg/mL) and Huh7 (IC50=0.29µg/mL) hepatoma cells with a 40% decrease in the IC50 compared to the non-encapsulated drug. Remarkably, non-malignant liver cells have a lower sensitivity to BA-TPQ-hydrogel cubes with 2-fold increased IC50 values compared to those of cancer cells. In addition, encapsulating BA-TPQ in the hydrogels amplifies the potency of the drug via down-regulation of MDM2 oncogenic protein and upregulation of p53 (a tumor suppressor) and p21 (cell proliferation suppressor) expression in HepG2 liver cancer cells. Moreover, enhanced inhibition of MDM2 protein expression by BA-TPQ-hydrogel cubes is independent of p53 status in Huh7 cells. This drug delivery platform of non-spherical shape provides a facile method for encapsulation of hydrophobic drugs and can facilitate the enhanced efficacy of BA-TPQ for liver cancer therapy. STATEMENT OF SIGNIFICANCE Many potent anticancer drugs are hydrophobic and lack tumor selectivity, which limits their application in cancer therapy. Although cubical hydrogels of poly(methacrylic acid) exhibit excellent biocompatibility and versatility, they have not been investigated for hydrophobic drug delivery due to poor mechanical stability and incompatibility between hydrophobic drugs and a hydrophilic hydrogel network. In this study, we provide a facile method to prepare a multilayer hydrogel-based platform with controlled nanostructure, cubical shape and redox-responsiveness for delivery of highly potent anticancer therapeutics, hydrophobic BA-TPQ. The BA-TPQ-hydrogel cubes have exceptional structural stability upon lyophilization which is advantageous for a long-term storage. The greatly enhanced trans-epithelial permeability and amplified anti-tumor activity of BA-TPQ are achieved by encapsulation in these hydrogel cubes. Furthermore, the anticancer BA-TPQ-hydrogel platform retains the selective activity of BA-TPQ to hepatocellular carcinoma cells. Overall, the produced BA-TPQ-hydrogel cubes demonstrate a high potential for clinical liver cancer therapy.
Collapse
Affiliation(s)
- Bing Xue
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States
| | - Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States
| | - Bhavitavya Nijampatnam
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Srinivasan Murugesan
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States.
| | - Sadanandan E Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294-3300, United States.
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Center of Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
42
|
Mathiyalagan R, Yang DC. Ginseng nanoparticles: a budding tool for cancer treatment. Nanomedicine (Lond) 2017; 12:1091-1094. [PMID: 28447867 DOI: 10.2217/nnm-2017-0070] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Ramya Mathiyalagan
- Graduate School of Biotechnology & Ginseng Bank, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Deok Chun Yang
- Graduate School of Biotechnology & Ginseng Bank, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
43
|
Doxorubicin-loaded platelets conjugated with anti-CD22 mAbs: a novel targeted delivery system for lymphoma treatment with cardiopulmonary avoidance. Oncotarget 2017; 8:58322-58337. [PMID: 28938559 PMCID: PMC5601655 DOI: 10.18632/oncotarget.16871] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/14/2017] [Indexed: 01/18/2023] Open
Abstract
B-cell lymphoma accounts for approximately 85% of all adult non-Hodgkin's lymphoma cases. Doxorubicin (DOX) is an indispensable drug for the treatment of non-Hodgkin's lymphoma. However, DOX causes severe cardiotoxicity, which limits its use in conventional treatment strategies. In this study, we developed a novel drug delivery system for lymphoma treatment: DOX-loaded platelets that were conjugated with anti-CD22 monoclonal antibodies (mAbs) (DOX-platelet-CD22). Platelets are bio- and immune-compatible drug carriers that can prolong the circulation time of drugs. Anti-CD22 mAb-labeled platelets can precisely deliver DOX to tumor cells. Our in vitro and in vivo experiments showed the enhanced antitumor activity and attenuated cardiotoxicity of DOX when delivered as DOX-platelet-CD22. Compared with other delivery systems, the uptake of DOX-platelet-CD22 by macrophage-like cells decreased. Moreover, DOX-platelet-CD22 showed platelet properties, such as tumor cell-induced platelet aggregation. Therefore, targeted chemotherapy that is mediated by DOX-platelet-CD22 is a promising option for lymphoma treatment.
Collapse
|
44
|
Enhanced and Extended Anti-Hypertensive Effect of VP5 Nanoparticles. Int J Mol Sci 2016; 17:ijms17121977. [PMID: 27898022 PMCID: PMC5187777 DOI: 10.3390/ijms17121977] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/11/2016] [Accepted: 11/18/2016] [Indexed: 02/05/2023] Open
Abstract
Hypertension has become a significant global public health concern and is also one of the most common risk factors of cardiovascular disease. Recent studies have shown the promising result of peptides inhibiting angiotensin converting enzyme (ACE) in lowering the blood pressure in both animal models and humans. However, the oral bioavailability and continuous antihypertensive effectiveness require further optimization. Novel nanoparticle-based drug delivery systems are helpful to overcome these barriers. Therefore, a poly-(lactic-co-glycolic) acid nanoparticle (PLGANPs) oral delivery system, of the antihypertensive small peptides Val-Leu-Pro-Val-Pro (VLPVP, VP5) model, was developed in this study and its antihypertensive effect was investigated in spontaneously hypertensive rats (SHRs) for the first time. The obtained VP5 nanoparticles (VP5-NPs) showed a small particle size of 223.7 ± 2.3 nm and high entrapment efficiency (EE%) of 87.37% ± 0.92%. Transmission electronic microscopy (TEM) analysis showed that the nanoparticles were spherical and homogeneous. The optimal preparation of VP5-NPs exhibited sustained release of VP5 in vitro and a 96 h long-term antihypertensive effect with enhanced efficacy in vivo. This study illustrated that PLGANPs might be an optimal formulation for oral delivery of antihypertensive small peptides and VP5-NPs might be worthy of further development and use as a potential therapeutic strategy for hypertension in the future.
Collapse
|
45
|
McCubrey JA, Lertpiriyapong K, Fitzgerald TL, Martelli AM, Cocco L, Rakus D, Gizak A, Libra M, Cervello M, Montalto G, Yang LV, Abrams SL, Steelman LS. Roles of TP53 in determining therapeutic sensitivity, growth, cellular senescence, invasion and metastasis. Adv Biol Regul 2016; 63:32-48. [PMID: 27776972 DOI: 10.1016/j.jbior.2016.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/20/2022]
Abstract
TP53 is a critical tumor suppressor gene that regulates cell cycle progression, apoptosis, cellular senescence and many other properties critical for control of normal cellular growth and death. Due to the pleiotropic effects that TP53 has on gene expression and cellular physiology, mutations at this tumor suppressor gene result in diverse physiological effects. T53 mutations are frequently detected in numerous cancers. The expression of TP53 can be induced by various agents used to treat cancer patients such as chemotherapeutic drugs and ionizing radiation. Radiation will induce Ataxia telangiectasia mutated (ATM) and other kinases that results in the phosphorylation and activation of TP53. TP53 is also negatively regulated by other mechanisms, such as ubiquitination by ligases such as MDM2. While TP53 has been documented to control the expression of many "classical" genes (e.g., p21Cip-1, PUMA, Bax) by transcriptional mechanisms for quite some time, more recently TP53 has been shown to regulate microRNA (miR) gene expression. Different miRs can promote oncogenesis (oncomiR) whereas others act to inhibit tumor progression (tumor suppressor miRs). Targeted therapies to stabilize TP53 have been developed by various approaches, MDM2/MDM4 inhibitors have been developed to stabilize TP53 in TP53-wild type (WT) tumors. In addition, small molecules have been isolated that will reactivate certain mutant TP53s. Both of these types of inhibitors are in clinical trials. Understanding the actions of TP53 may yield novel approaches to suppress cancer, aging and other health problems.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Massimo Libra
- Department of Bio-Medical Sciences, University of Catania, Catania, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Guiseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
46
|
Shen J, Bi J, Tian H, Jin Y, Wang Y, Yang X, Yang Z, Kou J, Li F. Preparation and evaluation of a self-nanoemulsifying drug delivery system loaded with Akebia saponin D-phospholipid complex. Int J Nanomedicine 2016; 11:4919-4929. [PMID: 27713630 PMCID: PMC5045231 DOI: 10.2147/ijn.s108765] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Akebia saponin D (ASD) exerts various pharmacological activities but with poor oral bioavailability. In this study, a self-nanoemulsifying drug delivery system (SNEDDS) based on the drug-phospholipid complex technique was developed to improve the oral absorption of ASD. METHODS ASD-phospholipid complex (APC) was prepared using a solvent-evaporation method and characterized by infrared spectroscopy, differential scanning calorimetry, morphology observation, and solubility test. Oil and cosurfactant were selected according to their ability to dissolve APC, while surfactant was chosen based on its emulsification efficiency in SNEDDS. Pseudoternary phase diagrams were constructed to determine the optimized APC-SNEDDS formulation, which was characterized by droplet size determination, zeta potential determination, and morphology observation. Robustness to dilution and thermodynamic stability of optimized formulation were also evaluated. Subsequently, pharmacokinetic parameters and oral bioavailability of ASD, APC, and APC-SNEDDS were investigated in rats. RESULTS The liposolubility significantly increased 11.4-fold after formation of APC, which was verified by the solubility test in n-octanol. Peceol (Glyceryl monooleate [type 40]), Cremophor® EL (Polyoxyl 35 castor oil), and Transcutol HP (Diethylene glycol monoethyl ether) were selected as oil, surfactant, and cosurfactant, respectively. The optimal formulation was composed of Glyceryl monooleate (type 40), Polyoxyl 35 castor oil, Diethylene glycol monoethyl ether, and APC (1:4.5:4.5:1.74, w/w/w/w), which showed a particle size of 148.0±2.7 nm and a zeta potential of -13.7±0.92 mV after dilution with distilled water at a ratio of 1:100 (w/w) and good colloidal stability. Pharmacokinetic studies showed that APC-SNEDDS exhibited a significantly greater Cmax1 (733.4±203.8 ng/mL) than ASD (437.2±174.2 ng/mL), and a greater Cmax2 (985.8±366.6 ng/mL) than ASD (180.5±75.1 ng/mL) and APC (549.7±113.5 ng/mL). Compared with ASD, Tmax1 and Tmax2 were both remarkably shortened by APC-SNEDDS. The oral bioavailability in rats was enhanced significantly to 183.8% and 431.8% by APC and APC-SNEDDS, respectively. CONCLUSION These results indicated that APC-SNEDDS was a promising drug delivery system to enhance the oral bioavailability of ASD.
Collapse
Affiliation(s)
- Jinyang Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing
| | - Jianping Bi
- Shandong Provincial Traditional Chinese Medical Hospital & Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan
| | - Hongli Tian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing
| | - Ye Jin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing
| | - Yuan Wang
- Traditional Chinese Medical Hospital of Pukou District
| | - Xiaolin Yang
- Key Laboratory of Pharmaceutical and Biological Marine Resources Research and Development of Jiangsu Province, Nanjing University of Chinese Medicine
| | - Zhonglin Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing
| |
Collapse
|
47
|
Qin JJ, Wang W, Voruganti S, Wang H, Zhang WD, Zhang R. Inhibiting NFAT1 for breast cancer therapy: New insights into the mechanism of action of MDM2 inhibitor JapA. Oncotarget 2016; 6:33106-19. [PMID: 26461225 PMCID: PMC4741752 DOI: 10.18632/oncotarget.5851] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/28/2015] [Indexed: 01/22/2023] Open
Abstract
Transcription factor NFAT1 has been recently identified as a new regulator of the MDM2 oncogene. Targeting the NFAT1-MDM2 pathway represents a novel approach to cancer therapy. We have recently identified a natural product MDM2 inhibitor, termed JapA. As a specific and potent MDM2 inhibitor, JapA inhibits MDM2 at transcriptional and post-translational levels. However, the molecular mechanism remains to be fully elucidated for its inhibitory effects on MDM2 transcription. Herein, we reported that JapA inhibited NFAT1 and NFAT1-mediated MDM2 transcription, which contributed to the anticancer activity of JapA. Its effects on the expression and activity of NFAT1 were examined in various breast cancer cell lines in vitro and in MCF-7 and MDA-MB-231 xenograft tumors in vivo. The specificity of JapA in targeting NFAT1 and NFAT1-MDM2 pathway and the importance of NFAT1 inhibition in JapA's anticancer activity were demonstrated using NFAT1 overexpression and knockdown cell lines and the pharmacological activators and inhibitors of NFAT1 signaling. Our results indicated that JapA inhibited NFAT1 signaling in breast cancer cells in vitro and in vivo, which plays a pivotal role in its anticancer activity. JapA inhibited the nuclear localization of NFAT1, disrupted the NFAT1-MDM2 P2 promoter complex, and induced NFAT1 proteasomal degradation, resulting in the repression of MDM2 transcription. In conclusion, JapA is a novel NFAT1 inhibitor and the NFAT1 inhibition is responsible for the JapA-induced repression of MDM2 transcription, contributing to its anticancer activity. The results may pave an avenue for validating the NFAT1-MDM2 pathway as a novel molecular target for cancer therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.,Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.,Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Sukesh Voruganti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Hui Wang
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Wei-Dong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.,Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| |
Collapse
|
48
|
Yin T, Cao X, Liu X, Wang J, Shi C, Su J, Zhang Y, Gou J, He H, Guo H, Tang X, Zhao Y. Interfacial molecular interactions based on the conformation recognition between the insoluble antitumor drug AD-1 and DSPC. Colloids Surf B Biointerfaces 2016; 146:902-9. [PMID: 27469574 DOI: 10.1016/j.colsurfb.2016.07.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/12/2016] [Accepted: 07/19/2016] [Indexed: 11/29/2022]
Abstract
In this study, molecular interactions between the anti-cancer agent 20(R)-25-methoxyl-dammarane-3β, 12β, 20-triol (AD-1) and phospholipid 1,2-Distearoyl-sn-glycero-3-phosphocholine (DSPC) were investigated using the Langmuir film balance technique. The characteristics of binary Langmuir monolayers consisting of DSPC and AD-1 were conducted on the basis of the surface pressure-area per molecule (π-A) isotherms. It was found that the drug was able to become efficiently inserted into preformed DSPC monolayers, indicating a preferential interaction between AD-1 and DSPC. For the examined lateral pressure at 20mN/m, the largest negative values of ΔGex were found for the AD-1/DSPC monolayer, which should be the most stable. Based on the calculated values of ΔGex, we found that the AD-1/DSPC systems exhibited the best mixed characteristics when the molar fraction of the AD-1 was 0.8; at that relative concentration, the AD-1 molecules can mix better and interact with the phospholipid molecules. In addition, the drug-DSPC binary supramolecular structure was also deposited on the mica plates as shown by atomic force microscopy (AFM). Finally, molecular docking calculations explained satisfactorily that, based on the conformations interactions (conformation recognition), even at an AD-1/DSPC molar ratio as high as 8:2, the interfacial stabilization of the AD-1/DSPC system was fairly strong due to hydrophobic interactions. A higher loading capacity of DSPC might be possible, as it is associated with a more flexible geometrical environment, which allows these supramolecular structures to accept larger increases in drug loading upon steric binding.
Collapse
Affiliation(s)
- Tian Yin
- School of Functional food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China; School of Chinese Materia Media, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiuxiu Cao
- School of Chinese Materia Media, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaolin Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jian Wang
- Key Laboratory of Structure-based Drug Design and Discovery, Shenyang Pharmaceutical University, ministry of Education, Shenyang 110016, China
| | - Caihong Shi
- School of Functional food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jia Su
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingxin Gou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haibing He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haiyan Guo
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yuqing Zhao
- School of Functional food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
49
|
Qin JJ, Wang W, Sarkar S, Zhang R. Oral delivery of anti-MDM2 inhibitor SP141-loaded FcRn-targeted nanoparticles to treat breast cancer and metastasis. J Control Release 2016; 237:101-14. [PMID: 27394681 DOI: 10.1016/j.jconrel.2016.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/27/2016] [Accepted: 07/05/2016] [Indexed: 01/21/2023]
Abstract
We have recently discovered a specific Murine Double Minute 2 (MDM2) oncogene inhibitor, called SP141, which exerts potent anticancer activity in various breast cancer models. However, its low oral bioavailability is the major hurdle for moving this drug to clinical trial. The present study was designed to discover and validate a novel nano-oral delivery system for this promising anticancer agent. Herein, we report the preparation, characterization, and evaluation of the efficacy and safety of the SP141-loaded IgG Fc-conjugated maleimidyl-poly(ethylene glycol)-co-poly(ε-caprolactone) (Mal-PEG-PCL) nanoparticles (SP141FcNP) as an orally cancer therapeutic agent. Our results indicated that SP141FcNP showed a biphasic release pattern and increased transepithelial transport in vitro and in vivo with the involvement of FcRn-mediated transcytosis. SP141FcNP also exhibited increased intestinal epithelial permeability, cellular uptake, and oral bioavailability, with extended blood circulation time, increased tumor accumulation, enhanced MDM2 inhibition, and stronger responses in anti-tumor growth and metastasis effects in vitro and in vivo, without apparent host toxicity. Collectively, this newly developed nanoparticle oral delivery system provides a basis for evaluation of SP141 as a potential clinical candidate for cancer therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Sushanta Sarkar
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
50
|
Yu J, Li Z, Wang W, Zhang Y, Li D, Liu Y, Shen S, Zhang R. Anticancer 20(R)-dammarane-3β,12β,20,25-tetrol-loaded polymeric micelles: Preparation, quantification and pharmacokinetics. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1022:13-20. [PMID: 27070116 PMCID: PMC5224911 DOI: 10.1016/j.jchromb.2016.03.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/11/2022]
Abstract
Polymeric micelles are effective drug-loading sites and often used to formulate poorly water-soluble agents. In the present study, the amphiphilic copolymer methoxy-capped poly(ethyleneglycol)-block-poly(Ɛ-caprolactone) (mPEG-b-PCL) was successfully developed for the delivery of 20(R)-dammarane-3β,12β,20,25-tetrol (25-OH-PPD), a natural anticancer product from Panax notoginseng. The 25-OH-PPD-loaded micelles were characterized by morphological observation and thermodynamic stability testing. The concentrations of 25-OH-PPD was determined by HPLC-MS/MS. The optimum MRM transition of 25-OH-PPD was selected at m/z 479.4.0→461.4. The chromatographic separation was achieved on a SB-C18 column (1.8μm, 2.1×50mm) with an optimized gradient mobile phase system. The extraction recoveries of plasma and various tissue homogenates were within the range of 81.1%-110.4% and the matrix effects ranged from 81.9% to 106.7%. The intra- and inter- day precision values (RSD%) were less than 12.0%, with accuracies ranging from 85.2% to 114.2%. In addition, 25-OH-PPD was found to be stable in different biological matrix after three freeze-thaw cycles, at room temperature and at -70°C for 4 weeks. The pharmacokinetics of 25-OH-PPD-loaded micelles was evaluated in rats. The micelles appeared as transparent liquid, stable and uniform spheres with an average particle size of 35.4±4.2nm. The maximum concentration of 25-OH-PPD in micelles was much lower than in free drug preparation. However, the drug in the micelles was released steadily, with a t1/2 of 9.1±4.0h, significantly longer than in free drug (3.3±1.4h). However, the drug concentrations in tissues after the micelle administration were lower than the levels after administration of the free drugs. In summary, the micelles were characterized by long circulation and sustained release, with an ability to avoid uptake by the reticuloendothelial system, providing a promising approach to deliver intravenous 25-OH-PPD for therapy.
Collapse
Affiliation(s)
- Junxian Yu
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhe Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Yang Zhang
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Dandan Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yi Liu
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Su Shen
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|