1
|
Ghasemian M, Kazeminava F, Naseri A, Mohebzadeh S, Abbaszadeh M, Kafil HS, Ahmadian Z. Recent progress in tannic acid based approaches as a natural polyphenolic biomaterial for cancer therapy: A review. Biomed Pharmacother 2023; 166:115328. [PMID: 37591125 DOI: 10.1016/j.biopha.2023.115328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/01/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023] Open
Abstract
Significant advancements have been noticed in cancer therapy for decades. Despite this, there are still many critical challenges ahead, including multidrug resistance, drug instability, and side effects. To overcome obstacles of these problems, various types of materials in biomedical research have been explored. Chief among them, the applications of natural compounds have grown rapidly due to their superb biological activities. Natural compounds, especially polyphenolic compounds, play a positive and great role in cancer therapy. Tannic acid (TA), one of the most famous polyphenols, has attracted widespread attention in the field of cancer treatment with unique structural, physicochemical, pharmaceutical, anticancer, antiviral, antioxidant and other strong biological features. This review concentrated on the basic structure along with the important role of TA in tuning oncological signal pathways firstly, and then focused on the use of TA in chemotherapy and preparation of delivery systems including nanoparticles and hydrogels for cancer therapy. Besides, the application of TA/Fe3+ complex coating in photothermal therapy, chemodynamic therapy, combined therapy and theranostics is discussed.
Collapse
Affiliation(s)
- Motaleb Ghasemian
- Department of Medicinal Chemistry, School of Pharmacy, Lorestan University of Medical Science, Khorramabad, Iran
| | - Fahimeh Kazeminava
- Department of Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ashkan Naseri
- Department of Applied Chemistry, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Soheila Mohebzadeh
- Department of Plant Production and Genetics, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mahmoud Abbaszadeh
- Department of Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Department of Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Zainab Ahmadian
- Department of Pharmaceutics, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
2
|
Kleszcz R, Majchrzak-Celińska A, Baer-Dubowska W. Tannins in cancer prevention and therapy. Br J Pharmacol 2023. [PMID: 37614022 DOI: 10.1111/bph.16224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Tannins are a heterogenous class of polyphenolic natural products with promising cancer chemopreventive and therapeutic potential. Studies undertaken over the last 30 years have demonstrated their capacity to target many cellular pathways and molecules important in the development of cancer. Recently, new mechanisms that might be important in anti-carcinogenic activity, such as inhibition of epithelial-to-mesenchymal transition, reduction of cancer stem cell creation, and modulation of cancer cells metabolism have been described. Along with the mechanisms underlying the anti-cancer activity of tannins, this review focuses on their possible application as chemosensitizers in adjuvant therapy and countering multidrug resistance. Furthermore, characteristic physicochemical properties of some tannins, particularly tannic acid, are useful in the formation of nanovehicles for anticancer drugs or the isolation of circulating cancer cells. These new potential applications of tannins deserve further studies. Well-designed clinical trials, which are scarce, are needed to assess the therapeutic effects of tannins themselves or as adjuvants in cancer treatment.
Collapse
Affiliation(s)
- Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznań, Poland
| | | | - Wanda Baer-Dubowska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
3
|
Dechsupa N, Khamto N, Chawapun P, Siriphong S, Innuan P, Suwan A, Luangsuep T, Photilimthana N, Maita W, Thanacharttanatchaya R, Sangthong P, Meepowpan P, Udomtanakunchai C, Kantapan J. Pentagalloyl Glucose-Targeted Inhibition of P-Glycoprotein and Re-Sensitization of Multidrug-Resistant Leukemic Cells (K562/ADR) to Doxorubicin: In Silico and Functional Studies. Pharmaceuticals (Basel) 2023; 16:1192. [PMID: 37765000 PMCID: PMC10535865 DOI: 10.3390/ph16091192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/12/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
Combining phytochemicals with chemotherapeutic drugs has demonstrated the potential to surmount drug resistance. In this paper, we explore the efficacy of pentagalloyl glucose (PGG) in modulating P-gp and reversing multidrug resistance (MDR) in drug-resistant leukemic cells (K562/ADR). The cytotoxicity of PGG was evaluated using a CCK-8 assay, and cell apoptosis was assessed using flow cytometry. Western blotting was used to analyze protein expression levels. P-glycoprotein (P-gp) activity was evaluated by monitoring the kinetics of P-gp-mediated efflux of pirarubicin (THP). Finally, molecular docking, molecular dynamics simulation, and molecular mechanics with generalized Born and surface area solvation (MM-GBSA) calculation were conducted to investigate drug-protein interactions. We found that PGG selectively induced cytotoxicity in K562/ADR cells and enhanced sensitivity to doxorubicin (DOX), indicating its potential as a reversal agent. PGG reduced the expression of P-gp and its gene transcript levels. Additionally, PGG inhibited P-gp-mediated efflux and increased intracellular drug accumulation in drug-resistant cells. Molecular dynamics simulations and MM-GBSA calculation provided insights into the binding affinity of PGG to P-gp, suggesting that PGG binds tightly to both the substrate and the ATP binding sites of P-gp. These findings support the potential of PGG to target P-gp, reverse drug resistance, and enhance the efficacy of anticancer therapies.
Collapse
Affiliation(s)
- Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (N.D.); (P.I.); (A.S.)
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Nopawit Khamto
- Faculty of Science, Department of Chemistry, Chiang Mai University, Chiang Mai 50200, Thailand (P.C.); (S.S.); (P.S.); (P.M.)
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornthip Chawapun
- Faculty of Science, Department of Chemistry, Chiang Mai University, Chiang Mai 50200, Thailand (P.C.); (S.S.); (P.S.); (P.M.)
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
- Interdisciplinary Program in Biotechnology, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sadanon Siriphong
- Faculty of Science, Department of Chemistry, Chiang Mai University, Chiang Mai 50200, Thailand (P.C.); (S.S.); (P.S.); (P.M.)
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
- Interdisciplinary Program in Biotechnology, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phattarawadee Innuan
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (N.D.); (P.I.); (A.S.)
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Authaphinya Suwan
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (N.D.); (P.I.); (A.S.)
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Thitiworada Luangsuep
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Nichakorn Photilimthana
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Witchayaporn Maita
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Rossarin Thanacharttanatchaya
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Padchanee Sangthong
- Faculty of Science, Department of Chemistry, Chiang Mai University, Chiang Mai 50200, Thailand (P.C.); (S.S.); (P.S.); (P.M.)
| | - Puttinan Meepowpan
- Faculty of Science, Department of Chemistry, Chiang Mai University, Chiang Mai 50200, Thailand (P.C.); (S.S.); (P.S.); (P.M.)
- Center of Excellence in Materials Science and Technology, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chatchanok Udomtanakunchai
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (N.D.); (P.I.); (A.S.)
- Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand; (T.L.); (N.P.); (W.M.); (R.T.); (C.U.)
| |
Collapse
|
4
|
Jing W, Xiaolan C, Yu C, Feng Q, Haifeng Y. Pharmacological effects and mechanisms of tannic acid. Biomed Pharmacother 2022; 154:113561. [PMID: 36029537 DOI: 10.1016/j.biopha.2022.113561] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022] Open
Abstract
In recent years, increasing attention has been paid to the pharmacological efficacy of tannins. Tannic acid (TA), the simplest hydrolysable tannin that has been approved by the FDA as a safe food additive, is one of the most important components of these traditional medicines. Studies have shown that TA displays a wide range of pharmacological activities, such as anti-inflammatory, neuroprotective, antitumor, cardioprotective, and anti-pathogenic effects. Here, we summarize the known pharmacological effects and associated mechanisms of TA. We focus on the effect and mechanism of TA in various animal models of inflammatory disease and organ, brain, and cardiovascular injury. Moreover, we discuss the possible molecular targets and signaling pathways of TA, in addition to the pharmacological effects of TA-based nanoparticles and TA in combination with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Wang Jing
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China.
| | - Chen Xiaolan
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| | - Chen Yu
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| | - Qin Feng
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, PR China
| | - Yang Haifeng
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| |
Collapse
|
5
|
Pacheco PA, Louvandini H, Giglioti R, Wedy BCR, Ribeiro JC, Verissimo CJ, Ferreira JFDS, Amarante AFT, Katiki LM. Phytochemicals modulation of P-Glycoprotein and its gene expression in an ivermectin resistant Haemonchus contortus isolate in vitro. Vet Parasitol 2022; 305:109713. [DOI: 10.1016/j.vetpar.2022.109713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/25/2022] [Accepted: 04/30/2022] [Indexed: 10/18/2022]
|
6
|
Phytochemicals in Malignant Pleural Mesothelioma Treatment-Review on the Current Trends of Therapies. Int J Mol Sci 2021; 22:ijms22158279. [PMID: 34361048 PMCID: PMC8348618 DOI: 10.3390/ijms22158279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare but highly aggressive tumor of pleura arising in response to asbestos fibers exposure. MPM is frequently diagnosed in the advanced stage of the disease and causes poor prognostic outcomes. From the clinical perspective, MPM is resistant to conventional treatment, thus challenging the therapeutic options. There is still demand for improvement and sensitization of MPM cells to therapy in light of intensive clinical studies on chemotherapeutic drugs, including immuno-modulatory and targeted therapies. One way is looking for natural sources, whole plants, and extracts whose ingredients, especially polyphenols, have potential anticancer properties. This comprehensive review summarizes the current studies on natural compounds and plant extracts in developing new treatment strategies for MPM.
Collapse
|
7
|
Zhang Q, Feng Z, Wang H, Su C, Lu Z, Yu J, Dushkin AV, Su W. Preparation of camptothecin micelles self-assembled from disodium glycyrrhizin and tannic acid with enhanced antitumor activity. Eur J Pharm Biopharm 2021; 164:75-85. [PMID: 33878433 DOI: 10.1016/j.ejpb.2021.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/30/2021] [Accepted: 04/11/2021] [Indexed: 10/21/2022]
Abstract
Natural compounds as carriers for hydrophobic drugs have been increasingly used in drug delivery systems. In this study, disodium glycyrrhizin (Na2GA), tannic acid (TA) and camptothecin (CPT) were firstly used to prepare the camptothecin solid dispersion (CPT SD). When dissolved in a solution medium, Na2GA self-assembled to form micelles and CPT was encapsulated in micelles, meanwhile, TA connected with Na2GA through hydrogen bonds to form a contract shell. The average diameter of the CPT-loaded micelles is 80 nm with the critical micellar concentration of 0.303 mg/mL, the zeta potential of -33 mV, the PDI of 0.25 and drug loading 6.22%. In vitro experiments confirmed that the drug-loaded micelles exhibited excellent stability and permeability in the intestinal environment. Furthermore, the formulation showed excellent anti-tumor activity in vitro and in vivo. These findings imply that this nano-micelles provide a more potential and efficacious oral drug formulation for chemotherapy.
Collapse
Affiliation(s)
- Qihong Zhang
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Zongmiao Feng
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Hui Wang
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Chen Su
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Zhaohui Lu
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Jingbo Yu
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Alexandr V Dushkin
- Institute of Solid State Chemistry and Mechanochemistry, Novosibirsk, Russia
| | - Weike Su
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, PR China; Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China.
| |
Collapse
|
8
|
Reker D, Shi Y, Kirtane AR, Hess K, Zhong GJ, Crane E, Lin CH, Langer R, Traverso G. Machine Learning Uncovers Food- and Excipient-Drug Interactions. Cell Rep 2021; 30:3710-3716.e4. [PMID: 32187543 PMCID: PMC7179333 DOI: 10.1016/j.celrep.2020.02.094] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 01/06/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
Inactive ingredients and generally recognized as safe compounds are regarded by the US Food and Drug Administration (FDA) as benign for human consumption within specified dose ranges, but a growing body of research has revealed that many inactive ingredients might have unknown biological effects at these concentrations and might alter treatment outcomes. To speed up such discoveries, we apply state-of-the-art machine learning to delineate currently unknown biological effects of inactive ingredients—focusing on P-glycoprotein (P-gp) and uridine diphosphate-glucuronosyltransferase-2B7 (UGT2B7), two proteins that impact the pharmacokinetics of approximately 20% of FDA-approved drugs. Our platform identifies vitamin A palmitate and abietic acid as inhibitors of P-gp and UGT2B7, respectively; in silico, in vitro, ex vivo, and in vivo validations support these interactions. Our predictive framework can elucidate biological effects of commonly consumed chemical matter with implications on food-and excipient-drug interactions and functional drug formulation development. Reker et al. use machine learning to identify biological activities of food and drug additives. Validation confirms vitamin A palmitate as an inhibitor of P-glycoprotein transport and abietic acid as an inhibitor of UGT2b7 metabolism. Such associations have important implications as food-or excipient-drug interactions.
Collapse
Affiliation(s)
- Daniel Reker
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; MIT-IBM Watson AI Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yunhua Shi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ameya R Kirtane
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kaitlyn Hess
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Grace J Zhong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Evan Crane
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chih-Hsin Lin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; MIT-IBM Watson AI Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Traverso
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; MIT-IBM Watson AI Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
9
|
A. Youness R, Kamel R, A. Elkasabgy N, Shao P, A. Farag M. Recent Advances in Tannic Acid (Gallotannin) Anticancer Activities and Drug Delivery Systems for Efficacy Improvement; A Comprehensive Review. Molecules 2021; 26:1486. [PMID: 33803294 PMCID: PMC7967207 DOI: 10.3390/molecules26051486] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Tannic acid is a chief gallo-tannin belonging to the hydrolysable tannins extracted from gall nuts and other plant sources. A myriad of pharmaceutical and biological applications in the medical field has been well recognized to tannic acid. Among these effects, potential anticancer activities against several solid malignancies such as liver, breast, lung, pancreatic, colorectal and ovarian cancers have been reported. Tannic acid was found to play a maestro-role in tuning several oncological signaling pathways including JAK/STAT, RAS/RAF/mTOR, TGF-β1/TGF-β1R axis, VEGF/VEGFR and CXCL12/CXCR4 axes. The combinational beneficial effects of tannic acid with other conventional chemotherapeutic drugs have been clearly demonstrated in literature such as a synergistic anticancer effect and enhancement of the chemo-sensitivity in several resistant cases. Yet, clinical applications of tannic acid have been limited owing to its poor lipid solubility, low bioavailability, off-taste, and short half-life. To overcome such obstacles, novel drug delivery systems have been employed to deliver tannic acid with the aim of improving its applications and/or efficacy against cancer cells. Among these drug delivery systems are several types of organic and metallic nanoparticles. In this review, the authors focus on the molecular mechanisms of tannic acid in tuning several neoplastic diseases as well as novel drug delivery systems that can be used for its clinical applications with an attempt to provide a systemic reference to promote the development of tannic acid as a cheap drug and/or drug delivery system in cancer management.
Collapse
Affiliation(s)
- Rana A. Youness
- The Molecular Genetics Research Team, Department of Pharmaceutical Biology, Faculty of Pharmacy andBiotechnology, German University in Cairo, Cairo 12622, Egypt;
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre, Cairo 12622, Egypt;
| | - Nermeen A. Elkasabgy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt;
| | - Ping Shao
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China;
| | - Mohamed A. Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
- Chemistry Department, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
10
|
Ding P, Wang Z, Wu Z, Hu M, Zhu W, Sun N, Pei R. Tannic Acid (TA)-Functionalized Magnetic Nanoparticles for EpCAM-Independent Circulating Tumor Cell (CTC) Isolation from Patients with Different Cancers. ACS APPLIED MATERIALS & INTERFACES 2021; 13:3694-3700. [PMID: 33442969 DOI: 10.1021/acsami.0c20916] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The majority of current methods of isolating circulating tumor cells (CTCs) rely on a biomarker. However, the isolation efficiency may be compromised due to the heterogeneity of CTCs. In this work, a simple and broad-spectrum method is established to efficiently isolate the heterogeneous CTCs from patient blood samples using tannic acid (TA)-functionalized magnetic nanoparticles (MNPs). The TA-functionalized MNPs (MNPs-TA) inhibit the nonspecific adhesion of peripheral blood mononuclear cell (PBMC) and enhance cancer cell capture, resulting from the unique interaction between TA and glycocalyx on cancer cells. The MNPs-TA was demonstrated to effectively capture seven kinds of cancer cells (HeLa, PC-3, T24, MAD-MB-231, MCF-7, HT1080, A549) from artificial samples (62.3-93.7%). Moreover, this epithelial cell adhesion molecule (EpCAM)-independent CTC isolation method was also tested using clinical blood samples from patients with different cancers (21 patients), which may provide a universal tool to detect CTCs in the clinic.
Collapse
Affiliation(s)
- Pi Ding
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhili Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zeen Wu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Mingchao Hu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
- The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Weipei Zhu
- The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Na Sun
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
11
|
Wang Y, Pi C, Feng X, Hou Y, Zhao L, Wei Y. The Influence of Nanoparticle Properties on Oral Bioavailability of Drugs. Int J Nanomedicine 2020; 15:6295-6310. [PMID: 32943863 PMCID: PMC7455773 DOI: 10.2147/ijn.s257269] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022] Open
Abstract
Oral administration has been the most common therapeutic regimen in various diseases because of its high safety, convenience, lower costs, and high compliance of patients. However, susceptible in hostile gastrointestinal (GI) environment, many drugs show poor permeability across GI tract mucus and intestinal epithelium with poor oral absorption and limited therapeutic efficacy. In recent years, nanoparticulate drug delivery systems (NDDS) have become a hot research spot because of their unique advantages including protecting drug from premature degrading and interacting with the physiological environment, increasing intracellular penetration, and enhancing drug absorption. However, a slight change in physicochemistry of nanoparticles can significantly impact their interaction with biological pathways and alter the oral bioavailability of drugs. Hence, this review focuses on the factors affecting oral bioavailability from two aspects. On the one hand, the factors are the biochemical and physiological barriers in oral drugs delivery. On the other hand, the factors are the nanoparticle properties including size, surface properties, and shape of nanoparticles.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Chao Pi
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Xianhu Feng
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yi Hou
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Ling Zhao
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Yumeng Wei
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
12
|
do Nascimento SB, de Lima Nascimento M, de Araújo LL, de Oliveira FM, do Carmo Vieira M, Duarte-Almeida JM, Siqueira JM, da Costa César I, Derendorf H, de Castro WV. Evaluation of the Effects of Maytenus ilicifolia on the Activities of Cytochrome P450 3A and P-glycoprotein. Curr Drug Metab 2020; 21:281-290. [DOI: 10.2174/1389200221666200512112718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/16/2020] [Accepted: 03/16/2020] [Indexed: 11/22/2022]
Abstract
Background:
Maytenus ilicifolia is a Brazilian popular medicine commonly used to treat ulcer and
gastritis. Despite the absence of toxicity regarding its consumption, possible interactions when co-administrated with
conventional drugs, are unknown.
Objective:
This study aimed to evaluate the effects of M. ilicifolia extracts on Cytochrome P450 3A (CYP3A) and
P-glycoprotein (P-gp) activities.
Method:
The extracts were obtained by infusion (MI) or turbo-extraction using hydro-acetonic solvent (MT70). The
content of polyphenols in each extract was determined. To assess the modulation of M. ilicifolia on P-gp activity, the
uptake of fexofenadine (FEX) by Caco-2 cells was investigated in the absence or presence of MI or MT70. The effect
on CYP3A activity was evaluated by the co-administration of midazolam (MDZ) with each extract in male Wistar
rats. The pharmacokinetic parameters of the drug were determined and compared with those from the control group.
The content of total phenolic compounds, tannins, and flavonoids on MT70 extract was about double of that found in
MI.
Results:
In the presence of the extracts, the uptake of the P-gp marker (FEX) by Caco-2 cells increased from
1.7 ± 0.4 ng.mg-1 protein (control) to 3.5 ± 0.2 ng.mg-1 protein (MI) and 4.4 ± 0.5 ng.mg-1 protein (MT70),
respectively. When orally co-administrated with MDZ (substrate of CYP3A), the extracts augmented the AUC(0-∞)
(Control: 911.7 ± 215.7 ng.h.mL-1; MI: 1947 ± 554.3 ng.h.mL-1; MT70: 2219.0 ± 506.3 ng.h.mL-1) and the
Cmax (Control: 407.7 ± 90.4 ng.mL-1; MI: 1770.5 ± 764.5 ng.mL-1; MT70: 1987.2 ± 544.9 ng.mL-1) of the drug in rats
indicating a 50% reduction of the oral Cl. No effect was observed when midazolam was given intravenously.
Conclusion:
The results suggest that M. ilicifolia can inhibit the intestinal metabolism and transport of drugs
mediated by CYP3A and P-gp, respectively, however, the involvement of other transporters and the clinical
relevance of such interaction still need to be clarified.
Collapse
Affiliation(s)
- Sara Batista do Nascimento
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| | - Mariana de Lima Nascimento
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| | - Laís Lobato de Araújo
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| | - Flávio Martins de Oliveira
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| | - Maria do Carmo Vieira
- Federal University of Grande Dourados R. Joao Rosa Goes, 1761-Vila Progresso, Dourados-MS, CEP: 79825-070, Brazil
| | - Joaquim Maurício Duarte-Almeida
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| | - João Máximo Siqueira
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| | - Isabela da Costa César
- Federal University of Minas Gerais, Av. Presidente Antonio Carlos, 667, Campus Pampulha, Belo Horizonte-MG, CEP: 31270-901, Brazil
| | - Hartmut Derendorf
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32611, United States
| | - Whocely Victor de Castro
- Federal University of Sao Joao del-Rei, Av. Sebastiao Goncalves Coelho, 400, Campus Centro-Oeste, Chanadour, Divinopolis-MG, CEP: 35501-296, Brazil
| |
Collapse
|
13
|
Huijghebaert S, De Bruyne P, Allegaert K, Vande Velde S, De Bruyne R, Van Biervliet S, Van Winckel M. Medical devices that look like medicines: safety and regulatory concerns for children in Europe. Arch Dis Child 2020; 105:147-154. [PMID: 31533915 DOI: 10.1136/archdischild-2018-316391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 07/28/2019] [Accepted: 08/23/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Medical devices (MedDevs) and medicines are assessed (and monitored) differently before and after launch. There are products for repeated oral ingestion that are marketed in the European Union as MedDevs. OBJECTIVES AND METHODS To illustrate the consequences of these differences in assessment, we compared the leaflet information of three MedDevs with the standards for medicines and with published evidence at launch. As examples, gelatin tannate (GT), its combination with tyndalised probiotics (TP) (GTTP) for diarrhoea and a gel containing hyaluronic acid (HA)/chondroitin sulfate (CS)/poloxamer (Pol407) (HACSPol) for gastro-oesophageal reflux disease were examined. RESULTS Applying standards for medicines, product composition is insufficiently defined in the MedDev leaflet (eg, plant origin, polymerisation grade, dose and ratio of the relevant constituents). As no age limit is mentioned in the leaflets, all 3 products allow use in children from birth onwards, although published clinical documentation in children was poor (GT) or lacking (GTTP and HACSPol). MedDev leaflets do not mention adverse events (AEs), while literature search suggests safety concerns such as tannic acid (TA) cytotoxicity, potentially more diarrhoea/AEs with TP, use of doses higher than established safe (TA and HA) and lack of chronic toxicity studies for oral Pol407. None refers to interactions with medicines, although some ingredients may affect medicine absorption. CONCLUSION Although these MedDevs require repeated oral intake as do medicines, their assessment and monitoring differ significantly from the standards for medicines. Compared with medicines, MedDevs for repeated oral use are poorly labelled and rely on very limited clinical information at market release.
Collapse
Affiliation(s)
| | - Pauline De Bruyne
- Department of Paediatric Gastroenteroloy, Ghent University Hospital - Ghent University, Gent, Belgium.,Department of Paediatric Gastroenterology, Erasmus MC, Rotterdam, The Netherlands
| | - Karel Allegaert
- Department of Paediatrics, Division of Neonatology, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Saskia Vande Velde
- Department of Paediatric Gastroenteroloy, Ghent University Hospital - Ghent University, Gent, Belgium
| | - Ruth De Bruyne
- Department of Paediatric Gastroenteroloy, Ghent University Hospital - Ghent University, Gent, Belgium
| | - Stephanie Van Biervliet
- Department of Paediatric Gastroenteroloy, Ghent University Hospital - Ghent University, Gent, Belgium
| | - Myriam Van Winckel
- Department of Paediatric Gastroenteroloy, Ghent University Hospital - Ghent University, Gent, Belgium
| |
Collapse
|
14
|
Li HQ, Xu JY, Fan XH, Wu SS. Optimization of the traditional processing method for precision detoxification of CaoWu through biomimetic linking kinetics and human toxicokinetics of aconitine as toxic target marker. JOURNAL OF ETHNOPHARMACOLOGY 2019; 242:112053. [PMID: 31271819 DOI: 10.1016/j.jep.2019.112053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/08/2019] [Accepted: 06/30/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE CaoWu (Aconiti Kusnezoffii Radix), well known for its high toxicity leading to fatal ventricular arrhythmias, is detoxified by HeZi (Terminalia Chebula Retz) decoction to prepare ZhiCaoWu (Aconiti Kusnezoffii Radix Preparata) as one part of ingredients of NaRu-3 pill which is used for the treatment of rheumatoid arthritis (RA). Aconitine (AC) is a highly toxic alkaloid of CaoWu and it is used as toxic target marker for the quality control (QC) of ZhiCaoWu. In the traditional processing method, the vanish of astringent or spicy feeling in tongue is the important detoxification indicator of ZhiCaoWu. However, how CaoWu is detoxified to ZhiCaoWu and whether the appropriate content of AC in ZhiCaoWu can be efficiently perceived after the empirical detoxification still lack factual basis. AIM OF THE STUDY The present study aimed to optimize the traditional processing method for precision detoxification of CaoWu through biomimetic linking kinetics and human toxicokinetics (TK) of AC, with a view of providing insights into the changes of toxic target marker. MATERIALS AND METHODS CaoWu medicinal slices (Mes) and coarse powder (Cop) were processed by blank HeZi decoction through the soaking method for 7 days. High-performance liquid chromatography (HPLC) was used for the analysis of the samples. The acidity of blank HeZi decoction and HeZi processing decoction was directly determined by pH meter. The non-compartment analysis (NCA) was used to have an intuitive appreciation for AC and pH changes in HeZi processing decoction while the compartment model method was used to build the biomimetic linking kinetics model with the covariate. The inter-species scaling of animal TK parameters was conducted to predict human AC TK profiles. The possible uptake ways of AC (rapid-release or extended-release) for humans were attempted to assess the poisoning risk of AC in NaRu-3 pill. Based on the target content of AC in ZhiCaoWu, the biomimetic linking kinetics model was explored to optimize the traditional processing detoxification method of CaoWu. The assays of determining inflammatory cytokines in lipopolysaccharides (LPS)-induced RAW264.7 cells were performed to investigate the inflammatory modulation effects of AC in vitro. RESULTS ZhiCaoWu was prepared by eliminating redundant AC in CaoWu through the repeatable replacement of HeZi processing decoction in which its acidity (pH) was affected. AC-pH changes in HeZi processing decoction were adequately depicted by a biomimetic linking kinetics model whose predictive power was determined by comparing the predictions of AC in ZhiCaoWu with the reported data. Rapid-release AC at the converted dose of 111.1 and 417.6 μg (0.011 and 0.042% of AC in NaRu-3 pill) reached maximum blood concentrations of 26.1 and 98.1 ng/mL at 0.3 h, in comparison with minimum human lethal concentration (100 ng/mL). Achieving the target content of AC (0.04%) in ZhiCaoWu or AC (0.011%) in NaRu-3 pill to precisely control the poisoning risk, the potential optimized protocols were that the processing time at 0.2-0.8% of AC in CaoWu was 2.0-4.4 days for Cop and 2.7-6.2 days for Mes. Correspondingly, pH values in HeZi processing decoction were 3.95 and 3.77 for Cop and Mes, respectively. Meanwhile, Lipopolysaccharides (LPS)-induced RAW264.7 cells were exposed to 0, 20, and 200 μM of AC for 12 h and AC at 20 μM enhanced the levels of IL-6, IL-10 and TNF-α. CONCLUSIONS Thus, for the first time, a biomimetic linking kinetics model was built to optimize the traditional detoxification method. Moreover, pH changes could be developed as surrogate endpoint for guiding the processing detoxification of CaoWu. Notably, setting the content limit of AC (0.011%) was very rational to control the poisoning risk of NaRu-3 pill. In addition, it was possible that there existed the more complex mechanisms of AC for inflammatory modulation in vitro.
Collapse
Affiliation(s)
- Han Qing Li
- State Clinical Trial Institution of New Drugs, International Mongolian Hospital of Inner Mongolia, Hohhot, 010065, China; Mongolian Medicine Laboratory, Mongolian Medicine Institute of Inner Mongolia, Hohhot, 010065, China.
| | - Jia Yin Xu
- Mongolian Pharmaceutical Preparation Center, International Mongolian Hospital of Inner Mongolia, Hohhot, 010065, China
| | - Xiao Hong Fan
- State Clinical Trial Institution of New Drugs, International Mongolian Hospital of Inner Mongolia, Hohhot, 010065, China
| | - Shan Shan Wu
- State Clinical Trial Institution of New Drugs, International Mongolian Hospital of Inner Mongolia, Hohhot, 010065, China
| |
Collapse
|
15
|
Chowdhury P, Nagesh PKB, Hatami E, Wagh S, Dan N, Tripathi MK, Khan S, Hafeez BB, Meibohm B, Chauhan SC, Jaggi M, Yallapu MM. Tannic acid-inspired paclitaxel nanoparticles for enhanced anticancer effects in breast cancer cells. J Colloid Interface Sci 2019; 535:133-148. [PMID: 30292104 PMCID: PMC6992213 DOI: 10.1016/j.jcis.2018.09.072] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022]
Abstract
Paclitaxel (PTX) is a gold standard chemotherapeutic agent for breast, ovarian, pancreatic and non-small cell lung carcinoma. However, in clinical use PTX can have adverse side effects or inadequate pharmacodynamic parameters, limiting its use. Nanotechnology is often employed to reduce the therapeutic dosage required for effective therapy, while also minimizing the systemic side effects of chemotherapy drugs. However, there is no nanoformulation of paclitaxel with chemosensitization motifs built in. With this objective, we screened eleven pharmaceutical excipients to develop an alternative paclitaxel nanoformulation using a self-assembly method. Based on the screening results, we observed tannic acid possesses unique properties to produce a paclitaxel nanoparticle formulation, i.e., tannic acid-paclitaxel nanoparticles. This stable TAP nanoformulation, referred to as TAP nanoparticles (TAP NPs), showed a spherical shape of ~ 102 nm and negative zeta potential of ~ -8.85. The presence of PTX in TAP NPs was confirmed by Fourier Transform Infrared (FTIR) spectra, thermogravimetric analyzer (TGA), and X-ray diffraction (XRD). Encapsulation efficiency of PTX in TAP NPs was determined to be ≥96%. Intracellular drug uptake of plain drug PTX on breast cancer cells (MDA-MB-231) shows more or less constant drug levels in 2 to 6 h, suggesting drug efflux by the P-gp transporters, over TAP NPs, in which PTX uptake was more than 95.52 ± 11.01% in 6 h, as analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Various biological assays such as proliferation, clonogenic formation, invasion, and migration confirm superior anticancer effects of TAP NPs over plain PTX at all tested concentrations. P-gp expression, beta-tubulin stabilization, Western blot, and microarray analysis further confirm the improved therapeutic potential of TAP NPs. These results suggest that the TAP nanoformulation provides an important reference for developing a therapeutic nanoformulation affording pronounced, enhanced effects in breast cancer therapy.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Elham Hatami
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Santosh Wagh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nirnoy Dan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Manish K Tripathi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sheema Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bilal B Hafeez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
16
|
Le Z, Chen Y, Han H, Tian H, Zhao P, Yang C, He Z, Liu L, Leong KW, Mao HQ, Liu Z, Chen Y. Hydrogen-Bonded Tannic Acid-Based Anticancer Nanoparticle for Enhancement of Oral Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:42186-42197. [PMID: 30444601 DOI: 10.1021/acsami.8b18979] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Oral chemotherapy has been emerging as a hopeful therapeutic regimen for the treatment of various cancers because of its high safety and convenience, lower costs, and high patient compliance. Currently, nanoparticulate drug delivery systems (NDDS) exhibit many unique advantages in mediating oral drug delivery; however, many anticancer drugs that were susceptible in hostile gastrointestinal (GI) environment showed poor permeability across intestinal epithelium, and most materials used as drug carriers are nonactive excipients and displayed no therapeutically relevant function, which leads to low oral bioavailability and therapeutic efficacy of anticancer drugs (e.g., paclitaxel). Inspired by these, in this study, paclitaxel (PTX) was used as a model drug, depending on intermolecular hydrogen-bonded interactions, PTX-loaded tannic acid/poly( N-vinylpyrrolidone) nanoparticles (PTX-NP) were produced by a flash nanoprecipitation (FNP) process. The optimized PTX-NP showed an average diameter of 54 nm with a drug encapsulation efficiency of 80% and loading capacity of 14.5%. Molecular dynamics simulations were carried out to illuminate the assembling mechanism of hydrogen-bonded PTX-NP. In vitro and in vivo results confirmed that PTX-NP showed pH-dependent intestinal site-specific drug release, P-gp inhibitory function by tannic acid (TA), prolonged intestinal retention, and improved trans-epithelial transport properties. Oral administration of PTX-NP generated a high oral delivery efficiency and relative oral bioavailability of 25.6% in rats, and further displayed a significant tumor-inhibition effect in a xenograft breast tumor model. These findings confirmed that our PTX-NP might be a promising oral drug formulation for chemotherapy.
Collapse
Affiliation(s)
- Zhicheng Le
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Yantao Chen
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation, College of Chemistry and Environmental Engineering , Shenzhen University , Shenzhen 518060 , China
| | - Honghua Han
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation, College of Chemistry and Environmental Engineering , Shenzhen University , Shenzhen 518060 , China
| | - Houkuan Tian
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Pengfei Zhao
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Chengbiao Yang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Zhiyu He
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Kam W Leong
- Department of Biomedical Engineering , Columbia University , New York , New York 10027 , United States
| | - Hai-Quan Mao
- Institute for NanoBioTechnology and Department of Materials Science and Engineering , Johns Hopkins University , Baltimore , Maryland 21218 , United States
- Department of Biomedical Engineering and Translational Tissue Engineering Center , Johns Hopkins University School of Medicine , Baltimore , Maryland 21287 , United States
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, GD Research Center for Functional Biomaterials Engineering and Technology , Sun Yat-sen University , Guangzhou 510275 , China
| |
Collapse
|
17
|
Ma BL, Ma YM. Pharmacokinetic herb–drug interactions with traditional Chinese medicine: progress, causes of conflicting results and suggestions for future research. Drug Metab Rev 2016; 48:1-26. [DOI: 10.3109/03602532.2015.1124888] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
18
|
Factors Influencing Oral Bioavailability of Thai Mango Seed Kernel Extract and Its Key Phenolic Principles. Molecules 2015; 20:21254-73. [PMID: 26633325 PMCID: PMC6332320 DOI: 10.3390/molecules201219759] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/25/2022] Open
Abstract
Mango seed kernel extract (MSKE) and its key components (gallic acid, GA; methyl gallate, MG; and pentagalloyl glucopyranose, PGG) have generated interest because of their pharmacological activities. To develop the potential use of the key components in MSKE as natural therapeutic agents, their pharmacokinetic data are necessary. Therefore, this study was performed to evaluate the factors affecting their oral bioavailability as pure compounds and as components in MSKE. The in vitro chemical stability, biological stability, and absorption were evaluated in Hanks' Balanced Salt Solution, Caco-2 cell and rat fecal lysates, and the Caco-2 cell model, respectively. The in vivo oral pharmacokinetic behavior was elucidated in Sprague-Dawley rats. The key components were unstable under alkaline conditions and in Caco-2 cell lysates or rat fecal lysates. The absorptive permeability coefficient followed the order MG > GA > PGG. The in vivo results exhibited similar pharmacokinetic trends to the in vitro studies. Additionally, the co-components in MSKE may affect the pharmacokinetic behaviors of the key components in MSKE. In conclusion, chemical degradation under alkaline conditions, biological degradation by intestinal cell and colonic microflora enzymes, and low absorptive permeability could be important factors underlying the oral bioavailability of these polyphenols.
Collapse
|
19
|
Spector NL, Robertson FC, Bacus S, Blackwell K, Smith DA, Glenn K, Cartee L, Harris J, Kimbrough CL, Gittelman M, Avisar E, Beitsch P, Koch KM. Lapatinib Plasma and Tumor Concentrations and Effects on HER Receptor Phosphorylation in Tumor. PLoS One 2015; 10:e0142845. [PMID: 26571496 PMCID: PMC4646457 DOI: 10.1371/journal.pone.0142845] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/26/2015] [Indexed: 01/04/2023] Open
Abstract
PURPOSE The paradigm shift in cancer treatment from cytotoxic drugs to tumor targeted therapies poses new challenges, including optimization of dose and schedule based on a biologically effective dose, rather than the historical maximum tolerated dose. Optimal dosing is currently determined using concentrations of tyrosine kinase inhibitors in plasma as a surrogate for tumor concentrations. To examine this plasma-tumor relationship, we explored the association between lapatinib levels in tumor and plasma in mice and humans, and those effects on phosphorylation of human epidermal growth factor receptors (HER) in human tumors. EXPERIMENTAL DESIGN Mice bearing BT474 HER2+ human breast cancer xenografts were dosed once or twice daily (BID) with lapatinib. Drug concentrations were measured in blood, tumor, liver, and kidney. In a randomized phase I clinical trial, 28 treatment-naïve female patients with early stage HER2+ breast cancer received lapatinib 1000 or 1500 mg once daily (QD) or 500 mg BID before evaluating steady-state lapatinib levels in plasma and tumor. RESULTS In mice, lapatinib levels were 4-fold higher in tumor than blood with a 4-fold longer half-life. Tumor concentrations exceeded the in vitro IC90 (~ 900 nM or 500 ng/mL) for inhibition of HER2 phosphorylation throughout the 12-hour dosing interval. In patients, tumor levels were 6- and 10-fold higher with QD and BID dosing, respectively, compared to plasma trough levels. The relationship between tumor and plasma concentration was complex, indicating multiple determinants. HER receptor phosphorylation varied depending upon lapatinib tumor concentrations, suggestive of changes in the repertoire of HER homo- and heterodimers. CONCLUSION Plasma lapatinib concentrations underestimated tumor drug levels, suggesting that optimal dosing should be focused on the site of action to avoid to inappropriate dose escalation. Larger clinical trials are required to determine optimal dose and schedule to achieve tumor concentrations that maximally inhibit HER receptors. TRIAL REGISTRATION CLINICAL TRIAL REGISTRATION NCT00359190.
Collapse
Affiliation(s)
- Neil L. Spector
- Department of Medicine, Duke Cancer Center, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| | - Faith C. Robertson
- Department of Medicine, Duke Cancer Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Sarah Bacus
- Targeted Molecular Diagnostics/Quintiles, Westmont, Illinois, United States of America
| | - Kimberly Blackwell
- Department of Medicine, Duke Cancer Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Deborah A. Smith
- GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| | - Kelli Glenn
- GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| | - Leanne Cartee
- GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| | - Jennifer Harris
- Pivot Oncology Consulting, Durham, North Carolina, United States of America
| | | | - Mark Gittelman
- Breast Cancer Specialists, Allentown, Pennsylvania, United States of America
| | - Eli Avisar
- Department of Surgery, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Peter Beitsch
- Dallas Surgical Group, Dallas, Texas, United States of America
| | - Kevin M. Koch
- GlaxoSmithKline, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
20
|
Mitchell C, Syed NIH, Gurney AM, Kennedy C. A Ca²⁺-dependent chloride current and Ca²⁺ influx via Ca(v)1.2 ion channels play major roles in P2Y receptor-mediated pulmonary vasoconstriction. Br J Pharmacol 2012; 166:1503-12. [PMID: 22320222 PMCID: PMC3417463 DOI: 10.1111/j.1476-5381.2012.01892.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 12/19/2011] [Accepted: 01/31/2012] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE ATP, UTP and UDP act at smooth muscle P2X and P2Y receptors to constrict rat intrapulmonary arteries, but the underlying signalling pathways are poorly understood. Here, we determined the roles of the Ca²⁺ -dependent chloride ion current (I(Cl,Ca)), Ca(v)1.2 ion channels and Ca²⁺ influx. EXPERIMENTAL APPROACH Isometric tension was recorded from endothelium-denuded rat intrapulmonary artery rings (i.d. 200-500 µm) mounted on a wire myograph. KEY RESULTS The I(Cl,Ca) blockers, niflumic acid and 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid and the Ca(v)1.2 channel blocker, nifedipine, reduced peak amplitude of contractions evoked by UTP and UDP by ∼45-50% and in a non-additive manner. Ca²⁺-free buffer inhibited responses by ∼70%. Niflumic acid and nifedipine similarly depressed contractions to ATP, but Ca²⁺-free buffer almost abolished the response. After peaking, contractions to UTP and UDP decayed slowly by 50-70% to a sustained plateau, which was rapidly inhibited by niflumic acid and nifedipine. Contractions to ATP, however, reversed rapidly and fully. Tannic acid contracted tissues per se and potentiated nucleotide-evoked contractions. CONCLUSIONS AND IMPLICATIONS I (Cl,Ca) and Ca²⁺ influx via Ca(v)1.2 ion channels contribute substantially and equally to contractions of rat intrapulmonary arteries evoked by UTP and UDP, via P2Y receptors. ATP also activates these mechanisms via P2Y receptors, but the greater dependence on extracellular Ca²⁺ most likely reflects additional influx through the P2X1 receptor pore. The lack of a sustained response to ATP is probably due to it acting at P2 receptor subtypes that desensitize rapidly. Thus multiple signalling mechanisms contribute to pulmonary artery vasoconstriction mediated by P2 receptors.
Collapse
Affiliation(s)
- Callum Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | | | | |
Collapse
|
21
|
Wink M, Ashour ML, El-Readi MZ. Secondary Metabolites from Plants Inhibiting ABC Transporters and Reversing Resistance of Cancer Cells and Microbes to Cytotoxic and Antimicrobial Agents. Front Microbiol 2012; 3:130. [PMID: 22536197 PMCID: PMC3332394 DOI: 10.3389/fmicb.2012.00130] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 03/19/2012] [Indexed: 12/14/2022] Open
Abstract
Fungal, bacterial, and cancer cells can develop resistance against antifungal, antibacterial, or anticancer agents. Mechanisms of resistance are complex and often multifactorial. Mechanisms include: (1) Activation of ATP-binding cassette (ABC) transporters, such as P-gp, which pump out lipophilic compounds that have entered a cell, (2) Activation of cytochrome p450 oxidases which can oxidize lipophilic agents to make them more hydrophilic and accessible for conjugation reaction with glucuronic acid, sulfate, or amino acids, and (3) Activation of glutathione transferase, which can conjugate xenobiotics. This review summarizes the evidence that secondary metabolites (SM) of plants, such as alkaloids, phenolics, and terpenoids can interfere with ABC transporters in cancer cells, parasites, bacteria, and fungi. Among the active natural products several lipophilic terpenoids [monoterpenes, diterpenes, triterpenes (including saponins), steroids (including cardiac glycosides), and tetraterpenes] but also some alkaloids (isoquinoline, protoberberine, quinoline, indole, monoterpene indole, and steroidal alkaloids) function probably as competitive inhibitors of P-gp, multiple resistance-associated protein 1, and Breast cancer resistance protein in cancer cells, or efflux pumps in bacteria (NorA) and fungi. More polar phenolics (phenolic acids, flavonoids, catechins, chalcones, xanthones, stilbenes, anthocyanins, tannins, anthraquinones, and naphthoquinones) directly inhibit proteins forming several hydrogen and ionic bonds and thus disturbing the 3D structure of the transporters. The natural products may be interesting in medicine or agriculture as they can enhance the activity of active chemotherapeutics or pesticides or even reverse multidrug resistance, at least partially, of adapted and resistant cells. If these SM are applied in combination with a cytotoxic or antimicrobial agent, they may reverse resistance in a synergistic fashion.
Collapse
Affiliation(s)
- Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University Heidelberg, Germany
| | | | | |
Collapse
|
22
|
Ryu HG, Jeong SJ, Kwon HY, Lee HJ, Lee EO, Lee MH, Choi SH, Ahn KS, Kim SH. Penta-O-galloyl-β-D-glucose attenuates cisplatin-induced nephrotoxicity via reactive oxygen species reduction in renal epithelial cells and enhances antitumor activity in Caki-2 renal cancer cells. Toxicol In Vitro 2011; 26:206-14. [PMID: 22172427 DOI: 10.1016/j.tiv.2011.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 09/23/2011] [Accepted: 11/17/2011] [Indexed: 01/26/2023]
Abstract
Cisplatin shows limited therapeutic efficacy due to serious side effects such as nephrotoxicity and hepatotoxicity. In the present study, we demonstrate that 1,2,3,4,6-penta-O-galloyl-β-d-glucose (PGG) has protective effects against cisplatin-induced cytotoxicity and apoptosis in normal human primary renal epithelial cells (HRCs) while showing synergistic effect against cisplatin-induced cell death in human Caki-2 renal cancer cells. PGG significantly blocked cisplatin-mediated cytotoxicity and reduced cisplatin-induced sub-G1 accumulation in HRCs. Consistently, PGG reduced the number of apoptotic cell populations by TdT-mediated dUTP nick end labeling (TUNEL) and Live/Dead assays in cisplatin-treated HRCs. Furthermore, PGG suppressed PARP cleavage and caspase-3 activation, cytochrome c release, up-regulation of bax and p53 in cisplatin-treated HRCs. Moreover, PGG attenuated reactive oxygen species (ROS) production mediated by cisplatin treatment, suggesting that PGG prevented cisplatin-induced apoptosis by inhibiting ROS generation in HRCs. Notably, PGG significantly enhanced cytotoxicity and PARP cleavage in cisplatin-treated Caki-2 renal cancer cells. Combination Index (CI) revealed synergism between PGG and cisplatin in Caki-2 cells. Taken together, our findings suggest the dual effects of PGG as a protective supplement against cisplatin-induced toxicity in normal renal cells and a combination chemotherapeutic drug with cisplatin in renal cancer cells.
Collapse
Affiliation(s)
- Ho-Geon Ryu
- College of Oriental Medicine, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Li Y, Revalde JL, Reid G, Paxton JW. Interactions of dietary phytochemicals with ABC transporters: possible implications for drug disposition and multidrug resistance in cancer. Drug Metab Rev 2011; 42:590-611. [PMID: 20433315 DOI: 10.3109/03602531003758690] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Common foods, such as fruits and vegetables, contain a large variety of secondary metabolites known as phytochemicals, many of which have been associated with health benefits. However, there is a limited knowledge of the processes by which these, mainly charged, phytochemicals (and/or their metabolites) are absorbed into the body, reach their biological target, and how they are eliminated. Recent studies have indicated that some of these phytochemicals are substrates and modulators of specific members of the superfamily of ABC transporting proteins. In this review, we present the reported interactions between the different classes of phytochemicals and ABC transporters and the mechanism by which they modulate the activity of these transporters. We also discuss the implications that such interactions may have on the pharmacokinetics of xenobiotics and the possible role of phytochemicals in the reversal of multidrug resistance in cancer chemotherapy.
Collapse
Affiliation(s)
- Yan Li
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
24
|
Xiang W, Gao A, Liang H, Li C, Gao J, Wang Q, Shuang B, Zhang J, Yan Y, Wang X. Reversal of P-glycoprotein-mediated multidrug resistance in vitro by milbemycin compounds in adriamycin-resistant human breast carcinoma (MCF-7/adr) cells. Toxicol In Vitro 2010; 24:1474-81. [PMID: 20656007 DOI: 10.1016/j.tiv.2010.07.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 07/07/2010] [Accepted: 07/19/2010] [Indexed: 12/25/2022]
Abstract
The effects of milbemycin A(4) (MB A(4)), milbemycin oxime A(4) (MBO A(4)) and milbemycin beta(1) (MB beta(1)) on reversing multidrug resistance (MDR) of tumor cells were firstly conducted according to the following research, including MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assay, the accumulation of adriamycin, the accumulation and efflux of rhodamine 123 (Rh123), the regulations of MDR1 gene, and expression of P-gp. The three milbemycins (5muM) showed strong potency to increase adriamycin cytotoxicity toward adriamycin-resistant human breast carcinoma cells MCF-7/adr with reversal fold (RF) of 21.42, 19.06 and 14.89, respectively. In addition, the mechanisms of milbemycins on P-glycoprotein (P-gp)-mediated MDR demonstrated that the milbemycins significantly increased the intracellular accumulations of adriamycin and Rh123 via inhibiting P-gp transport function. Based on the analysis of the P-gp and MDR1 gene expression using flow cytometry and RT-PCR, the results revealed that milbemycin compounds, particularly MB A(4), could regulate down the expression of the P-gp and MDR1 gene. These findings suggest that the milbemycins probably represent promising agents for overcoming MDR in cancer therapy, and especially MB A(4) is better modulator with the lowest toxicity.
Collapse
Affiliation(s)
- Wensheng Xiang
- School of Life Science, Northeast Agricultural University, Xiangfang District, Harbin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gao A, Wang X, Xiang W, Liang H, Gao J, Yan Y. Reversal of P-glycoprotein-mediated multidrug resistance in vitro by doramectin and nemadectin. J Pharm Pharmacol 2010; 62:393-9. [DOI: 10.1211/jpp.62.03.0016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Objectives
Multidrug resistance (MDR) is a serious obstacle encountered in cancer treatment. This study was performed to explore the reversal of MDR by doramectin from the avermectin family and nemadectin belonging to the milbemycin family.
Methods
The MTT assay was used to evaluate the abilities of the two compounds to reverse drug resistance in adriamycin-resistant human breast carcinoma cells (MCF-7/adr). Intracellular accumulation of adriamycin was determined by HPLC. The effects of the two compounds on inhibiting P-glycoprotein (P-gp) efflux was demonstrated by accumulation of rhodamine 123 in MCF-7/adr cells. To investigate the mechanism of reversal by the two compounds, the expressions of P-gp and the MDR1 gene encoding P-gp were tested by flow cytometry and reverse-transcriptase PCR.
Key findings
Doramectin and nemadectin at the high dose of 8 μmol/l significantly increased the sensitivity of MCF-7/adr cells to adriamycin by 49.35- and 23.97-fold, respectively. They also increased the intracellular accumulation of adriamycin and rhodamine 123 in MCF-7/adr cells in a dose-dependent manner. Expression of both P-gp and MDR1 were down-regulated.
Conclusions
Doramectin and nemadectin are promising agents for overcoming MDR in cancer therapy. Doramectin was more potent in reversing MDR.
Collapse
Affiliation(s)
- Aili Gao
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Xiangjing Wang
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Wensheng Xiang
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Hongsheng Liang
- Department of Neurosurgery and Key Laboratory in Cell Transplantation in Ministry of Health of China; The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiguo Gao
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Yijun Yan
- School of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
26
|
Zhang J, Li L, Kim SH, Hagerman AE, Lü J. Anti-cancer, anti-diabetic and other pharmacologic and biological activities of penta-galloyl-glucose. Pharm Res 2009; 26:2066-80. [PMID: 19575286 PMCID: PMC2822717 DOI: 10.1007/s11095-009-9932-0] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 06/17/2009] [Indexed: 12/22/2022]
Abstract
1, 2, 3, 4, 6-penta-O-galloyl-beta-D-glucose (PGG) is a polyphenolic compound highly enriched in a number of medicinal herbals. Several in vitro and a handful of in vivo studies have shown that PGG exhibits multiple biological activities which implicate a great potential for PGG in the therapy and prevention of several major diseases including cancer and diabetes. Chemically and functionally, PGG appears to be distinct from its constituent gallic acid or tea polyphenols. For anti-cancer activity, three published in vivo preclinical cancer model studies with PGG support promising efficacy to selectively inhibit malignancy without host toxicity. Potential mechanisms include anti-angiogenesis; anti-proliferative actions through inhibition of DNA replicative synthesis, S-phase arrest, and G(1) arrest; induction of apoptosis; anti-inflammation; and anti-oxidation. Putative molecular targets include p53, Stat3, Cox-2, VEGFR1, AP-1, SP-1, Nrf-2, and MMP-9. For anti-diabetic activity, PGG and analogues appear to improve glucose uptake. However, very little is known about the absorption, pharmacokinetics, and metabolism of PGG, or its toxicity profile. The lack of a large quantity of highly pure PGG has been a bottleneck limiting in vivo validation of cancer preventive and therapeutic efficacies in clinically relevant models.
Collapse
Affiliation(s)
- Jinhui Zhang
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Li Li
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Sung-Hoon Kim
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
- Cancer Preventive Material Development Research Center and Institute, College of Oriental Medicine, Kyunghee University, Republic of Korea
| | - Ann E. Hagerman
- Department of Chemistry & Biochemistry, Miami University, Oxford OH 45056
| | - Junxuan Lü
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| |
Collapse
|