51
|
He Q, Yang J, Zhang G, Chen D, Zhang M, Pan Z, Wang Z, Su L, Zeng J, Wang B, Wang H, Chen P. Sanhuang Jiangtang tablet protects type 2 diabetes osteoporosis via AKT-GSK3β-NFATc1 signaling pathway by integrating bioinformatics analysis and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2021; 273:113946. [PMID: 33647426 DOI: 10.1016/j.jep.2021.113946] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/07/2021] [Accepted: 02/14/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanhuang Jiangtang tablet (SHJTT), has been widely used to treat type 2 diabetes mellitus (T2DM). However, the potential and mechanism of SHJTT in treating type 2 diabetes osteoporosis (T2DOP) has not been reported. AIM OF THE STUDY The aim of this work was to investigate the role and the underlying molecular mechanism of SHJTT in managing type 2 diabetes osteoporosis. MATERIALS AND METHODS The target genes of each component consisting of SHJTT were obtained by searching the ETCM database. The target genes of osteoporosis and diabetes were individually acquired by analyzing the DisGeNET and OMIM disease databases. Then the potential therapeutic genes were obtained from the intersection of the herbal medicine targets and the disease targets which were imported into the R and STRING platform for the analysis of GO terms, KEGG pathways and PPI network. The key modules of PPI network were constructed by Cytoscape software. Finally, leptin receptor deficiency (db/db) mice were confirmed as an animal model of type 2 diabetic osteoporosis (T2DOP) through phenotype assessment and the key genes of SHJTT against T2DOP were validated by quantitative real-time PCR (qRT-PCR). RESULTS A total of 786 target genes of SHJTT were obtained from ETCM. Simultaneously, a total of 3906 osteoporosis and type 2 diabetes associated targets were acquired from DisGeNET and OMIM databases. Then, 97 common targets were found by overlapping them. On the basis of the GO and KEGG enrichment analysis and PPI network, we found that the related pathway of SHJTT in type 2 diabetes osteoporosis was AKT-GSK3β-NFATc1 pathway which is tightly associated with osteoclast differentiation. The expression of key genes including Akt1, Mapk3, Gsk3β, Mmp9, Nfkb1 were significantly down-regulated by SHJTT in T2DOP mice (p < 0.05). CONCLUSIONS SHJTT had a protective effect on T2DOP via regulating AKT-GSK3β-NFATc1 signaling pathway. This study might provide a theoretical basis for the application of SHJTT for the treatment of type 2 diabetic osteoporosis.
Collapse
Affiliation(s)
- Qi He
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Junzheng Yang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Gangyu Zhang
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Delong Chen
- Department of Orthopaedic Surgery, Clifford Hospital, Jinan University, Guangzhou, 510006, PR China
| | - Meng Zhang
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, 450003, PR China
| | - Zhaofeng Pan
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Zihao Wang
- Queen's University Belfast, University Road, Belfast, Northen Ireland, BT7 1NN, United Kingdom
| | - Lijun Su
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Jiaxu Zeng
- 1(st) School of Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Baohua Wang
- Department of Endocrinology, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China.
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzho, 510405, PR China.
| | - Peng Chen
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzho, 510405, PR China.
| |
Collapse
|
52
|
Levit SL, Tang C. Polymeric Nanoparticle Delivery of Combination Therapy with Synergistic Effects in Ovarian Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1048. [PMID: 33923947 PMCID: PMC8072532 DOI: 10.3390/nano11041048] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
Treatment of ovarian cancer is challenging due to late stage diagnosis, acquired drug resistance mechanisms, and systemic toxicity of chemotherapeutic agents. Combination chemotherapy has the potential to enhance treatment efficacy by activation of multiple downstream pathways to overcome drug resistance and reducing required dosages. Sequence of delivery and the dosing schedule can further enhance treatment efficacy. Formulation of drug combinations into nanoparticles can further enhance treatment efficacy. Due to their versatility, polymer-based nanoparticles are an especially promising tool for clinical translation of combination therapies with tunable dosing schedules. We review polymer nanoparticle (e.g., micelles, dendrimers, and lipid nanoparticles) carriers of drug combinations formulated to treat ovarian cancer. In particular, the focus on this review is combinations of platinum and taxane agents (commonly used first line treatments for ovarian cancer) combined with other small molecule therapeutic agents. In vitro and in vivo drug potency are discussed with a focus on quantifiable synergistic effects. The effect of drug sequence and dosing schedule is examined. Computational approaches as a tool to predict synergistic drug combinations and dosing schedules as a tool for future nanoparticle design are also briefly discussed.
Collapse
Affiliation(s)
- Shani L Levit
- Chemical and Life Science Engineering Department, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Christina Tang
- Chemical and Life Science Engineering Department, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
53
|
Gui G, Fan Z, Ning Y, Yuan C, Zhang B, Xu Q. Optimization, Characterization and in vivo Evaluation of Paclitaxel-Loaded Folate-Conjugated Superparamagnetic Iron Oxide Nanoparticles. Int J Nanomedicine 2021; 16:2283-2295. [PMID: 33776433 PMCID: PMC7992116 DOI: 10.2147/ijn.s287434] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/19/2021] [Indexed: 12/28/2022] Open
Abstract
Background Paclitaxel (PTX) has interesting anticancer activity. However, it is insoluble in water, which seriously hinders its use in clinical. Superparamagnetic iron oxide nanoparticles (SPIONs) are used as an ideal drug delivery system. Therefore, we proposed a folic acid (FA) targeting drug-loaded SPIONs to reduce its adverse reaction. Methods To improve the hydrophilicity of PTX, the structure of PTX was modified by succinic anhydride to obtain 2ʹ-succinate paclitaxel (SPTX). FA conjugated Polyethylene glycol (PEG)/polyethyleneimine (PEI)-SPIONs SPTX-loaded nanoparticles (SPTX@FA@PEG/PEI-SPIONs) were prepared by solvent volatilization and hydrogen bond adsorption, and the nano-formulation was optimized by response surface methodology (RSM). The characteristics, antitumor effect in vitro, pharmacokinetics, and biodistribution of SPTX@FA@PEG/PEI-SPIONs were evaluated. Results SPTX was successfully loaded on the surface of FA@PEG/PEI-SPIONs. The formation of SPTX@FA@PEG/PEI-SPIONs was exhibited water-dispersive monodispersity with high stability by RSM, and dynamic light scattering (DLS) was 178.1±3.12 nm, particle size observed in the transmission electron microscope (TEM) was 13.01±1.10 nm, and the encapsulation efficiency (EE) and loading efficiency (LE) were 81.1±1.66% and 14.8±1.46%, respectively. It enhanced the stability in normal physiological condition, accelerated drug release at tumorous pH, and preferentially prolonged the circulation time. In vitro, the SPTX@FA@PEG/PEI-SPIONs significantly targeted to folate receptor (FR) positive cancers cell (HNE-1) via the receptor-ligand mediated pathway, resulting in effective cytotoxic activity. Pharmacokinetic results demonstrated that SPTX@FA@PEG/PEI-SPIONs (t1/2=3.41 h) had longer than free SPTX or PTX (t1/2=1.67 h) in rats in vivo. Tissue distribution studies showed that SPTX@FA@PEG/PEI-SPIONs were present at high levels in the liver and help in targeting the folate receptors present on the kidneys. Conclusion These results suggest that SPTX@FA@PEG/PEI-SPIONs offer a highly promising approach to control drug release, improve drug pharmacokinetics and actively target the nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Gang Gui
- College of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Ziqi Fan
- College of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Yihuan Ning
- College of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| | - Cancan Yuan
- Department of Pharmacy, Luohe City People's Hospital, Luohe, 462000, People's Republic of China
| | - Baolin Zhang
- College of Materials Science and Engineering, Guilin University of Technology, Guilin, 541004, People's Republic of China
| | - Qin Xu
- College of Pharmacy, Guilin Medical University, Guilin, 541004, People's Republic of China
| |
Collapse
|
54
|
Guo Q, Wu Y, Guo X, Cao L, Xu F, Zhao H, Zhu J, Wen H, Ju X, Wu X. The RNA-Binding Protein CELF2 Inhibits Ovarian Cancer Progression by Stabilizing FAM198B. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:169-184. [PMID: 33335801 PMCID: PMC7734233 DOI: 10.1016/j.omtn.2020.10.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/11/2020] [Indexed: 02/06/2023]
Abstract
An increasing number of studies have clarified the functional roles of RNA-binding proteins (RBPs) in driving post-transcriptional mechanisms of cancer progression. In this study, we integrated data from the RBP database and Gene Expression Omnibus (GEO) data with RNA sequencing (RNA-seq) data from 10 ovarian cancer tissues and 8 normal ovarian tissues and identified an RBP, CUGBP- and ETR-3-like family 2 (CELF2). We found that CELF2 expression was downregulated in ovarian cancer and positively correlated with the overall survival (OS) and progression-free survival (PFS) of patients with ovarian cancer. Altered CELF2 expression led to changes in the proliferation, migration, and invasion of ovarian cancer cells in vitro and in vivo. CELF2 expression increased the stability of its target, FAM198B, by binding to AU/U-rich elements (AREs) in the 3' untranslated region (3' UTR). FAM198B knockdown restored the CELF2-mediated suppression of proliferation and migration. We also found that CELF2/FAM198B may repress ovarian cancer progression by inhibiting the mitogen-activated protein kinase/extracellular-regulated protein kinase (MAPK/ERK) signaling pathway. Finally, a curcumin-induced increase in CELF2 expression resulted in increased ovarian cancer cell sensitivity to cisplatin. Our study elucidated a novel mechanism by which the CELF2/FAM198B axis regulates proliferation and metastasis in ovarian cancer, providing novel, potential therapeutic targets for ovarian cancer.
Collapse
Affiliation(s)
- Qinhao Guo
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Xueqi Guo
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Lijie Cao
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Fei Xu
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Haiyun Zhao
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Jun Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Hao Wen
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Xingzhu Ju
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Xiaohua Wu
- Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong-An Road, Shanghai 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| |
Collapse
|
55
|
Nanoparticles in precision medicine for ovarian cancer: From chemotherapy to immunotherapy. Int J Pharm 2020; 591:119986. [DOI: 10.1016/j.ijpharm.2020.119986] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/23/2020] [Accepted: 10/11/2020] [Indexed: 12/24/2022]
|
56
|
Polychemotherapy with Curcumin and Doxorubicin via Biological Nanoplatforms: Enhancing Antitumor Activity. Pharmaceutics 2020; 12:pharmaceutics12111084. [PMID: 33187385 PMCID: PMC7697177 DOI: 10.3390/pharmaceutics12111084] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/31/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is a well-known chemotherapeutic agent extensively applied in the field of cancer therapy. However, similar to other chemotherapeutic agents such as cisplatin, paclitaxel, docetaxel, etoposide and oxaliplatin, cancer cells are able to obtain chemoresistance that limits DOX efficacy. In respect to dose-dependent side effect of DOX, enhancing its dosage is not recommended for effective cancer chemotherapy. Therefore, different strategies have been considered for reversing DOX resistance and diminishing its side effects. Phytochemical are potential candidates in this case due to their great pharmacological activities. Curcumin is a potential antitumor phytochemical isolated from Curcuma longa with capacity of suppressing cancer metastasis and proliferation and affecting molecular pathways. Experiments have demonstrated the potential of curcumin for inhibiting chemoresistance by downregulating oncogene pathways such as MMP-2, TGF-β, EMT, PI3K/Akt, NF-κB and AP-1. Furthermore, coadministration of curcumin and DOX potentiates apoptosis induction in cancer cells. In light of this, nanoplatforms have been employed for codelivery of curcumin and DOX. This results in promoting the bioavailability and internalization of the aforementioned active compounds in cancer cells and, consequently, enhancing their antitumor activity. Noteworthy, curcumin has been applied for reducing adverse effects of DOX on normal cells and tissues via reducing inflammation, oxidative stress and apoptosis. The current review highlights the anticancer mechanism, side effects and codelivery of curcumin and DOX via nanovehicles.
Collapse
|
57
|
Ashrafizadeh M, Zarrabi A, Hashemipour M, Vosough M, Najafi M, Shahinozzaman M, Hushmandi K, Khan H, Mirzaei H. Sensing the scent of death: Modulation of microRNAs by Curcumin in gastrointestinal cancers. Pharmacol Res 2020; 160:105199. [DOI: 10.1016/j.phrs.2020.105199] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
|
58
|
Wang JY, Song YQ, Peng J, Luo HL. Nanostructured Lipid Carriers Delivering Sorafenib to Enhance Immunotherapy Induced by Doxorubicin for Effective Esophagus Cancer Therapy. ACS OMEGA 2020; 5:22840-22846. [PMID: 32954132 PMCID: PMC7495447 DOI: 10.1021/acsomega.0c02072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/17/2020] [Indexed: 06/11/2023]
Abstract
The tumor microenvironment (TME) plays a significant role in weakening the effect of cancer immunotherapy, which calls for the remodeling of TME. Herein, we fabricated a nanostructured lipid carrier (NLC) to codeliver doxorubicin (Dox) and sorafenib (Sfn) as a drug delivery system (NLC/D-S). The Sfn was expected to regulate the TME of esophagus cancer. As a result, the immune response induced by Dox-related immunogenicity cell death could be fully realized. Our results demonstrated that Sfn was able to remodel the TME through downregulation of regulatory T cells (Treg), activation of effector T cells, and relieving of PD-1 expression, which achieved synergistic effect on the inhibition of primary tumor but also subsequent strong immune response on the regeneration of distant tumor.
Collapse
|
59
|
Concomitant analysis of dasatinib and curcuminoids in a pluronic-based nanoparticle formulation using a novel HPLC method. Chromatographia 2020. [DOI: 10.1007/s10337-020-03956-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
60
|
Avan Z, Biabani Ardakani J, Talebpour Amiri F, Abedi SM, Hosseinimehr SJ. The Potential Usefulness of 99mTc-HYNIC-(Ser)3-LTVPWY Peptide for Predicting HER2 Status Alteration After Chemotherapy in Ovarian Tumor-Bearing Mice. Cancer Biother Radiopharm 2020; 37:862-869. [DOI: 10.1089/cbr.2020.4004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Zahra Avan
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Javad Biabani Ardakani
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fereshteh Talebpour Amiri
- Department of Anatomy and Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
61
|
Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, Wu S, Deng Y, Zhang J, Shao A. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front Mol Biosci 2020; 7:193. [PMID: 32974385 DOI: 10.3389/fmolb.2020.00193/bibtex] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/21/2020] [Indexed: 05/26/2023] Open
Abstract
Nanotechnology has been extensively studied and exploited for cancer treatment as nanoparticles can play a significant role as a drug delivery system. Compared to conventional drugs, nanoparticle-based drug delivery has specific advantages, such as improved stability and biocompatibility, enhanced permeability and retention effect, and precise targeting. The application and development of hybrid nanoparticles, which incorporates the combined properties of different nanoparticles, has led this type of drug-carrier system to the next level. In addition, nanoparticle-based drug delivery systems have been shown to play a role in overcoming cancer-related drug resistance. The mechanisms of cancer drug resistance include overexpression of drug efflux transporters, defective apoptotic pathways, and hypoxic environment. Nanoparticles targeting these mechanisms can lead to an improvement in the reversal of multidrug resistance. Furthermore, as more tumor drug resistance mechanisms are revealed, nanoparticles are increasingly being developed to target these mechanisms. Moreover, scientists have recently started to investigate the role of nanoparticles in immunotherapy, which plays a more important role in cancer treatment. In this review, we discuss the roles of nanoparticles and hybrid nanoparticles for drug delivery in chemotherapy, targeted therapy, and immunotherapy and describe the targeting mechanism of nanoparticle-based drug delivery as well as its function on reversing drug resistance.
Collapse
Affiliation(s)
- Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qiang Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shijie Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
62
|
Yan YB, Tian Q, Zhang JF, Xiang Y. Antitumor effects and molecular mechanisms of action of natural products in ovarian cancer. Oncol Lett 2020; 20:141. [PMID: 32934709 PMCID: PMC7471673 DOI: 10.3892/ol.2020.12001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is a common malignancy and the second leading cause of mortality among females with genital tract cancer. At present, postoperative platinum drugs and paclitaxel-based chemotherapy is the gold standard treatment for ovarian cancer. However, patients who receive this chemotherapy often develop cumulative toxic effects and are prone to chemotherapy resistance. Therefore, it is necessary to determine more effective treatment options that would be better tolerated by patients. Recent studies have reported the therapeutic effects of numerous natural products in patients with ovarian cancer. Notably, these natural ingredients do not induce adverse effects in healthy cells and tissues, suggesting that natural products may serve as a safe alternative treatment for ovarian cancer. The antitumor effects of natural products are attributed to suppression of cell proliferation and metastasis, stimulation of autophagy, improved chemotherapy sensitivity, and induction of apoptosis. The present review focused on the antitumor effects of several natural products, including curcumin, resveratrol, ginsenosides, (-)-epigallocatechin-3-gallate and quercetin, which are increasingly being investigated as therapeutic options in ovarian cancer, and discussed the molecular mechanisms involved in cell proliferation, apoptosis, autophagy, metastasis and sensitization.
Collapse
Affiliation(s)
- Yun-Bo Yan
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Qing Tian
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China.,Department of Cell Biology and Genetics, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Ji-Fang Zhang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China.,Department of Cell Biology and Genetics, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
63
|
Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, Wu S, Deng Y, Zhang J, Shao A. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front Mol Biosci 2020; 7:193. [PMID: 32974385 PMCID: PMC7468194 DOI: 10.3389/fmolb.2020.00193] [Citation(s) in RCA: 449] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Nanotechnology has been extensively studied and exploited for cancer treatment as nanoparticles can play a significant role as a drug delivery system. Compared to conventional drugs, nanoparticle-based drug delivery has specific advantages, such as improved stability and biocompatibility, enhanced permeability and retention effect, and precise targeting. The application and development of hybrid nanoparticles, which incorporates the combined properties of different nanoparticles, has led this type of drug-carrier system to the next level. In addition, nanoparticle-based drug delivery systems have been shown to play a role in overcoming cancer-related drug resistance. The mechanisms of cancer drug resistance include overexpression of drug efflux transporters, defective apoptotic pathways, and hypoxic environment. Nanoparticles targeting these mechanisms can lead to an improvement in the reversal of multidrug resistance. Furthermore, as more tumor drug resistance mechanisms are revealed, nanoparticles are increasingly being developed to target these mechanisms. Moreover, scientists have recently started to investigate the role of nanoparticles in immunotherapy, which plays a more important role in cancer treatment. In this review, we discuss the roles of nanoparticles and hybrid nanoparticles for drug delivery in chemotherapy, targeted therapy, and immunotherapy and describe the targeting mechanism of nanoparticle-based drug delivery as well as its function on reversing drug resistance.
Collapse
Affiliation(s)
- Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qiang Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shijie Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
64
|
Ashrafizadeh M, Zarrabi A, Orouei S, Saberifar S, Salami S, Hushmandi K, Najafi M. Recent advances and future directions in anti-tumor activity of cryptotanshinone: A mechanistic review. Phytother Res 2020; 35:155-179. [PMID: 33507609 DOI: 10.1002/ptr.6815] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/29/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
In respect to the enhanced incidence rate of cancer worldwide, studies have focused on cancer therapy using novel strategies. Chemotherapy is a common strategy in cancer therapy, but its adverse effects and chemoresistance have limited its efficacy. So, attempts have been directed towards minimally invasive cancer therapy using plant derived-natural compounds. Cryptotanshinone (CT) is a component of salvia miltiorrihiza Bunge, well-known as Danshen and has a variety of therapeutic and biological activities such as antioxidant, anti-inflammatory, anti-diabetic and neuroprotective. Recently, studies have focused on anti-tumor activity of CT against different cancers. Notably, this herbal compound is efficient in cancer therapy by targeting various molecular signaling pathways. In the present review, we mechanistically describe the anti-tumor activity of CT with an emphasis on molecular signaling pathways. Then, we evaluate the potential of CT in cancer immunotherapy and enhancing the efficacy of chemotherapy by sensitizing cancer cells into anti-tumor activity of chemotherapeutic agents, and elevating accumulation of anti-tumor drugs in cancer cells. Finally, we mention strategies to enhance the anti-tumor activity of CT, for instance, using nanoparticles to provide targeted drug delivery.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.,Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul, Turkey
| | - Sima Orouei
- MSc. Student, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sedigheh Saberifar
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Saeed Salami
- DVM. Graduated, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
65
|
Li J, Zheng L, Wang R, Sun D, Liang S, Wu J, Liu Y, Tian X, Li T, Yang Y, Han L. Synergistic Combination of Sodium Aescinate-Stabilized, Polymer-Free, Twin-Like Nanoparticles to Reverse Paclitaxel Resistance. Int J Nanomedicine 2020; 15:5839-5853. [PMID: 32848393 PMCID: PMC7428345 DOI: 10.2147/ijn.s259432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The development of paclitaxel (PTX) resistance seriously restricts its clinical efficacy. An attractive option for combating resistance is inhibiting the expression of P-glycoprotein (P-gp) in tumor cells. We have reported that flavokawain A (FKA) inhibited P-gp protein expression in PTX-resistant A549 (A549/T) cells, indicating that FKA combined with PTX may reverse PTX resistance. However, due to the variable pharmacokinetics of FKA and PTX, the conventional cocktail combination in clinics may cause uncertainty of treatment efficacy in vivo. MATERIALS AND METHODS To synergistically elevate the anti-cancer activity of PTX and FKA in vivo, the national medical products administration (NMPA) approved sodium aescinate (Aes) was utilized to stabilize hydrophobic PTX and FKA to form polymer-free twin like PTX-A nanoparticles (NPs) and FKA-A NPs. RESULTS The resulting nanoparticles prepared simply by nanoprecipitation possessed similar particle size, good stability and ultrahigh drug loadings of up to 50%. With the aid of Aes, these two drugs accumulated in tumor tissue by passive targeting and were efficiently taken up by A549/T cells; this resulted in significant suppression of tumor growth in A549/T homograft mice at a low PTX dose (2.5 mg·kg-1). Synergistic effects and reversed PTX resistance were achieved by the combination of PTX-A NPs and FKA-A NPs by inhibiting P-gp expression in tumor cells. CONCLUSION Using NMPA-approved Aes to prepare twin-like nanoparticles without introducing any new materials provides an efficient platform for combination chemotherapy and clinical translation.
Collapse
Affiliation(s)
- Juan Li
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Lei Zheng
- Department of Pharmacy, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250031, People’s Republic of China
| | - Rongmei Wang
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Deqing Sun
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Shuang Liang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, People’s Republic of China
| | - Jing Wu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Xiaona Tian
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Tingting Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Yang Yang
- China National Center for Biotechnology Development, Beijing100039, People’s Republic of China
| | - Leiqiang Han
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| |
Collapse
|
66
|
Gong K, Dong Y, Wang L, Duan Y, Yu J, Sun Y, Bai M, Duan Y. Nanoparticle BAF312@CaP-NP Overcomes Sphingosine-1-Phosphate Receptor-1-Mediated Chemoresistance Through Inhibiting S1PR1/P-STAT3 Axis in Ovarian Carcinoma. Int J Nanomedicine 2020; 15:5561-5571. [PMID: 32801704 PMCID: PMC7414939 DOI: 10.2147/ijn.s248667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/10/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose Platinum/paclitaxel-based chemotherapy is the strategy for ovarian cancer, but chemoresistance, inherent or acquired, occurs and hinders therapy. Therefore, further understanding of the mechanisms of drug resistance and adoption of novel therapeutic strategies are urgently needed. Methods In this study, we report that sphingosine-1-phosphate receptor-1 (S1PR1)-mediated chemoresistance for ovarian cancer. Then we developed nanoparticles with a hydrophilic PEG2000 chain and a hydrophobic DSPE and biodegradable CaP (calcium ions and phosphate ions) shell with pH sensitivity as a delivery system (CaP-NPs) to carry BAF312, a selective antagonist of S1PR1 (BAF312@CaP-NPs), to overcome the cisplatin (DDP) resistance of the ovarian cancer cell line SKOV3DR. Results We found that S1PR1 affected acquired chemoresistance in ovarian cancer by increasing the phosphorylated-signal transduction and activators of transcription 3 (P-STAT3) level. The mean size and zeta potential of BAF312@CaP-NPs were 116 ± 4.341 nm and −9.67 ± 0.935 mV, respectively. The incorporation efficiency for BAF312 in the CaP-NPs was 76.1%. The small size of the nanoparticles elevated their enrichment in the tumor, and the degradable CaP shell with smart pH sensitivity of the BAF312@CaP-NPs ensured the release of BAF312 in the acidic tumor niche. BAF312@CaP-NPs caused substantial cytotoxicity in DDP-resistant ovarian cancer cells by downregulating S1PR1 and P-STAT3 levels. Conclusion We found that BAF312@CaP-NPs act as an effective and selective delivery system for overcoming S1PR1-mediated chemoresistance in ovarian carcinoma by inhibiting S1PR1 and P-STAT3.
Collapse
Affiliation(s)
- Ke Gong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| | - Liting Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| | - Yi Duan
- Department of Clinical Medicine, North Sichuan Medical College, Sichuan 637100, People's Republic of China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| | - Min Bai
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| |
Collapse
|
67
|
Wan J, Wang J, Zhou M, Rao Z, Ling X. A cell membrane vehicle co-delivering sorafenib and doxorubicin remodel the tumor microenvironment and enhance immunotherapy by inducing immunogenic cell death in lung cancer cells. J Mater Chem B 2020; 8:7755-7765. [PMID: 32735004 DOI: 10.1039/d0tb01052a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cancer immunotherapy is a promising approach for cancer therapy but is usually hindered by the inhibition of the tumor microenvironment (TME). Herein, we developed a cell membrane vehicle (CV) to co-deliver doxorubicin (Dox) and sorafenib (Sfn) as a drug delivery system (CV/D-S) to regulate the TME and sensitize the immunogenic cell death (ICD)-induced immune response against tumors. The CV/D-S showed high stability, acid-responsive drug release, high biocompatibility with tumor-specific cellular uptake, and target-ability that preferably resulted in the in vitro and in vivo anticancer performance. Most importantly, the Dox in the DDS can induce significant ICD while Sfn was able to remodel the TME, downregulate Treg, activate effector T cells and relieve programmed cell death protein 1 (PD-1) expression. As a result, the synergistic effect of Dox and Sfn achieved strong immune response in CV/D-S treated mice, which is believed to open a new window for the design and development of future platforms for the more effective immunotherapy of cancer.
Collapse
Affiliation(s)
- Jun Wan
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P. R. China
| | | | | | | | | |
Collapse
|
68
|
Yang B, Hao A, Chen L. Mirror siRNAs loading for dual delivery of doxorubicin and autophagy regulation siRNA for multidrug reversing chemotherapy. Biomed Pharmacother 2020; 130:110490. [PMID: 32712530 DOI: 10.1016/j.biopha.2020.110490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
The multidrug resistance (MDR) which widely observed in multiple cancer types is responsible for the poor chemotherapy benefits of doxorubicin (Dox). Here in our study, Dox was firstly loaded into a scramble siRNA and then condensed by polyethyleneimine (PEI) 25k together with anti-autophagy siRNA, the obtained PEI/Si-D containing mirror RNAs was further coated with hyaluronic acid (HA) to shield the surface charge of PEI and offer tumor-homing property that finally developed a platform for effective cancer chemotherapy (HP/Si-D). Our results revealed that the obtained HP/Si-D was showed high stability and biocompatibility with promising transfection profile. As a result, the anti-autophagy siRNA downregulated autophagy level of target cells, which further decreased ATP supply to enhance drug retention and cell cycle arrest. These results contributed significantly to reverse the MDR of A549/Dox (Dox resistance A549 cell line) cells with promising in vitro and in vivo results, which suggested the potential of effective MDR cancer therapy using synergistic anti-autophagy and chemotherapy.
Collapse
Affiliation(s)
- Bo Yang
- Department of Thoracic Surgery, Anyang Tumour Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan 455000, China
| | - Anlin Hao
- Department of Thoracic Surgery, Anyang Tumour Hospital (The Fourth Affiliated Hospital of Henan University of Science and Technology), Anyang, Henan 455000, China
| | - Lin Chen
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
69
|
Ashrafizadeh M, Zarrabi A, Hashemi F, Moghadam ER, Hashemi F, Entezari M, Hushmandi K, Mohammadinejad R, Najafi M. Curcumin in cancer therapy: A novel adjunct for combination chemotherapy with paclitaxel and alleviation of its adverse effects. Life Sci 2020; 256:117984. [PMID: 32593707 DOI: 10.1016/j.lfs.2020.117984] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
Dealing with cancer is of importance due to enhanced incidence rate of this life-threatening disorder. Chemotherapy is an ideal candidate in overcoming and eradication of cancer. To date, various chemotherapeutic agents have been applied in cancer therapy and paclitaxel (PTX) is one of them. PTX is a key member of taxane family with potential anti-tumor activity against different cancers. Notably, PTX has demonstrated excellent proficiency in elimination of cancer in clinical trials. This chemotherapeutic agent is isolated from Taxus brevifolia, and is a tricyclic diterpenoid. However, resistance of cancer cells into PTX chemotherapy has endangered its efficacy. Besides, administration of PTX is associated with a number of side effects such as neurotoxicity, hepatotoxicity, cardiotoxicity and so on, demanding novel strategies in obviating PTX issues. Curcumin is a pharmacological compound with diverse therapeutic effects including anti-tumor, anti-oxidant, anti-inflammatory, anti-diabetic and so on. In the current review, we demonstrate that curcumin, a naturally occurring nutraceutical compound is able to enhance anti-tumor activity of PTX against different cancers. Besides, curcumin administration reduces adverse effects of PTX due to its excellent pharmacological activities. These topics are discussed with an emphasis on molecular pathways to provide direction for further studies in revealing other signaling networks.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey; Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzia, Istanbul 34956, Turkey
| | - Farid Hashemi
- DVM, Graduated, Young Researcher and Elite Club, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|