101
|
Chang YT, Wang CCN, Wang JY, Lee TE, Cheng YY, Morris-Natschke SL, Lee KH, Hung CC. Tenulin and isotenulin inhibit P-glycoprotein function and overcome multidrug resistance in cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 53:252-262. [PMID: 30668405 PMCID: PMC6421864 DOI: 10.1016/j.phymed.2018.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/24/2018] [Accepted: 09/03/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Multidrug resistance (MDR) in cancer is one of the main obstacles in treatment with chemotherapy. Drug efflux through P-glycoprotein is the major mechanism involved in MDR. A potential strategy to provide the best possible clinical outcomes is to develop P-glycoprotein (P-gp) inhibitors from natural products. PURPOSE The present study investigated the effects of the natural sesquiterpene lactone tenulin and its derivative isotenulin on human P-gp; the mechanisms of kinetic interactions were also explored. METHODS The human P-gp (ABCB1/Flp-In™-293) stable expression cells were established by using the Flp-In™ system. The effects of tenulin and isotenulin on cell viability were evaluated by SRB assays in established cell lines, sensitive cancer cell line (HeLaS3), and resistant cancer cell line (KB-vin). The transporter inhibition ability was evaluated by calcein-AM uptake assays. The P-gp inhibition kinetics of tenulin and isotenulin were evaluated by rhodamine123 and doxorubicin efflux assays. The ATPase activity was evaluated with the Pgp-Glo™ Assay System. RESULTS Tenulin and isotenulin significantly inhibited the P-gp efflux function by stimulating P-gp ATPase activity. Tenulin and isotenulin interacted with the effluxes of rhodamine 123 and doxorubicin through a competitive and noncompetitive mechanism, respectively. The combinations of tenulin and isotenulin with chemotherapeutic drugs significantly resensitized MDR cancer cells. CONCLUSION These results suggested that tenulin and isotenulin are potential candidates to be developed for synergistic treatment of MDR cancers.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Antineoplastic Agents, Phytogenic/pharmacology
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Screening Assays, Antitumor
- HeLa Cells
- Humans
- Lactones/pharmacology
- Rhodamine 123/pharmacology
- Sesquiterpenes/pharmacology
Collapse
Affiliation(s)
- Ying-Tzu Chang
- Department of Pharmacy, College of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan, ROC
| | - Charles C N Wang
- Department of Bioinformatics and Medical Engineering, Asia University. 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan, ROC
| | - Jiun-Yi Wang
- Department of Healthcare Administration, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan, ROC
| | - Tsui-Er Lee
- Office of Physical Education, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan, ROC
| | - Yung-Yi Cheng
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, United States.; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan, ROC
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, United States.; Chinese Medicine Research and Development Center, China Medical University and Hospital, 2 Yude Road, Taichung 40447, Taiwan, ROC
| | - Chin-Chuan Hung
- Department of Pharmacy, College of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan, ROC; Department of Pharmacy, China Medical University Hospital, 2 Yude Road, Taichung 40447, Taiwan, ROC.
| |
Collapse
|
102
|
Zhang H, Jiang H, Zhang H, Liu J, Hu X, Chen L. Ribophorin II potentiates P-glycoprotein- and ABCG2-mediated multidrug resistance via activating ERK pathway in gastric cancer. Int J Biol Macromol 2019; 128:574-582. [PMID: 30710584 DOI: 10.1016/j.ijbiomac.2019.01.195] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022]
Abstract
Multidrug resistance (MDR) is a critical reason of cancer chemotherapy failure. Ribophorin II (RPN2) has emerged as a vital regulator of MDR in multiple cancers including gastric cancer (GC). However, the roles and molecular mechanisms of RPN2 in MDR have not been well featured till now. The present study aimed to explore the roles and molecular mechanisms of RPN2 in MDR of drug-resistant GC cells. Results showed that the expressions of RPN2, multidrug resistance 1 (MDR1), and ATP binding cassette subfamily G member 2 (ABCG2) were upregulated in SGC7901/DDP and SGC7901/VCR cells. Knockdown of RPN2 alleviated MDR through downregulating MDR1 and ABCG2 expressions in SGC7901/DDP and SGC7901/VCR cells. RPN2 depletion inhibited the activation of MEK/ERK pathway. RPN2 overexpression enhanced MDR by upregulating P-glycoprotein (P-gp) and ABCG2 protein expressions in SGC7901/DDP or SGC7901/VCR cells, while this effect of RPN2 was abrogated by ERK knockdown or treatment with ERK inhibitor PD98059. Our findings suggested that RPN2 potentiated P-gp- and ABCG2-mediated MDR via activating MEK/ERK pathway in GC, hinting the critical values of RPN2 in ameliorating MDR and providing a promising target for GC therapy.
Collapse
Affiliation(s)
- Hongzhi Zhang
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China.
| | - Huijuan Jiang
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Huixiang Zhang
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Juncai Liu
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Xigang Hu
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Lei Chen
- Department of Radiotherapy, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| |
Collapse
|
103
|
Gawel AM, Godlewska M, Grech-Baran M, Stachurska A, Gawel D. MIX2: A Novel Natural Multi-Component Modulator of Multidrug-Resistance and Hallmarks of Cancer Cells. Nutr Cancer 2019; 71:334-347. [PMID: 30676767 DOI: 10.1080/01635581.2018.1560480] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Multidrug resistance is one of the key barriers suppressing the effectiveness of drug therapies of malignant tumors. Here, we report a study on the effect of a mix of natural extracts (MIX2) prepared from fresh fruits of Prunus spinosa, Crataegus monogyna, Sorbus aucuparia, and Euonymus europaeus on the classic hallmarks of cancer cells and the expression of multidrug resistance proteins. In the studies, HeLa and T98G cell lines, and classic methods of molecular biology, including RT-qPCR, Western blot, flow cytometry, and confocal imaging, were used. Additionally, migration, adhesion, and proliferation assays were performed. The obtained results indicate that the MIX2 cocktail presents strong anti-cancer properties. MIX2 is not toxic, but at the same time significantly alters the migration, proliferation, and adhesion of tumor cells. Furthermore, it was found that cells exposed to the mixture presented a significantly reduced expression level of genes associated with MDR, including ABCB1, which encodes for glycoprotein P. In vitro data showed that MIX2 effectively sensitizes tumor cells to doxorubicin. We postulate that modulation of the multidrug resistance phenotype of tumors with the use of MIX2 may be considered as a safe and applicable tool in sustaining drug delivery therapies of malignancies.
Collapse
Affiliation(s)
- Agata M Gawel
- a Department of Biochemistry and Molecular Biology , Centre of Postgraduate Medical Education , Warsaw , Poland
| | - Marlena Godlewska
- a Department of Biochemistry and Molecular Biology , Centre of Postgraduate Medical Education , Warsaw , Poland
| | - Marta Grech-Baran
- b Laboratory of Plant Pathogenesis , Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland
| | - Anna Stachurska
- c Department of Immunohematology , Centre of Postgraduate Medical Education , Warsaw , Poland
| | - Damian Gawel
- a Department of Biochemistry and Molecular Biology , Centre of Postgraduate Medical Education , Warsaw , Poland
| |
Collapse
|
104
|
Belen’kii LI, Evdokimenkova YB. The literature of heterocyclic chemistry, part XVII, 2017. ADVANCES IN HETEROCYCLIC CHEMISTRY 2019:337-418. [DOI: 10.1016/bs.aihch.2019.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
105
|
Li H, Jiao R, Mu J, Xu S, Li X, Wang X, Li Z, Xu J, Hua H, Li D. Bioactive Natural Spirolactone-Type 6,7- seco- ent-Kaurane Diterpenoids and Synthetic Derivatives. Molecules 2018; 23:molecules23112914. [PMID: 30413071 PMCID: PMC6278314 DOI: 10.3390/molecules23112914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/04/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023] Open
Abstract
Diterpenoids are widely distributed natural products and have caused considerable interest because of their unique skeletons and antibacterial and antitumor activities and so on. In light of recent discoveries, ent-kaurane diterpenoids, which exhibit a wide variety of biological activities, such as anticancer and anti-inflammatory activities, pose enormous potential to serve as a promising candidate for drug development. Among them, spirolactone-type 6,7-seco-ent-kaurane diterpenoids, with interesting molecular skeleton, complex oxidation patterns, and bond formation, exhibit attractive activities. Furthermore, spirolactone-type diterpenoids have many modifiable sites, which allows for linking to various substituents, suitable for further medicinal study. Hence, some structurally modified derivatives with improved cytotoxicity activities are also achieved. In this review, natural bioactive spirolactone-type diterpenoids and their synthetic derivatives were summarized.
Collapse
Affiliation(s)
- Haonan Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Runwei Jiao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Jiahui Mu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Shengtao Xu
- Department of Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China.
| | - Xianhua Wang
- School of Public Health, Qingdao University, Qingdao 266021, China.
| | - Zhanlin Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Jinyi Xu
- Department of Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
106
|
Antiproliferative Effects of Alkaloid Evodiamine and Its Derivatives. Int J Mol Sci 2018; 19:ijms19113403. [PMID: 30380774 PMCID: PMC6274956 DOI: 10.3390/ijms19113403] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/20/2018] [Accepted: 10/24/2018] [Indexed: 12/18/2022] Open
Abstract
Alkaloids, a category of natural products with ring structures and nitrogen atoms, include most U.S. Food and Drug Administration approved plant derived anti-cancer agents. Evodiamine is an alkaloid with attractive multitargeting antiproliferative activity. Its high content in the natural source ensures its adequate supply on the market and guarantees further medicinal study. To the best of our knowledge, there is no systematic review about the antiproliferative effects of evodiamine derivatives. Therefore, in this article the review of the antiproliferative activities of evodiamine will be updated. More importantly, the antiproliferative activities of structurally modified new analogues of evodiamine will be summarized for the first time.
Collapse
|
107
|
Muthusamy G, Gunaseelan S, Prasad NR. Ferulic acid reverses P-glycoprotein-mediated multidrug resistance via inhibition of PI3K/Akt/NF-κB signaling pathway. J Nutr Biochem 2018; 63:62-71. [PMID: 30342318 DOI: 10.1016/j.jnutbio.2018.09.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/27/2018] [Accepted: 09/27/2018] [Indexed: 02/07/2023]
Abstract
In this study, the modulatory effect of ferulic acid on P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) was examined in KB ChR8-5 resistant cells and drug-resistant tumor xenografts. We observed that ferulic acid enhanced the cytotoxicity of doxorubicin and vincristine in the P-gp overexpressing KB ChR8-5 cells. Further, ferulic acid enhances the doxorubicin induced γH2AX foci formation and synergistically augmented doxorubicin-induced apoptotic signaling in the drug-resistant cells. It has also been noticed that NF-κB nuclear translocation was suppressed by ferulic acid and that this response might be associated with the modulation of phosphatidyinositol 3-kinase (PI3K)/Akt/signaling pathway. We also found that ferulic acid and doxorubicin combination reduced the size of KB ChR8-5 tumor xenograft by threefold as compared to doxorubicin-alone treated group. Thus, ferulic acid contributes to the reversal of the MDR through suppression of P-gp expression via the inhibition of PI3K/Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ganesan Muthusamy
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, 608002, Tamilnadu, India
| | - Srithar Gunaseelan
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, 608002, Tamilnadu, India
| | - Nagarajan Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, 608002, Tamilnadu, India.
| |
Collapse
|
108
|
Krauß J, Bracher F. Pharmacokinetic Enhancers (Boosters)-Escort for Drugs against Degrading Enzymes and Beyond. Sci Pharm 2018; 86:scipharm86040043. [PMID: 30262788 DOI: 10.3390/scipharm86040043] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 02/04/2023] Open
Abstract
Pharmacokinetic enhancers (boosters) are compounds used in combination with a primary therapeutic agent (drug) and are not used for their direct effects on the disease but because they enhance or restore the activity of the primary agent. Hence, in certain cases, they represent an indispensable escort for enzyme-labile drugs. Pharmacokinetic enhancers can exert their activity on different ways. In the most common case, they inhibit enzymes such as human cytochrome P450 enzymes in the liver or other organs and, thereby, block or reduce undesired metabolism and inactivation of the primary drug. In this review, an overview will be given on the therapeutically most important classes of pharmacokinetic enhancers like β-lactamase inhibitors, inhibitors of CYP (cytochrome P450) enzymes in HIV therapy and hepatitis C, boosters for fluoropyrimidine-type anticancer agents, compounds utilized for enabling therapy of Parkinson's disease with levodopa, and others. Inhibitors of efflux pumps in both pathogenic bacteria and tumor cells will be addresses shortly.
Collapse
Affiliation(s)
- Jürgen Krauß
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians University, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Franz Bracher
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians University, Butenandtstr. 5-13, 81377 Munich, Germany.
| |
Collapse
|
109
|
Sugisawa N, Ohnuma S, Ueda H, Murakami M, Sugiyama K, Ohsawa K, Kano K, Tokuyama H, Doi T, Aoki J, Ishida M, Kudoh K, Naitoh T, Ambudkar SV, Unno M. Novel Potent ABCB1 Modulator, Phenethylisoquinoline Alkaloid, Reverses Multidrug Resistance in Cancer Cell. Mol Pharm 2018; 15:4021-4030. [PMID: 30052463 DOI: 10.1021/acs.molpharmaceut.8b00457] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
ATP-binding cassette (ABC) transporters, which are concerned with the efflux of anticancer drugs from cancer cells, have a pivotal role in multidrug resistance (MDR). In particular, ABCB1 is a well-known ABC transporter that develops MDR in many cancer cells. Some ABCB1 modulators can reverse ABCB1-mediated MDR; however, no modulators with clinical efficacy have been approved. The aim of this study was to identify novel ABCB1 modulators by using high-throughput screening. Of the 5861 compounds stored at Tohoku University, 13 compounds were selected after the primary screening via a fluorescent plate reader-based calcein acetoxymethylester (AM) efflux assay. These 13 compounds were validated in a flow cytometry-based calcein AM efflux assay. Two isoquinoline derivatives were identified as novel ABCB1 inhibitors, one of which was a phenethylisoquinoline alkaloid, (±)-7-benzyloxy-1-(3-benzyloxy-4-methoxyphenethyl)-1,2,3,4-tetrahydro-6-methoxy-2-methylisoquinoline oxalate. The compound, a phenethylisoquinoline alkaloid, was subsequently evaluated in the cytotoxicity assay and shown to significantly enhance the reversal of ABCB1-mediated MDR. In addition, the compound activated the ABCB1-mediated ATP hydrolysis and inhibited the photolabeling of ABCB1 with [125I]-iodoarylazidoprazosin. Furthermore, the compound also reversed the resistance to paclitaxel without increasing the toxicity in the ABCB1-overexpressing KB-V1 cell xenograft model. Overall, we concluded that the newly identified phenethylisoquinoline alkaloid reversed ABCB1-mediated MDR through direct interaction with the substrate-binding site of ABCB1. These findings may contribute to the development of more potent and less toxic ABCB1 modulators, which could overcome ABCB1-mediated MDR.
Collapse
Affiliation(s)
- Norihiko Sugisawa
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan
| | - Shinobu Ohnuma
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan
| | - Hirofumi Ueda
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Megumi Murakami
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan.,Laboratory of Cell Biology, Center for Cancer Research , National Cancer Institute, NIH , Bethesda , Maryland 20892 , United States
| | - Kyoko Sugiyama
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Kosuke Ohsawa
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Hidetoshi Tokuyama
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Takayuki Doi
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Masaharu Ishida
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan
| | - Katsuyoshi Kudoh
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan
| | - Takeshi Naitoh
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research , National Cancer Institute, NIH , Bethesda , Maryland 20892 , United States
| | - Michiaki Unno
- Department of Surgery , Tohoku University Graduate School of Medicine , Sendai 980-8574 , Japan
| |
Collapse
|
110
|
The Effects of Synthetically Modified Natural Compounds on ABC Transporters. Pharmaceutics 2018; 10:pharmaceutics10030127. [PMID: 30096910 PMCID: PMC6161255 DOI: 10.3390/pharmaceutics10030127] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance (MDR) is a major hurdle which must be overcome to effectively treat cancer. ATP-binding cassette transporters (ABC transporters) play pivotal roles in drug absorption and disposition, and overexpression of ABC transporters has been shown to attenuate cellular/tissue drug accumulation and thus increase MDR across a variety of cancers. Overcoming MDR is one desired approach to improving the survival rate of patients. To date, a number of modulators have been identified which block the function and/or decrease the expression of ABC transporters, thereby restoring the efficacy of a range of anticancer drugs. However, clinical MDR reversal agents have thus far proven ineffective and/or toxic. The need for new, effective, well-tolerated and nontoxic compounds has led to the development of natural compounds and their derivatives to ameliorate MDR. This review evaluates whether synthetically modifying natural compounds is a viable strategy to generate potent, nontoxic, ABC transporter inhibitors which may potentially reverse MDR.
Collapse
|
111
|
Wang B, Ma LY, Wang JQ, Lei ZN, Gupta P, Zhao YD, Li ZH, Liu Y, Zhang XH, Li YN, Zhao B, Chen ZS, Liu HM. Discovery of 5-Cyano-6-phenylpyrimidin Derivatives Containing an Acylurea Moiety as Orally Bioavailable Reversal Agents against P-Glycoprotein-Mediated Mutidrug Resistance. J Med Chem 2018; 61:5988-6001. [PMID: 29975529 DOI: 10.1021/acs.jmedchem.8b00335] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
P-glycoprotein (ABCB1)-mediated multidrug resistance (MDR) has become a major obstacle in successful cancer chemotherapy, which attracted much effort to develop clinically useful compounds to reverse MDR. Here, we designed and synthesized a novel series of derivatives with a 5-cyano-6-phenylpyrimidine scaffold and evaluated their potential reversal activities against MDR. Among these compounds, 55, containing an acylurea appendage, showed the most potent activity in reversing paclitaxel resistance in SW620/AD300 cells. Further studies demonstrated 55 could increase accumulation of PTX, interrupt ABCB1-mediated Rh123 accumulation and efflux, stimulate ABCB1 ATPase activity, and especially have no effect on CYP3A4 activity, which avoid drug interaction caused toxicity. More importantly, 55 significantly enhanced the efficacy of PTX against the SW620/AD300 cell xenograft without obvious side effects for orally intake. Given all that, the pyrimidine-acylurea based ABCB1 inhibitor may be a promising lead in developing new efficacious ABCB1-dependent MDR modulator.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Administration, Oral
- Biological Availability
- Cell Line, Tumor
- Dose-Response Relationship, Drug
- Drug Design
- Drug Resistance, Multiple/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Intracellular Space/drug effects
- Intracellular Space/metabolism
- Models, Molecular
- Molecular Docking Simulation
- Protein Conformation
- Protein Transport/drug effects
- Pyrimidines/chemistry
- Pyrimidines/metabolism
- Pyrimidines/pharmacokinetics
- Pyrimidines/pharmacology
- Urea/chemistry
Collapse
Affiliation(s)
- Bo Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Li-Ying Ma
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences , St. John's University , Queens , New York 11439 , United States
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences , St. John's University , Queens , New York 11439 , United States
| | - Pranav Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences , St. John's University , Queens , New York 11439 , United States
| | - Yuan-Di Zhao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Zhong-Hua Li
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Ying Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Xin-Hui Zhang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Ya-Nan Li
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Bing Zhao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences , St. John's University , Queens , New York 11439 , United States
| | - Hong-Min Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education; Key Laboratory of Henan Province for Drug Quality and Evaluation; School of Pharmaceutical Sciences , Zhengzhou University , Zhengzhou 450001 , P. R. China
| |
Collapse
|
112
|
Shi M, Zhang J, Li X, Pan S, Li J, Yang C, Hu H, Qiao M, Chen D, Zhao X. Mitochondria-targeted delivery of doxorubicin to enhance antitumor activity with HER-2 peptide-mediated multifunctional pH-sensitive DQAsomes. Int J Nanomedicine 2018; 13:4209-4226. [PMID: 30140154 PMCID: PMC6054761 DOI: 10.2147/ijn.s163858] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Introduction Multidrug resistance (MDR) of breast cancer is the major challenge to successful chemotherapy while mitochondria-targeting therapy was a promising strategy to overcome MDR. Materials and methods In this study, HER-2 peptide-PEG2000-Schiff base-cholesterol (HPSC) derivate was synthesized successfully and incorporated it on the surface of the doxorubicin (DOX)-loaded dequalinium (DQA) chloride vesicle (HPS-DQAsomes) to treat drug-resistant breast cancer. Evaluations were performed using human breast cancer cell and DOX-resistant breast cancer cell lines (MCF-7 and MCF-7/ADR). Results The particle size of HPS-DQAsomes was ~110 nm with spherical shape. In vitro cytotoxicity assay indicated that HPS-DQAsomes could increase the cytotoxicity against MCF-7/ADR cell line. Cellular uptake and mitochondria-targeting assay demonstrated that HPS-DQAsomes could target delivering therapeutical agent to mitochondria and inducing mitochondria-driven apoptosis process. In vivo antitumor assay suggested that HPS-DQAsomes could reach favorable antitumor activity due to both tumor targetability and sub-organelles’ targetability. Histological assay also indicated that HPS-DQAsomes showed a strong apoptosis-inducing effect. No obvious systematic toxicity of HPS-DQAsomes could be observed. Conclusion In summary, multifunctional HPS-DQAsomes provide a novel and versatile approach for overcoming MDR via mitochondrial pathway in cancer treatment.
Collapse
Affiliation(s)
- Menghao Shi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Jiulong Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Xiaowei Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Shuang Pan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Jie Li
- Department of Pharmacy, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, People's Republic of China
| | - Chunrong Yang
- College Pharmacy of Jiamusi University, Jiamusi, Heilongjiang 154007, People's Republic of China
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Mingxi Qiao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| | - Xiuli Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China,
| |
Collapse
|
113
|
Nevskaya AA, Matveeva MD, Borisova TN, Niso M, Colabufo NA, Boccarelli A, Purgatorio R, de Candia M, Cellamare S, Voskressensky LG, Altomare CD. A New Class of 1-Aryl-5,6-dihydropyrrolo[2,1-a]isoquinoline Derivatives as Reversers of P-Glycoprotein-Mediated Multidrug Resistance in Tumor Cells. ChemMedChem 2018; 13:1588-1596. [PMID: 29802687 DOI: 10.1002/cmdc.201800177] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/16/2018] [Indexed: 12/20/2022]
Abstract
A number of aza-heterocyclic compounds, which share the 5,6-dihydropyrrolo[2,1-a]isoquinoline (DHPIQ) scaffold with members of the lamellarin alkaloid family, were synthesized and evaluated for their ability to reverse in vitro multidrug resistance in cancer cells through inhibition of P-glycoprotein (P-gp) and/or multidrug-resistance-associated protein 1. Most of the investigated DHPIQ compounds proved to be selective P-gp modulators, and the most potent modulator, 8,9-diethoxy-1-(3,4-diethoxyphenyl)-3-(furan-2-yl)-5,6-dihydropyrrolo[2,1-a]isoquinoline-2-carbaldehyde, attained sub-micromolar inhibitory potency (IC50 : 0.19 μm). Schiff bases prepared by the condensation of some 1-aryl-DHPIQ aldehydes with p-aminophenol also proved to be of some interest, and one of them, 4-((1-(4-fluorophenyl)-5,6-dihydro-8,9-dimethoxypyrrolo[2,1-a]isoquinolin-2-yl)methyleneamino)phenol, had an IC50 value of 1.01 μm. In drug combination assays in multidrug-resistant cells, some DHPIQ compounds, at nontoxic concentrations, significantly increased the cytotoxicity of doxorubicin in a concentration-dependent manner. Studies of structure-activity relationships and investigation of the chemical stability of Schiff bases provided physicochemical information useful for molecular optimization of lamellarin-like cytotoxic drugs active toward chemoresistant tumors as well as nontoxic reversers of P-gp-mediated multidrug resistance in tumor cells.
Collapse
Affiliation(s)
- Alisa A Nevskaya
- Organic Chemistry Department, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - Maria D Matveeva
- Organic Chemistry Department, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - Tatiana N Borisova
- Organic Chemistry Department, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - Mauro Niso
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Nicola A Colabufo
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Angelina Boccarelli
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Rosa Purgatorio
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Saverio Cellamare
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Leonid G Voskressensky
- Organic Chemistry Department, Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - Cosimo D Altomare
- Department of Pharmacy-Drug Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| |
Collapse
|
114
|
Liquid Chromatography Coupled with Linear Ion Trap Hybrid OrbitrapMass Spectrometry for Determination of Alkaloids in Sinomeniumacutum. Molecules 2018; 23:molecules23071634. [PMID: 29973556 PMCID: PMC6099952 DOI: 10.3390/molecules23071634] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 01/15/2023] Open
Abstract
The characterization of alkaloids is challenging because of the diversity of structures and the complicated fragmentation of collision induced structural dissociation in mass spectrometry. In this study, we analyzed the alkaloids in Sinomenium acutum (Thunb.) Rehderet Wil by high resolution mass spectrometry. Chromatographic separation was achieved on a Phenomenex Kinetex C18 (2.1 mm × 100 mm, 2.6 μm) column with a mobile phase consisting of acetonitrile and water (0.1% formic acid) under gradient elution. A total of 52 alkaloids were well separated and 45 of them were structurally characterized, including morphinans, aporphines, benzylisoquinolines, and protoberberines. Specially, mass spectrometric study of the morphinan alkaloids were explicitly investigated. Electrostatic potential plot from simulation was calculated for determination of protonation sites. Further fragmentation analysis suggested that the C₃H₇N, CH₄O, and H₂O elimination was displayed in MS² spectrum. These fragmentation pathways are universal for morphinan alkaloids having methoxy substituted cyclohexenone or cyclohexadienone moieties. Additionally, for nitrogen oxides, an ion-neutral complex intermediate is involved in the fragmentation process, generating additional oxygenated ions. All these results provided the universal rules of fragmentation used for detection of alkaloids, and will be expected to be highly useful for comprehensive study of multi-components in the herbal medicine analysis.
Collapse
|
115
|
AboulFotouh K, Allam AA, El-Badry M, El-Sayed AM. Self-emulsifying drug–delivery systems modulate P-glycoprotein activity: role of excipients and formulation aspects. Nanomedicine (Lond) 2018; 13:1813-1834. [DOI: 10.2217/nnm-2017-0354] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Self-emulsifying drug–delivery systems (SEDDS) have been widely employed to ameliorate the oral bioavailability of P-glycoprotein (P-gp) substrate drugs and to overcome multidrug resistance in cancer cells. However, the role of formulation aspects in the reduced P-gp activity is not fully understood. In this review, we first explore the role of various SEDDS excipients in the reduced P-gp activity with the main emphasis on the effective excipient concentration range for excipient-mediated modulation of P-gp activity and then we discuss the synergistic effect of various formulation aspects on the excipient-mediated modulation of P-gp activity. This review provides an approach to develop a rationally designed SEDDS to overcome P-gp-mediated drug efflux.
Collapse
Affiliation(s)
- Khaled AboulFotouh
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Ayat A Allam
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mahmoud El-Badry
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Ahmed M El-Sayed
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
116
|
Zhang J, Du Z, Pan S, Shi M, Li J, Yang C, Hu H, Qiao M, Chen D, Zhao X. Overcoming Multidrug Resistance by Codelivery of MDR1-Targeting siRNA and Doxorubicin Using EphA10-Mediated pH-Sensitive Lipoplexes: In Vitro and In Vivo Evaluation. ACS APPLIED MATERIALS & INTERFACES 2018; 10:21590-21600. [PMID: 29798663 DOI: 10.1021/acsami.8b01806] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The therapeutic efficacy of chemotherapy is dramatically hindered by multidrug resistance (MDR), which is induced by the overexpression of P-glycoprotein (P-gp). The codelivery of an antitumor drug and siRNA is an effective strategy recently applied in overcoming P-gp-related MDR. In this study, a multifunctional drug delivery system with both pH-sensitive feature and active targetability was designed, in which MDR1-siRNA and DOX were successfully loaded. The resulting carrier EphA10 antibody-conjugated pH-sensitive doxorubicin (DOX), MDR1-siRNA coloading lipoplexes (shortened as DOX + siRNA/ePL) with high serum stability had favorable physicochemical properties. DOX + siRNA/ePL exhibited an incremental cellular uptake, enhanced P-gp downregulation efficacy, as well as a better cell cytotoxicity in human breast cancer cell line/adriamycin drug-resistant (MCF-7/ADR) cells. The results of the intracellular colocalization study indicated that DOX + siRNA/ePL possessed the ability for pH-responsive rapid endosomal escape in a time-dependent characteristic. Meanwhile, the in vivo antitumor activities suggested that DOX + siRNA/ePL could prolong the circulation time as well as specifically accumulate in the tumor cells via receptor-mediated endocytosis after intravenous administration into the blood system. The histological study further demonstrated that DOX + siRNA/ePL could inhibit the proliferation, induce apoptosis effect, and downregulate the P-gp expression in vivo. Altogether, DOX + siRNA/ePL was expected to be a suitable codelivery system for overcoming the MDR effect.
Collapse
Affiliation(s)
- Jiulong Zhang
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Zhouqi Du
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Shuang Pan
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Menghao Shi
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Jie Li
- Mudanjiang Medical University , Tongxiang Street No. 3 , Mudanjiang , Heilongjiang 157011 , PR China
| | - Chunrong Yang
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Haiyang Hu
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Mingxi Qiao
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Dawei Chen
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| | - Xiuli Zhao
- School of Pharmacy , Shenyang Pharmaceutical University , 103 Wenhua Road , Shenyang , Liaoning 110016 , PR China
| |
Collapse
|
117
|
Zhou H, Lv S, Zhang D, Deng M, Zhang X, Tang Z, Chen X. A polypeptide based podophyllotoxin conjugate for the treatment of multi drug resistant breast cancer with enhanced efficiency and minimal toxicity. Acta Biomater 2018; 73:388-399. [PMID: 29694920 DOI: 10.1016/j.actbio.2018.04.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
Abstract
Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown significant activity against P-glycoprotein (P-gp) mediated multi drug resistant cancer cells. However, because of the poor aqueous solubility and high toxicity, PPT cannot be used in clinical cancer therapy. In order to enhance the efficiency and reduce side effect of PPT, a polypeptide based PPT conjugate PLG-g-mPEG-PPT was developed and used for the treatment of multi drug resistant breast cancer. The PLG-g-mPEG-PPT was prepared by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol) (PLG-g-mPEG) via ester bonds. The PPT conjugates self-assembled into nanoparticles with average sizes about 100 nm in aqueous solution. Western blotting assay showed that the PLG-g-mPEG-PPT could effectively inhibit the expression of P-gp in the multiple drug resistant MCF-7/ADR cells. In vitro cytotoxicity assay indicated that the resistance index (RI) values of PLG-g-mPEG-PPT on different drug-resistant cancer cell lines exhibited 57-270 folds reduction than of traditional microtubule inhibitor chemotherapeutic drug PTX or DTX. Hemolysis assay demonstrated that the conjugation greatly decreased the hemolytic activity of free PPT. Maximum tolerated dose (MTD) of PLG-g-mPEG-PPT increased greatly (13.3 folds) as compared to that of free PPT. In vivo study showed that the PLG-g-mPEG-PPT conjugate remarkably enhanced the antitumor efficacy against MCF-7/ADR xenograft tumors with a tumor suppression rate (TSR) of 82.5%, displayed significantly improved anticancer efficacy as compared to free PPT (TSR = 37.1%) with minimal toxicity when both of the two formulations were used in MTD. STATEMENT OF SIGNIFICANCE The development of multiple drug resistance (MDR) of cancer cells is the main cause of chemotherapy failure. The over-expression of P-glycoprotein (P-gp) has been recognized to be the most important cause of MDR in cancer. Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown strong activity against P-gp mediated multidrug resistant cancer cells by simultaneously inhibiting the over-expression of P-gp and the growth of cancer cells. However, PPT can not be used in clinical cancer treatment due to its poor aqueous solubility and high toxicity. Herein, we developed a polypeptide based PPT conjugate PLG-g-mPEG-PPT by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol). The PLG-g-mPEG-PPT shows significantly decreased hemolytic activity, greatly improved maximum tolerated dose and remarkably enhanced antitumor efficacy against MCF-7/ADR xenograft tumors as compared to free PPT.
Collapse
|
118
|
Synthesis and biological evaluation of 2,5-disubstituted furan derivatives as P-glycoprotein inhibitors for Doxorubicin resistance in MCF-7/ADR cell. Eur J Med Chem 2018; 151:546-556. [DOI: 10.1016/j.ejmech.2018.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/04/2018] [Accepted: 04/04/2018] [Indexed: 12/29/2022]
|
119
|
Ma P, Chen J, Bi X, Li Z, Gao X, Li H, Zhu H, Huang Y, Qi J, Zhang Y. Overcoming Multidrug Resistance through the GLUT1-Mediated and Enzyme-Triggered Mitochondrial Targeting Conjugate with Redox-Sensitive Paclitaxel Release. ACS APPLIED MATERIALS & INTERFACES 2018; 10:12351-12363. [PMID: 29569435 DOI: 10.1021/acsami.7b18437] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Multidrug resistance (MDR) is thought to be the major obstacle leading to the failure of paclitaxel (PTX) chemotherapy. To solve this problem, a glucose transporter-mediated and matrix metalloproteinase 2 (MMP2)-triggered mitochondrion-targeting conjugate [glucose-polyethylene glycol (PEG)-peptide-triphenylphosponium-polyamidoamine (PAMAM)-PTX] composed of a PAMAM dendrimer and enzymatic detachable glucose-PEG was constructed for mitochondrial delivery of PTX. The conjugate was characterized by a 30 nm sphere particle, MMP2-sensitive PEG outer layer detachment from PAMAM, and glutathione (GSH)-sensitive PTX release. It showed higher cellular uptake both in glucose transporter 1 (GLUT1) overexpressing MCF-7/MDR monolayer cell (2D) and multicellular tumor spheroids (3D). The subcellular location study showed that it could specifically accumulate in the mitochondria. Moreover, it exhibited higher cytotoxicity against MCF-7/MDR cells, which significantly reverse the MDR of MCF-7/MDR cells. The MDR reverse might be caused by reducing the ATP content through destroying the mitochondrial membrane as well as by down-regulating P-gp expression. In vivo imaging and tissue distribution indicated more conjugate accumulated in the tumor of the tumor-bearing mice model. Consequently, the conjugate showed better tumor inhibition rate and lower body weight loss, which demonstrated that it possessed high efficiency and low toxicity. This study provides glucose-mediated GLUT targeting, MMP2-responsive PEG detachment, triphenylphosponium-mediated mitochondria targeting, and a GSH-sensitive intracellular drug release conjugate that has the potential to be exploited for overcoming MDR of PTX.
Collapse
Affiliation(s)
- Pengkai Ma
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Jianhua Chen
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Xinning Bi
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Zhihui Li
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Xing Gao
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Hongpin Li
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Hongyu Zhu
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Yunfang Huang
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Jing Qi
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| | - Yujie Zhang
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Yangguang South Street , Beijing 102488 , China
| |
Collapse
|
120
|
Tikhomirov AS, Lin CY, Volodina YL, Dezhenkova LG, Tatarskiy VV, Schols D, Shtil AA, Kaur P, Chueh PJ, Shchekotikhin AE. New antitumor anthra[2,3-b]furan-3-carboxamides: Synthesis and structure-activity relationship. Eur J Med Chem 2018; 148:128-139. [DOI: 10.1016/j.ejmech.2018.02.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/06/2018] [Accepted: 02/09/2018] [Indexed: 11/16/2022]
|
121
|
Chen SR, Dai Y, Zhao J, Lin L, Wang Y, Wang Y. A Mechanistic Overview of Triptolide and Celastrol, Natural Products from Tripterygium wilfordii Hook F. Front Pharmacol 2018; 9:104. [PMID: 29491837 PMCID: PMC5817256 DOI: 10.3389/fphar.2018.00104] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/30/2018] [Indexed: 12/28/2022] Open
Abstract
Triptolide and celastrol are predominantly active natural products isolated from the medicinal plant Tripterygium wilfordii Hook F. These compounds exhibit similar pharmacological activities, including anti-cancer, anti-inflammation, anti-obesity, and anti-diabetic activities. Triptolide and celastrol also provide neuroprotection and prevent cardiovascular and metabolic diseases. However, toxicity restricts the further development of triptolide and celastrol. In this review, we comprehensively review therapeutic targets and mechanisms of action, and translational study of triptolide and celastrol. We systemically discuss the structure-activity-relationship of triptolide, celastrol, and their derivatives. Furthermore, we propose the use of structural derivatives, targeted therapy, and combination treatment as possible solutions to reduce toxicity and increase therapeutic window of these potent natural products from T. wilfordii Hook F.
Collapse
Affiliation(s)
- Shao-Ru Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yan Dai
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jing Zhao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
122
|
The dual-inhibitory effect of miR-338-5p on the multidrug resistance and cell growth of hepatocellular carcinoma. Signal Transduct Target Ther 2018. [PMID: 29527329 PMCID: PMC5837112 DOI: 10.1038/s41392-017-0003-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chemotherapeutic treatments against hepatocellular carcinoma (HCC) are necessary for both inoperable patients to improve prospects for survival and surgery patients to improve the outcome after surgical resection. However, multidrug resistance (MDR) is a major obstacle to obtaining desirable results. Currently, increasing the chemotherapy sensitivity of tumor cells or discovering novel tumor inhibitors is an effective therapeutic strategy to solve this issue. In the present study, we uncovered the dual-inhibitory effect of miR-338-5p: on the one hand, it could downregulate ABCB1 expression and sensitize HCC cells to doxorubicin and vinblastine by directly targeting the 3′-untranslated region (3′-UTR) of ABCB1, while, on the other hand, it could suppress the proliferation of HCC cells by directly targeting the 3′-UTR of EGFR and reducing EGFR expression. Since EGFR regulates ABCB1 levels, the indirect action of miR-338-5p in ABCB1 modulation was revealed, in which miR-338-5p inhibits ABCB1 expression by targeting the EGFR/ERK1/2 signaling pathway. These data indicate that the miR-338-5p/EGFR/ABCB1 regulatory loop plays a critical role in HCC, and a negative correlation between miR-338-5p and EGFR or ABCB1 was also detected in HCC clinical samples. In conclusion, these findings reveal a critical role for miR-338-5p in the regulation of MDR and proliferation of HCC, suggesting the potential therapeutic implications of miR-338-5p in HCC treatment. A small RNA molecule inhibits the growth of liver cancer cells while also making the cells sensitive to the anti-cancer drugs. These twin effects of the natural microRNA miR-338-5p were discovered by researchers in China, led by Chunzhu Li and Jin Ren at the Center for Drug Safety Evaluation and Research in Shanghai. MicroRNAs control gene activity by interacting with the messenger RNA copies of genes that guide synthesis of the proteins the genes encode. The research identified a gene whose expression miR-338-5p inhibits to restrict the growth of hepatocellular carcinoma – the most common form of liver cancer. This is also one of the most drug-resistant forms of liver cancer. A different gene whose activity miR-338-5p controls to sensitize cells to chemotherapeutic drugs was also identified. Using miR-338-5p to treat liver cancer warrants further investigation.
Collapse
|
123
|
A vascular disrupting agent overcomes tumor multidrug resistance by skewing macrophage polarity toward the M1 phenotype. Cancer Lett 2018; 418:239-249. [PMID: 29337108 DOI: 10.1016/j.canlet.2018.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/01/2018] [Accepted: 01/08/2018] [Indexed: 12/12/2022]
Abstract
Multidrug resistance (MDR) mediated by ATP-binding cassette (ABC) transporters is the major obstacle for chemotherapeutic success. Although attempts have been made to circumvent ABC transporter-mediated MDR in past decades, there is still no effective agent in clinic. Here, we identified a vascular disrupting agent, Z-GP-DAVLBH, that significantly inhibited the growth of multidrug-resistant human hepatoma HepG2/ADM and human breast cancer MCF-7/ADR tumor xenografts, although these cells were insensitive to Z-GP-DAVLBH in vitro. Z-GP-DAVLBH increased the secretion of granulocyte-macrophage colony-stimulating factor in tumor tissues and serum of tumor-bearing mice to skew tumor-associated macrophages from the pro-tumor M2 phenotype to the antitumor M1 phenotype, thereby contributing to the induction of HepG2/ADM and MCF-7/ADR cell apoptosis. Our findings shed new light on the underlying mechanisms of VDAs in the treatment of drug-resistant tumors and provide strong evidence that Z-GP-DAVLBH should be a promising agent for overcoming MDR.
Collapse
|
124
|
Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS One 2017. [PMID: 29121120 DOI: 10.1371/journal.pone.0187925.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cancer is considered one of the primary diseases that cause morbidity and mortality in millions of people worldwide and due to its prevalence, there is undoubtedly an unmet need to discover novel anticancer drugs. However, the traditional process of drug discovery and development is lengthy and expensive, so the application of in silico techniques and optimization algorithms in drug discovery projects can provide a solution, saving time and costs. A set of 617 approved anticancer drugs, constituting the active domain, and a set of 2,892 natural products, constituting the inactive domain, were employed to build predictive models and to index natural products for their anticancer bioactivity. Using the iterative stochastic elimination optimization technique, we obtained a highly discriminative and robust model, with an area under the curve of 0.95. Twelve natural products that scored highly as potential anticancer drug candidates are disclosed. Searching the scientific literature revealed that few of those molecules (Neoechinulin, Colchicine, and Piperolactam) have already been experimentally screened for their anticancer activity and found active. The other phytochemicals await evaluation for their anticancerous activity in wet lab.
Collapse
|
125
|
Rayan A, Raiyn J, Falah M. Nature is the best source of anticancer drugs: Indexing natural products for their anticancer bioactivity. PLoS One 2017; 12:e0187925. [PMID: 29121120 PMCID: PMC5679595 DOI: 10.1371/journal.pone.0187925] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/27/2017] [Indexed: 01/10/2023] Open
Abstract
Cancer is considered one of the primary diseases that cause morbidity and mortality in millions of people worldwide and due to its prevalence, there is undoubtedly an unmet need to discover novel anticancer drugs. However, the traditional process of drug discovery and development is lengthy and expensive, so the application of in silico techniques and optimization algorithms in drug discovery projects can provide a solution, saving time and costs. A set of 617 approved anticancer drugs, constituting the active domain, and a set of 2,892 natural products, constituting the inactive domain, were employed to build predictive models and to index natural products for their anticancer bioactivity. Using the iterative stochastic elimination optimization technique, we obtained a highly discriminative and robust model, with an area under the curve of 0.95. Twelve natural products that scored highly as potential anticancer drug candidates are disclosed. Searching the scientific literature revealed that few of those molecules (Neoechinulin, Colchicine, and Piperolactam) have already been experimentally screened for their anticancer activity and found active. The other phytochemicals await evaluation for their anticancerous activity in wet lab.
Collapse
Affiliation(s)
- Anwar Rayan
- Drug Discovery Informatics Lab, QRC - Qasemi Research Center, Al-Qasemi Academic College, Baka EL-Garbiah, Israel
- Drug Discovery and Development Laboratory, Institute of Applied Research - The Galilee Society, Shefa-Amr, Israel
- * E-mail: (AR); (MF)
| | - Jamal Raiyn
- Drug Discovery Informatics Lab, QRC - Qasemi Research Center, Al-Qasemi Academic College, Baka EL-Garbiah, Israel
| | - Mizied Falah
- Faculty of Medicine in the Galilee, Bar-Ilan University, Ramat Gan, Tel Aviv, Israel
- Galilee Medical Center, Nahariya, Israel
- * E-mail: (AR); (MF)
| |
Collapse
|
126
|
Design and synthesis of novel nitrogen mustard-evodiamine hybrids with selective antiproliferative activity. Bioorg Med Chem Lett 2017; 27:4989-4993. [DOI: 10.1016/j.bmcl.2017.10.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/26/2017] [Accepted: 10/07/2017] [Indexed: 11/22/2022]
|
127
|
Liu CM, Kao CL, Tseng YT, Lo YC, Chen CY. Ginger Phytochemicals Inhibit Cell Growth and Modulate Drug Resistance Factors in Docetaxel Resistant Prostate Cancer Cell. Molecules 2017; 22:E1477. [PMID: 28872603 PMCID: PMC6151784 DOI: 10.3390/molecules22091477] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/02/2017] [Indexed: 12/12/2022] Open
Abstract
Ginger has many bioactive compounds with pharmacological activities. However, few studies are known about these bioactive compounds activity in chemoresistant cells. The aim of the present study was to investigate the anticancer properties of ginger phytochemicals in docetaxel-resistant human prostate cancer cells in vitro. In this study, we isolated 6-gingerol, 10-gingerol, 4-shogaol, 6-shogaol, 10-shogaol, and 6-dehydrogingerdione from ginger. Further, the antiproliferation activity of these compounds was examined in docetaxel-resistant (PC3R) and sensitive (PC3) human prostate cancer cell lines. 6-gingerol, 10-gingerol, 6-shogaol, and 10-shogaol at the concentration of 100 μM significantly inhibited the proliferation in PC3R but 6-gingerol, 6-shogaol, and 10-shogaol displayed similar activity in PC3. The protein expression of multidrug resistance associated protein 1 (MRP1) and glutathione-S-transferase (GSTπ) is higher in PC3R than in PC3. In summary, we isolated the bioactive compounds from ginger. Our results showed that 6-gingerol, 10-gingerol, 6-shogaol, and 10-shogaol inhibit the proliferation of PC3R cells through the downregulation of MRP1 and GSTπ protein expression.
Collapse
Affiliation(s)
- Chi-Ming Liu
- Department of Nursing, Tzu Hui Institute of Technology, Pingtung County 92641, Taiwan.
| | - Chiu-Li Kao
- Department of Nursing, Tzu Hui Institute of Technology, Pingtung County 92641, Taiwan.
| | - Yu-Ting Tseng
- Department of Pharmacology, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yi-Ching Lo
- Department of Pharmacology, School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chung-Yi Chen
- School of Medical and Health Sciences, Fooyin University, Ta-Liao District, Kaohsiung 83102, Taiwan.
| |
Collapse
|