1
|
Xue J, Li Q, Wang Y, Yin R, Zhang J. Insight into the structure, oligomerization, and the role in drug resistance of human UDP-glucuronosyltransferases. Arch Toxicol 2025; 99:1153-1165. [PMID: 39812829 DOI: 10.1007/s00204-024-03929-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025]
Abstract
Human UDP-glucuronosyltransferases (UGTs) are pivotal phase II metabolic enzymes facilitating the transfer of glucuronic acid from UDP-glucuronic acid (UDPGA) to various substrates. UGTs are classic type I transmembrane glycoproteins, mainly localized in the endoplasmic reticulum (ER) membrane. This review comprehensively explores UGTs, encompassing gene expression, functional characteristics, substrate specificity, and metabolic mechanisms. A recent analysis of C-terminal structures, compared with original data, underscores the pivotal role of α3, α4, and β4 functional domains in selectively recognizing diverse glycosyl donors. Accumulating evidence suggests that UGTs function as homo- and heterodimers, with oligomers likely stabilizing UGTs and modulating their activity. The review sheds light on the implications of UGT oligomerization on substrate glucuronidation and the interplay between protein-protein interaction and glucuronidation activity. UGT-mediated drug resistance, often underestimated, emerges as a clinically relevant form of chemical resistance, with delineated outcomes in tumors and other diseases. This review provides a multifaceted exploration of the physiological significance of UGTs, spanning genetics, proteins, oligomerization, drug resistance, and more, offering insights into their metabolic mechanisms. Understanding interactions between UGT isoforms is crucial for predicting drug-drug interactions, preventing drug toxicity, and enabling precision treatment.
Collapse
Affiliation(s)
- Jia Xue
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiuyi Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruxi Yin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Zhao S, Wang L, Huang X, Xiao Y, Li M, Huang X, Chen X, Li S, Xie J, Liu P, Wang YD, Chen WD. Design, Synthesis, and Biological Evaluation of Covalently Mucoadhesive Derivatives as Nonsystemic Intestine-Targeted TGR5 Agonists. J Med Chem 2024; 67:17701-17712. [PMID: 39321318 DOI: 10.1021/acs.jmedchem.4c01637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Takeda G-protein-coupled receptor 5 (TGR5) is considered a promising therapeutic target for treating type 2 diabetes mellitus (T2DM), obesity, and other metabolism-related diseases. Although many TGR5 agonists have been identified, they might cause some side effects in the gallbladder and the heart. To reduce these side effects and improve glucose-lowering capability, we first designed and synthesized a series of 4-phenoxynicotinamide intestine-targeted TGR5 agonist derivatives containing maleimides in the side chains with different linker lengths. All of the target compounds demonstrated significant TGR5 agonistic activity, among which compound 22b displayed the best TGR5 agonistic activity. Additionally, compound 22b displayed low Caco-2 cell permeability and strong mucoadhesion to mucin and the rat intestine. In C57BL/6J, diet-induced obese, and db/db mice, compound 22b demonstrated a robust and prolonged hypoglycemic effect along with an acceptable safety profile.
Collapse
Affiliation(s)
- Shizhen Zhao
- The First Affiliated Hospital of Henan University, Kaifeng 475000, China
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China
| | - Le Wang
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China
| | - Xiaotong Huang
- The First Affiliated Hospital of Henan University, Kaifeng 475000, China
| | - Yali Xiao
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China
| | - Mengqi Li
- The First Affiliated Hospital of Henan University, Kaifeng 475000, China
| | - Xueyuan Huang
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China
| | - Xueyu Chen
- The First Affiliated Hospital of Henan University, Kaifeng 475000, China
| | - Shengjie Li
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China
| | - Jing Xie
- The First Affiliated Hospital of Henan University, Kaifeng 475000, China
| | - Peng Liu
- Hebi Key Laboratory of Cardiovascular Diseases, Hebi Key Laboratory of Energy Metabolism, People's Hospital of Hebi, Henan University, Kaifeng 475000, China
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wei-Dong Chen
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475000, China
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot 010110, China
| |
Collapse
|
3
|
Qin YT, Liu X, An JX, Chen Z, Niu MT, Yan X, Li QR, Rao ZY, Zhang XZ. Oral Saccharomyces cerevisiae-Guided Enzyme Prodrug Therapy Combined with Immunotherapy for the Treatment of Orthotopic Colorectal Cancer. ACS NANO 2024; 18:23497-23507. [PMID: 39146387 DOI: 10.1021/acsnano.4c07115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Colorectal cancer (CRC) is a major global health concern, and the development of effective treatment strategies is crucial. Enzyme prodrug therapy (EPT) shows promise in combating tumors but faces challenges in achieving sustained expression of therapeutic enzymes and optimal biological distribution. To address these issues, a fungi-triggered in situ chemotherapeutics generator (named as SC@CS@5-FC) was constructed via oral delivery of a prodrug (5-fluorocytosine, 5-FC) for the treatment of orthotopic colorectal tumor. When SC@CS@5-FC targets the tumor through tropism by Saccharomyces cerevisiae (SC), the chemotherapeutic generator could be degraded under abundant hyaluronidase (HAase) in the tumor microenvironment by an enzyme-responsive gate to release prodrug (5-FC). And nontoxic 5-FC was catalyzed to toxic chemotherapy drug 5-fluorouracil (5-FU) by cytosine deaminase (CD) of SC. Meanwhile, SC and zinc-coordinated chitosan nanoparticles could be used as immune adjuvants to activate antigen-presenting cells and further enhance the therapeutic effect. Our results demonstrated that SC@CS@5-FC could effectively inhibit tumor growth and prolong mouse survival in an orthotopic colorectal cancer model. This work utilizes living SC as a dynamotor and positioning system for the chemotherapeutic generator SC@CS@5-FC, providing a strategy for oral enzyme prodrug therapy for the treatment of orthotopic colorectal.
Collapse
Affiliation(s)
- You-Teng Qin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xinhua Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Jia-Xin An
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Zhu Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Mei-Ting Niu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xiao Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Qian-Ru Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Zhi-Yong Rao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
4
|
Zhang YJ, Liang JX, Xu YS, Liu YX, Cui Y, Qiao ZY, Wang H. Covalent drugs based on small molecules and peptides for disease theranostics. Biomater Sci 2024; 12:564-580. [PMID: 37975197 DOI: 10.1039/d3bm01138k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Biomacromolecules, such as proteins, nucleic acids and polysaccharides, are widely distributed in the human body, and some of them have been recognized as the targets of drugs for disease theranostics. Drugs typically act on targets in two ways: non-covalent bond and covalent bond. Non-covalent bond-based drugs have some disadvantages, such as structural instability and environmental sensitivity. Covalent interactions between drugs and targets have a longer action time, higher affinity and controllability than non-covalent interactions of conventional drugs. With the development of artificial intelligence, covalent drugs have received more attention and have been developed rapidly in pharmaceutical research in recent years. From the perspective of covalent drugs, this review summarizes the design methods and the effects of covalent drugs. Finally, we discuss the application of covalent peptide drugs and expect to provide a new reference for cancer treatment.
Collapse
Affiliation(s)
- Ying-Jin Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P.R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P.R. China.
| | - Jian-Xiao Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P.R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, P.R. China
| | - Yin-Sheng Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P.R. China.
| | - Yi-Xuan Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P.R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, P.R. China
| | - Yingying Cui
- Department of Food and Drug, Laiwu Vocational and Technical, College, Jinan, China
| | - Zeng-Ying Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P.R. China.
| | - Hao Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P.R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P.R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, P.R. China
| |
Collapse
|
5
|
Verma H, Narendra G, Raju B, Singh PK, Silakari O. Dihydropyrimidine Dehydrogenase-Mediated Resistance to 5-Fluorouracil: Mechanistic Investigation and Solution. ACS Pharmacol Transl Sci 2022; 5:1017-1033. [PMID: 36407958 PMCID: PMC9667542 DOI: 10.1021/acsptsci.2c00117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Indexed: 11/29/2022]
Abstract
5-Fluorouracil (5-FU) is one of the most widely used chemotherapeutics for the treatment of cancers associated with the aerodigestive tract, breast, and colorectal system. The efficacy of 5-FU is majorly affected by dihydropyrimidine dehydrogenase (DPD) as it degrades more than 80% of administered 5-FU into an inactive metabolite, dihydrofluorouracil. Herein we discuss the molecular mechanism of this inactivation by analyzing the interaction pattern and electrostatic complementarity of the DPD-5-FU complex. The basis of DPD overexpression in cancer cell lines due to significantly distinct levels of the miRNAs (miR-134, miR-27b, and miR-27a) compared to normal cells has also been outlined. Additionally, some kinases including sphingosine kinase 2 (SphK2) have been reported to correlate with DPD expression. Currently, to address this problem various strategies are reported in the literature, including 5-FU analogues (bypass the DPD-mediated inactivation), DPD downregulators (regulate the DPD expression levels in tumors), inhibitors (as promising adjuvants), and formulation development loaded with 5-FU (liposomes, nanoparticles, nanogels, etc.), which are briefly discussed in this Review.
Collapse
Affiliation(s)
- Himanshu Verma
- Molecular
Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab147002, India
| | - Gera Narendra
- Molecular
Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab147002, India
| | - Baddipadige Raju
- Molecular
Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab147002, India
| | - Pankaj Kumar Singh
- Integrative
Physiology and Pharmacology, Institute of Biomedicine, Faculty of
Medicine, University of Turku, FI-20520Turku, Finland
| | - Om Silakari
- Molecular
Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab147002, India
| |
Collapse
|
6
|
Analytical and bioanalytical HPLC method for simultaneous estimation of 5-fluorouracil and sonidegib. Bioanalysis 2021; 14:29-45. [PMID: 34786955 DOI: 10.4155/bio-2021-0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: To develop a new sensitive RP-HPLC method for simultaneous estimation of 5-fluorouracil (5-FU) and sonidegib (SDG). Materials & methods: Analytical and bioanalytical methods for simultaneous quantification of 5-FU and SDG in bulk, nanoformulations and in rat plasma were developed and validated using a gradient elution technique. Results: Separation of the analytes was effected on a Luna® C18 LC column using a mobile mixture comprising acetonitrile and acidified water. 5-FU and SDG were extracted from plasma matrix using liquid-liquid extraction. The applicability of the method was verified through single-dose oral pharmacokinetic study in Wistar rats. Conclusion: The developed methods allow a specific, sensitive and steady analytical procedure for the simultaneous estimation of 5-FU and SDG in nanoformulations and biological matrix.
Collapse
|
7
|
Liu L, Tian C, Dong B, Xia M, Cai Y, Hu R, Chu X. Models to evaluate the barrier properties of mucus during drug diffusion. Int J Pharm 2021; 599:120415. [PMID: 33647411 DOI: 10.1016/j.ijpharm.2021.120415] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
Mucus is widely disseminated in the nasal cavity, oral cavity, respiratory tract, eyes, gastrointestinal tract, and reproductive tract to prevent the invasion of pathogenic bacteria and toxins. The mucus layer through its continuous secretion can prevent the passage of macromolecular substances such as pathogenic bacteria and toxins, thereby reducing the occurrence of inflammation. Without a doubt, mucus also hinders oral absorption. The physiological and biochemical properties of intestinal mucus and the different types of mucus barrier models need to be predominated. To find ways to increase the bioavailability of drugs in the future, this article summarizes mucus composition, barrier properties, mucus models, and mucoadhesive/mucopenetrating particles to highlight the information they can afford. Collectively, the review seeks to provide a state-of-the-art roadmap for researchers who must contend with this critical barrier to drug delivery.
Collapse
Affiliation(s)
- Liu Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Chunling Tian
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Baoqi Dong
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Mengqiu Xia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Ye Cai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Rongfeng Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
8
|
Pakdel M, Raissi H, Hosseini ST. Evaluation the synergistic antitumor effect of methotrexate-camptothecin codelivery prodrug from self-assembly process to acid-catalyzed both drugs release: A comprehensive theoretical study. J Comput Chem 2020; 41:1486-1496. [PMID: 32190916 DOI: 10.1002/jcc.26192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/10/2019] [Accepted: 03/09/2020] [Indexed: 12/19/2022]
Abstract
Therapeutic efficiency of amphiphilic methotrexate-camptothecin (MTX-CPT) prodrug compared to free drug mixture (MTX/CPT) has been investigated using all-atom molecular dynamics simulation and first principles density functional theory calculations. This comparison revealed that MTX-CPT prodrug tends to form spherical self-assembled nanoparticle (NP), while free MTX/CPT mixture forms rod-shape NP. These observations are attributed to a structural defect in the MTX-CPT prodrug and solvation free energies of MTX, CPT and MTX-CPT molecules. The results provided evidence that noncovalent interactions (NCIs) among the pharmaceutical drugs play a very important role in anticancer agents aggregation process, leading to enhanced stability of the self-assembled NPs. It is found that the stability of MTX-CPT self-assembled NP is greater than the MTX/CPT NP due to the synergistic effect of hydrogen bonding between monomers and solvent (water). Moreover, the noncatalyzed as well as catalyzed hydrolysis reactions of MTX-CPT prodrug are theoretically studied at the PCM(water)//M06-2X/6-31G(d,p) computational level to shed additional light on the role of acidic condition in tumor tissues. We found that the ester hydrolysis in mild acidic solutions is a concerted reaction. In an agreement between theory and experiment, we also confirmed that the activation energies of the catalyzed-hydrolysis steps are much lower than the activation energies of the corresponding steps in the noncatalyzed reaction. Thus, the MTX-CPT prodrug reveals very promising properties as a pH-controlled drug delivery system.
Collapse
Affiliation(s)
- Majid Pakdel
- Department of Chemistry, Faculty of Science, University of Birjand, Birjand, Iran
| | - Heidar Raissi
- Department of Chemistry, Faculty of Science, University of Birjand, Birjand, Iran
| | - Seyede T Hosseini
- Department of Chemistry, Faculty of Science, University of Birjand, Birjand, Iran
| |
Collapse
|
9
|
Pasban S, Raissi H, Pakdel M, Farzad F. Enhance the efficiency of 5-fluorouracil targeted delivery by using a prodrug approach as a novel strategy for prolonged circulation time and improved permeation. Int J Pharm 2019; 568:118491. [PMID: 31276765 DOI: 10.1016/j.ijpharm.2019.118491] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/18/2019] [Accepted: 06/30/2019] [Indexed: 11/25/2022]
Abstract
Due to the toxicity and resistance to treatment with anticancer drugs, various methods are used to improve their efficacy in cancer treatment. In this present study, in order to overcome the limitation of 5-Fluorouracil (5-FU), prodrug strategy has been pursued with using density functional theory (DFT) and molecular dynamics simulation (MDs). The main objective of this study is to examine the mechanisms of drug release from its prodrug form by using the intrinsic reaction coordinate (IRC) calculations. The reaction mechanisms of 5-FU prodrug (EMC-5-FU) in the presence of lactic acid (LA) and water molecule were theoretically studied. The IRC calculations were carried out at the M06-2X/6-311G** level in the aqueous phase through the mechanism of ester hydrolysis to obtain energies, the geometry optimization of all stationary points along the potential energy surfaces (PES), and also to determine the harmonic vibrational frequencies. The results herein presented suggest that three reaction pathways and transition states TS1 to TS2 are involved along the calculated potential energy surface. We found that the drug molecule is released in the third step and this occurs by separation CH2O group in the presence of water molecule with the highest energy barrier about 25.9 kcal/mol. Since the carbon nanotubes (CNTs) can act as drug delivery vehicles and deliver anticancer drugs directly to the target cells. Therefore in DFT section, the interaction mechanism of CNTs with 5-FU prodrug is studied by means of DFT method. The atoms in molecules (AIM) and the non-covalent interactions (NCI) between the CNTs and prodrug are used in order to examine the strength and type of interaction between them. The result of negative binding energy values of CNT-prodrug interaction show the stability of these complexes. Our theoretical results show that the more favorable interaction occurs when the prodrug is located inside the carbon nanotube. Furthermore, for design and development of intracellular drug delivery systems, steered molecular dynamics (SMD) simulations was used to investigate the possibility of encapsulated prodrug-CNT penetration through a (1-palmitoyl-2-oleoyl phosphatidylcholine) POPC lipid bilayer. For this purpose, the forces of penetration and the free energies of rupture of POPC bilayer with a Prodrug-CNT were studied. Our simulation results show that encapsulated prodrug-carbon nanotube does not permanently destroy the POPC membrane structure.
Collapse
Affiliation(s)
- Samaneh Pasban
- Chemistry Department, University of Birjand, Birjand, Iran.
| | - Heidar Raissi
- Chemistry Department, University of Birjand, Birjand, Iran
| | - Majid Pakdel
- Chemistry Department, University of Birjand, Birjand, Iran
| | | |
Collapse
|
10
|
Verma H, Singh Bahia M, Choudhary S, Kumar Singh P, Silakari O. Drug metabolizing enzymes-associated chemo resistance and strategies to overcome it. Drug Metab Rev 2019; 51:196-223. [DOI: 10.1080/03602532.2019.1632886] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Himanshu Verma
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | - Shalki Choudhary
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Pankaj Kumar Singh
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
11
|
Zhang R, Song XQ, Liu RP, Ma ZY, Xu JY. Fuplatin: An Efficient and Low-Toxic Dual-Prodrug. J Med Chem 2019; 62:4543-4554. [PMID: 31002510 DOI: 10.1021/acs.jmedchem.9b00128] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As FDA-approved chemotherapeutic agents, cisplatin, oxaliplatin, and 5-fluorouracil are widely used in clinic but limited by severe side-effects. To ameliorate their respective defects, a series of "dual-prodrug" by linking oxoplatin and 5-FU were designed and synthesized. The assembled compounds 10-17, named Fuplatin, exhibited much higher cytotoxicity against the tested cancer cells while lower cytotoxicity toward the human normal lung cells than free drugs or their combinations. Among them, 14 enhanced cellular accumulation with 62- and 825-fold amount of oxaliplatin and 8 at 9 h, respectively, significantly induced DNA damage and cell apoptosis, and inhibited migration and invasion in HCT-116 cells. Compound 14 arrested the cell cycle at S and G2 phases and up-regulated thymidylate synthase and p53, consistent with the results of the combination, suggesting 14 adopted a collaborative mode of 5-FU and oxaliplatin to kill cancer cells. In vivo, compound 14 showed high antitumor effect and no observable toxicity in NOD/SCID mice bearing HCT-116 tumors.
Collapse
Affiliation(s)
- Ran Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy , Tianjin Medical University , Tianjin 300070 , China
| | - Xue-Qing Song
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy , Tianjin Medical University , Tianjin 300070 , China
| | - Rui-Ping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy , Tianjin Medical University , Tianjin 300070 , China
| | - Zhong-Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy , Tianjin Medical University , Tianjin 300070 , China
| | - Jing-Yuan Xu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy , Tianjin Medical University , Tianjin 300070 , China
| |
Collapse
|
12
|
Machine Learning Approach for Determining the Formation of β-Lactam Antibiotic Complexes with Cyclodextrins Using Multispectral Analysis. Molecules 2019; 24:molecules24040743. [PMID: 30791432 PMCID: PMC6413071 DOI: 10.3390/molecules24040743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/15/2019] [Accepted: 02/17/2019] [Indexed: 12/18/2022] Open
Abstract
The problem of determining the formation of complexes of β-lactam antibiotics with cyclodextrins (CDs) and the interactions involved in this process were addressed by machine learning on multispectral images. Complexes of β-lactam antibiotics, including cefuroxime axetil, cefetamet pivoxil, and pivampicillin, as well as CDs, including αCD, βCD, γCD, hydroxypropyl-αCD, methyl-βCD, hydroxypropyl-βCD, and hydroxypropyl-γCD, were prepared in all combinations. Thermograms confirming the formation of cyclodextrin complexes were obtained using differential scanning calorimetry. Transmission Fourier-transform infrared (tFTIR) and complementary attenuated total reflectance FTIR (ATR) coupled with machine learning were techniques chosen as a nondestructive alternative. The machine learning algorithm was used to determine the formation of complexes in samples using solely their tFTIR and ATR spectra at the prediction stage. Parameterized method 7 (PM7) was used to support the analysis by molecular modeling of the complexes. The model developed through machine learning properly distinguished samples with formed complexes form noncomplexed samples with a cross-validation accuracy of 90.4%. Analysis of the contribution of spectral bands to the model indicated interactions of ester groups of β-lactam antibiotics with CDs, as well as some interactions of cephem ring in cefetamet pivoxil and penam moiety in pivampicillin. Molecular modeling with PM7 helped to explain experimental results and allowed to propose possible binding modes.
Collapse
|