1
|
Anandi L, Garcia J, Ros M, Janská L, Liu J, Carmona-Fontaine C. Direct visualization of emergent metastatic features within an ex vivo model of the tumor microenvironment. Life Sci Alliance 2025; 8:e202403053. [PMID: 39419548 PMCID: PMC11487089 DOI: 10.26508/lsa.202403053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemic conditions such as hypoxia and nutrient starvation, together with interactions with stromal cells, are critical drivers of metastasis. These conditions arise deep within tumor tissues, and thus, observing nascent metastases is exceedingly challenging. We thus developed the 3MIC-an ex vivo model of the tumor microenvironment-to study the emergence of metastatic features in tumor cells in a 3-dimensional (3D) context. Here, tumor cells spontaneously create ischemic-like conditions, allowing us to study how tumor spheroids migrate, invade, and interact with stromal cells under different metabolic conditions. Consistent with previous data, we show that ischemia increases cell migration and invasion, but the 3MIC allowed us to directly observe and perturb cells while they acquire these pro-metastatic features. Interestingly, our results indicate that medium acidification is one of the strongest pro-metastatic cues and also illustrate using the 3MIC to test anti-metastatic drugs on cells experiencing different metabolic conditions. Overall, the 3MIC can help dissecting the complexity of the tumor microenvironment for the direct observation and perturbation of tumor cells during the early metastatic process.
Collapse
Affiliation(s)
- Libi Anandi
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Jeremy Garcia
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Manon Ros
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Libuše Janská
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Josephine Liu
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Carlos Carmona-Fontaine
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| |
Collapse
|
2
|
Fu XJ, Li N, Wu J, Wang ZY, Liu RR, Niu JB, Taleb M, Yuan S, Liu HM, Song J, Zhang SY. Discovery of novel pyrazolo[1,5-a]pyrimidine derivatives as potent reversal agents against ABCB1-mediated multidrug resistance. Eur J Med Chem 2024; 277:116761. [PMID: 39151276 DOI: 10.1016/j.ejmech.2024.116761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
The P-glycoprotein (ABCB1)-mediated multidrug resistance (MDR) has emerged as a significant impediment to the efficacy of cancer chemotherapy in clinical therapy, which could promote the development of effective agents for MDR reversal. In this work, we reported the exploration of novel pyrazolo [1,5-a]pyrimidine derivatives as potent reversal agents capable of enhancing the sensitivity of ABCB1-mediated MDR MCF-7/ADR cells to paclitaxel (PTX). Among them, compound 16q remarkably increased the sensitivity of MCF-7/ADR cells to PTX at 5 μM (IC50 = 27.00 nM, RF = 247.40) and 10 μM (IC50 = 10.07 nM, RF = 663.44). Compound 16q could effectively bind and stabilize ABCB1, and does not affect the expression and subcellular localization of ABCB1 in MCF-7/ADR cells. Compound 16q inhibited the function of ABCB1, thereby increasing PTX accumulation, and interrupting the accumulation and efflux of the ABCB1-mediated Rh123, thus resulting in exhibiting good reversal effects. In addition, due to the potent reversal effects of compound 16q, the abilities of PTX to inhibit tubulin depolymerization, and induce cell cycle arrest and apoptosis in MCF-7/ADR cells under low-dose conditions were restored. These results indicate that compound 16q might be a promising potent reversal agent capable of revising ABCB1-mediated MDR, and pyrazolo [1,5-a]pyrimidine might represent a novel scaffold for the discovery of new ABCB1-mediated MDR reversal agents.
Collapse
Affiliation(s)
- Xiang-Jing Fu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Na Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Ji Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Zi-Yue Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Rui-Rui Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mohammad Taleb
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China; State Key Laboratory of Esophageal Cancer Prevention &Treatment, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Xia HY, Zhao Y, Meng RY, Chen AZ, Wang SB, Kankala RK. Transition Metal (Molybdenum)-Doped Drug-like Conformational Nanoarchitectonics with Altered Valence States (Mn 2+/Mn 4+ and Mo 5+/Mo 6+) for Augmented Cancer Theranostics. Mol Pharm 2024; 21:5864-5879. [PMID: 39378327 DOI: 10.1021/acs.molpharmaceut.4c00896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Despite the advancements in cancer therapy, delivering active pharmaceutical ingredients (APIs) using nanoparticles remains challenging due to the failed conveyance of the required drug payload, poor targeting ability, and poor biodistribution, hampering their clinical translation. Recently, the appropriate design of materials with intrinsic therapeutic functionalities has garnered enormous interest in the development of various intelligent therapeutic nanoplatforms. In this study, we demonstrate the fabrication of transition metal (molybdenum, Mo)-doped manganese dioxide (MnO2) nanoarchitectures, exhibiting diagnostic (magnetic resonance imaging, MRI) and therapeutic (chemodynamic therapy, CDT) functionalities. The facile hydrothermal approach-assisted Mo-doped MnO2 flower-like nanostructures offered tailorable morphologies in altered dimensions, precise therapeutic effects, exceptional biocompatibility, and biodegradability in the tumor microenvironment. The resultant defects due to doped Mo species exhibited peroxidase and oxidase activities, improving glutathione (GSH) oxidation. The two sets of variable valence metal ion pairs (Mn2+/Mn4+ and Mo5+/Mo6+) and their interplay could substantially improve the Fenton-like reaction and generate toxic hydroxyl radicals (•OH), thus achieving CDT-assisted antitumor effects. As inherent T1-MRI agents, these MnO2 nanoparticles displayed excellent MRI efficacy in vitro. Together, we believe that these conformational Mo-doped MnO2 nanoarchitectures with two pairs of variable valence states could potentiate drugless therapy in pharmaceutics.
Collapse
Affiliation(s)
- Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ying Zhao
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ruo-Yin Meng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| |
Collapse
|
4
|
Kölz C, Gaugaz FZ, Handin N, Schaeffeler E, Tremmel R, Winter S, Klein K, Zanger UM, Artursson P, Schwab M, Nies AT. In silico and biological analyses of missense variants of the human biliary efflux transporter ABCC2: effects of novel rare missense variants. Br J Pharmacol 2024; 181:4593-4609. [PMID: 39096023 DOI: 10.1111/bph.16508] [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: 03/11/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND AND PURPOSE The ATP-dependent biliary efflux transporter ABCC2, also known as multidrug resistance protein 2 (MRP2), is essential for the cellular disposition and detoxification of various xenobiotics including drugs as well as endogenous metabolites. Common functionally relevant ABCC2 genetic variants significantly alter drug responses and contribute to side effects. The aim of this study was to determine functional consequences of rare variants identified in subjects with European ancestry using in silico tools and in vitro analyses. EXPERIMENTAL APPROACH Targeted next-generation sequencing of the ABCC2 gene was used to identify novel variants in European subjects (n = 143). Twenty-six in silico tools were used to predict functional consequences. For biological validation, transport assays were carried out with membrane vesicles prepared from cell lines overexpressing the newly identified ABCC2 variants and estradiol β-glucuronide and carboxydichlorofluorescein as the substrates. KEY RESULTS Three novel rare ABCC2 missense variants were identified (W227R, K402T, V489F). Twenty-five in silico tools predicted W227R as damaging and one as potentially damaging. Prediction of functional consequences was not possible for K402T and V489F and for the common linked variants V1188E/C1515Y. Characterisation in vitro showed increased function of W227R, V489F and V1188E/C1515Y for both substrates, whereas K402T function was only increased for carboxydichlorofluorescein. CONCLUSION AND IMPLICATIONS In silico tools were unable to accurately predict the substrate-dependent increase in function of ABCC2 missense variants. In vitro biological studies are required to accurately determine functional activity to avoid misleading consequences for drug therapy.
Collapse
Affiliation(s)
- Charlotte Kölz
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | | | - Niklas Handin
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Roman Tremmel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
| | - Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Ding L, Guo H, Zhang J, Zheng M, Zhang W, Wang L, Du Q, Zhou C, Xu Y, Wu H, He Q, Yang B. Zosuquidar Promotes Antitumor Immunity by Inducing Autophagic Degradation of PD-L1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400340. [PMID: 39229920 DOI: 10.1002/advs.202400340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/30/2024] [Indexed: 09/05/2024]
Abstract
The intracellular distribution and transportation process are essential for maintaining PD-L1 (programmed death-ligand 1) expression, and intervening in this cellular process may provide promising therapeutic strategies. Here, through a cell-based high content screening, it is found that the ABCB1 (ATP binding cassette subfamily B member 1) modulator zosuquidar dramatically suppresses PD-L1 expression by triggering its autophagic degradation. Mechanistically, ABCB1 interacts with PD-L1 and impairs COP II-mediated PD-L1 transport from ER (endoplasmic reticulum) to Golgi apparatus. The treatment of zosuquidar enhances ABCB1-PD-L1 interaction and leads the ER retention of PD-L1, which is subsequently degraded in the SQSTM1-dependent selective autophagy pathway. In CT26 mouse model and a humanized xenograft mouse model, zosuquidar significantly suppresses tumor growth and accompanies by increased infiltration of cytotoxic T cells. In summary, this study indicates that ABCB1 serves as a negative regulator of PD-L1, and zosuquidar may act as a potential immunotherapy agent by triggering PD-L1 degradation in the early secretory pathway.
Collapse
Affiliation(s)
- Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, 311100, China
| | - Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wenjie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Longsheng Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qianqian Du
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chen Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanjun Xu
- The Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Honghai Wu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Cancer Center of Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| |
Collapse
|
6
|
Espona-Fiedler M, Patthey C, Lindblad S, Sarró I, Öhlund D. Overcoming therapy resistance in pancreatic cancer: New insights and future directions. Biochem Pharmacol 2024; 229:116492. [PMID: 39153553 DOI: 10.1016/j.bcp.2024.116492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is predicted to become the second leading cause of cancer deaths by 2030 and this is mostly due to therapy failure. Limited treatment options and resistance to standard-of-care (SoC) therapies makes PDAC one of the cancer types with poorest prognosis and survival rates [1,2]. Pancreatic tumors are renowned for their poor response to therapeutic interventions including targeted therapies, chemotherapy and radiotherapy. Herein, we review hallmarks of therapy resistance in PDAC and current strategies aiming to tackle escape mechanisms and to re-sensitize cancer cells to therapy. We will further provide insights on recent advances in the field of drug discovery, nanomedicine, and disease models that are setting the ground for future research.
Collapse
Affiliation(s)
- Margarita Espona-Fiedler
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| | - Cedric Patthey
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden
| | - Stina Lindblad
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden
| | - Irina Sarró
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Universitat de Barcelona, Barcelona, Spain
| | - Daniel Öhlund
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| |
Collapse
|
7
|
Feng M, Fan X, Shi J, Shan S, Li S, He S, Ding M, Li Z. Terpenoids from quinoa reverse drug resistance of colon cancer by upregulating miR-495-3p. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8916-8927. [PMID: 38962946 DOI: 10.1002/jsfa.13718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/09/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Quinoa contains far more nutrients than any traditional grain crop. It is known that terpenoids in quinoa have anti-inflammatory and antitumor effects, but their role in reversing drug resistance remains unclear. RESULTS Our previous studies showed that quinoa-derived terpenoid compounds (QBT) can inhibit the occurrence and development of colon cancer. This study further indicates that QBT markedly reverse drug resistance of colon cancer. The results showed that QBT combined with 5-fluorouracil (5-Fu) treatment significantly enhanced the chemotherapy sensitivity of HCT-8/Fu, compared with 5-Fu treatment alone. Moreover, we found that QBT significantly reduced the expression of drug-resistant proteins (P-gp, MRP1, BCRP), and increased the accumulation of chemotherapy drugs. Taking P-gp as the target for biogenesis prediction analysis, results showed that upregulation of miR-495-3p enhanced the chemosensitivity of drug-resistant HCT-8/Fu cells. Besides, the results showed that miR-495-3p was abnormally methylated in HCT-8/Fu compared with HCT-8 colon cancer cells. The expression of methyltransferases DNMT1, DNMT3a and DNMT3b was abnormal. After QBT treatment, the expression level of methyltransferases returned to normal. In addition, the QBT + 5Fu group showed inhibition of tumors in nude mice. CONCLUSION QBT treatment downregulated the expression of drug-resistant protein P-gp by inhibiting the methylation of miR-495-3p, and enhanced the accumulation of 5-Fu in vivo, which in turn reversed its chemoresistance. This suggests that QBT has potential ability as a new drug-resistance reversal agent in colorectal cancer. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Mangmang Feng
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Xiaxia Fan
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Jiangying Shi
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Shuhua Shan
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Songtao Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Shuiling He
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| | - Man Ding
- School of Life Science, Shanxi University, Taiyuan, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China
| |
Collapse
|
8
|
Zhong S, Li Y, Zhu X, Li Z, Jin C, Zhao Y. Knockdown of circ_0006225 overcomes resistance to cisplatin and suppresses growth in lung cancer by miR-1236-3p/ANKRD22 axis. J Biochem Mol Toxicol 2024; 38:e23830. [PMID: 39467212 DOI: 10.1002/jbt.23830] [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: 05/23/2023] [Revised: 07/23/2024] [Accepted: 08/22/2024] [Indexed: 10/30/2024]
Abstract
Cisplatin-based chemotherapy is the mainstay of therapeutic agents for lung cancer. Hence, we investigated the role and mechanism of circ_0006225 in tumorigenesis and cisplatin resistance in lung cancer. Levels of circ_0006225, microRNA (miR)-1236-3p and ankyrin repeat domain 22 (ANKRD22) were detected. Cell cisplatin (DDP) sensitivity and growth were determined by Cell Counting Kit-8, cell colony formation, 5-ethynyl-2'-deoxyuridine, and murine xenograft assays, respectively. A high level of circ_0006225 in lung cancer tissues and cells with cisplatin resistance was observed. Circ_0006225 deletion elevated cisplatin sensitivity and constrained proliferation in DDP-resistant lung cancer cells in vitro. Mechanistically, Circ_0006225 was confirmed to modulate ANKRD22 by sequestering miR-1236-3p. Furthermore, the suppressive effects of circ_0006225 downregulation on cisplatin resistance and proliferation in DDP-resistant lung cancer cells were reversed by miR-1236-3p inhibition or ANKRD22 overexpression. Besides that, circ_0006225 silencing also repressed cisplatin resistance and tumor growth in lung cancer in vivo. In conclusion, knockdown of circ_0006225 restrained the growth and reduced cisplatin resistance in lung cancer by the miR-1236-3p/ANKRD22 axis, suggesting a better effective therapeutic target for overcoming cisplatin resistance in lung cancer patients.
Collapse
Affiliation(s)
- Shengyi Zhong
- Department of Cardiothorcic Surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Yanxing Li
- Department of Cardiothorcic Surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Xiaofeng Zhu
- Department of Cardiothorcic Surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Zhenhua Li
- Department of Cardiothorcic Surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Chi Jin
- Department of Cardiothorcic Surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Yuming Zhao
- Department of Cardiothorcic Surgery, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
9
|
Ge Y, Janson V, Liu H. Comprehensive review on leucine-rich pentatricopeptide repeat-containing protein (LRPPRC, PPR protein): A burgeoning target for cancer therapy. Int J Biol Macromol 2024; 282:136820. [PMID: 39476900 DOI: 10.1016/j.ijbiomac.2024.136820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Leucine-rich pentatricopeptide repeat-containing (LRPPRC), known as the gene mutations that cause Leigh Syndrome French Canadian, encodes a high molecular weight PPR protein (157,905 Da), LRPPRC. LRPPRC binds to DNA, RNA, and proteins to regulate transcription and translation, leading to changes in cell fate. Increasing evidence indicates that LRPPRC plays a pivotal role in various human diseases, particularly cancer in recent years. Here, we review the structure, function, molecular mechanism, as well as inhibitors of LRPPRC. LRPPRC expression elevates in most cancer types and high expression of LRPPRC predicts the poor prognosis of cancer patients. Targeting LRPPRC suppresses tumor progression by affecting several cancer hallmarks, including signal transduction, cancer metabolism, and immune regulation. LRPPRC is a promising target in cancer research, serving as both a biomarker and therapeutic target. Further studies are required to extend the understanding of LRPPRC function and molecular mechanism, as well as to refine novel therapeutic strategies targeting LRPPRC in cancer therapy.
Collapse
Affiliation(s)
- Yunxiao Ge
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Victor Janson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China.
| |
Collapse
|
10
|
Parisi C, Laneri F, Martins TJ, Fraix A, Sortino S. Nitric Oxide-Photodelivering Materials with Multiple Functionalities: From Rational Design to Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39445390 DOI: 10.1021/acsami.4c13478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The achievement of materials that are able to release therapeutic agents under the control of light stimuli to improve therapeutic efficacy is a significant challenge in health care. Nitric oxide (NO) is one of the most studied molecules in the fascinating realm of biomedical sciences, not only for its crucial role as a gaseous signaling molecule in the human body but also for its great potential as an unconventional therapeutic in a variety of diseases including cancer, bacterial and viral infections, and neurodegeneration. Handling difficulties due to its gaseous nature, reduced region of action due to its short half-life, and strict dependence of the biological effects on its concentration and generation site are critical questions to be solved for appropriate therapeutic uses of NO. Light-activatable NO precursors, namely, NO photodonors (NOPDs), address the above issues since they are stable in the dark and permit in a noninvasive fashion the remote-controlled delivery of NO on demand with great spatiotemporal precision. Engineering biocompatible materials with NOPDs and their combination with additional imaging, therapeutic, and phototherapeutic components leads to intriguing light-responsive multifunctional constructs exhibiting promising potential for biomedical applications. This contribution illustrates the most significant progress made over the last five years in achieving engineered materials including nanoparticles, gels, and thin films, sharing the common feature to deliver NO under the exclusive control of the biocompatible visible/near infrared light inputs. We will highlight the logical design behind the fabrication of these systems, illustrating the potential therapeutic applications with particular emphasis on cancer and bacterial infections.
Collapse
Affiliation(s)
- Cristina Parisi
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Francesca Laneri
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Tassia J Martins
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Aurore Fraix
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Salvatore Sortino
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| |
Collapse
|
11
|
Shi Z, Zeng Y, Luo J, Wang X, Ma G, Zhang T, Huang P. Endogenous Magnetic Lipid Droplet-Mediated Cascade-Targeted Sonodynamic Therapy as an Approach to Reversing Breast Cancer Multidrug Resistance. ACS NANO 2024; 18:28659-28674. [PMID: 39387174 DOI: 10.1021/acsnano.4c05938] [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: 10/12/2024]
Abstract
Multidrug resistance (MDR) has emerged as a major barrier to effective breast cancer treatment, contributing to high rates of chemotherapy failure and disease recurrence. There is thus a pressing need to overcome MDR and to facilitate the efficient and precise treatment of breast cancer in a targeted manner. In this study, endogenous functional lipid droplets (IR780@LDs-Fe3O4/OA) were developed and used to effectively overcome the limited diffusion distance of reactive oxygen species owing to their amenability to cascade-targeted delivery, thereby facilitating precise and effective sonodynamic therapy (SDT) for MDR breast cancer. Initially, IR780@LDs-Fe3O4/OA was efficiently enriched within tumor sites in a static magnetic field, achieving the visualization of tumor treatment. Subsequently, the cascade-targeted SDT combined with the Fenton effect induced lysosome membrane permeabilization and relieved lysosomal sequestration, thus elevating drug concentration at the target site. This treatment approach also suppressed ATP production, thereby inhibiting P-glycoprotein-mediated chemotherapeutic drug efflux. This cascade-targeted SDT strategy significantly increased the sensitivity of MDR cells to doxorubicin, increasing the IC50 value of doxorubicin by approximately 10-fold. Moreover, the cascade-targeted SDT also altered the gene expression profiles of MDR cells and suppressed the expression of MDR-related genes. In light of these promising results, the combination of cascade-targeted SDT and conventional chemotherapy holds great clinical promise as an effective treatment modality with excellent biocompatibility that can improve MDR breast cancer patient outcomes.
Collapse
Affiliation(s)
- Zhan Shi
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Yiqing Zeng
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Jiali Luo
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Xue Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Guangrong Ma
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Tao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, No. 66 Dongxin Avenue, Binjiang District, Hangzhou 310053, P. R. China
| |
Collapse
|
12
|
Wang S, Wang SQ, Chen XB, Xu Q, Deng H, Teng QX, Chen ZS, Zhang X, Chen FE. Cell-Based Screen Identifies a Highly Potent and Orally Available ABCB1 Modulator for Treatment of Multidrug Resistance. J Med Chem 2024. [PMID: 39425773 DOI: 10.1021/acs.jmedchem.4c01081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Targeting ABCB1 is a promising strategy in combating multidrug resistance. Our cell-based phenotypic screening led to the discovery of novel triazolo[1,5-a]pyrimidone-based ABCB1 modulators. Notably, WS-917 was identified as a significant contributor to heightened sensitization of human colorectal adenocarcinoma cells (SW620/Ad300) to paclitaxel (IC50 = 5 nM). Mechanistic elucidation revealed that this compound substantially augmented intracellular paclitaxel and [3H]-paclitaxel, concurrently mitigating the efflux of [3H]-paclitaxel in SW620/Ad300 through the inhibition of ABCB1 efflux. The cellular thermal shift assay underscored its ability to stabilize ABCB1 through direct binding. Additionally, WS-917 induced stimulation of ABCB1 ATPase activity while exhibiting negligible inhibitory effect against CYP3A4. Remarkable was its capacity to enhance the sensitivity of SW620/Ad300 to paclitaxel, as well as the sensitivity of CT26/TAXOL to paclitaxel and PD-L1 inhibitor (Atezolizumab) in vivo, all achieved without inducing observable toxicity. The discovery of WS-917 holds promise for the development of more potent ABCB1 modulators.
Collapse
Affiliation(s)
- Shuai Wang
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin 133002, China
- Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China
| | - Sai-Qi Wang
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xiao-Bing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Qian Xu
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China
- Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China
| | - Hao Deng
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China
- Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China
| | - Qiu-Xu Teng
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, New York 11439, United States
| | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Fen-Er Chen
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin 133002, China
- Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
13
|
Oshchepkova A, Chernikov I, Miroshnichenko S, Patutina O, Markov O, Savin I, Staroseletz Y, Meschaninova M, Puchkov P, Zhukov S, Kupryushkin M, Maslov M, Sen’kova A, Vlassov V, Chernolovskaya E, Zenkova M. Extracellular vesicle mimetics as delivery vehicles for oligonucleotide-based therapeutics and plasmid DNA. Front Bioeng Biotechnol 2024; 12:1437817. [PMID: 39493304 PMCID: PMC11528538 DOI: 10.3389/fbioe.2024.1437817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Small membrane particles called extracellular vesicles (EVs) transport biologically active cargo between cells, providing intercellular communication. The clinical application of EVs is limited due to the lack of scalable and cost-effective approaches for their production and purification, as well as effective loading strategies. Methods Here we used EV mimetics produced by cell treatment with the actin-destabilizing agent cytochalasin B as an alternative to EVs for the delivery of therapeutic nucleic acids. Results Cytochalasin-B-inducible nanovesicles (CINVs) delivered a fully modified N-(methanesulfonyl)- or mesyl (µ-) antisense oligonucleotide to B16 melanoma cells, selectively decreasing the level of target microRNA-21 with effectiveness comparable to that observed upon Lipofectamine 2000-mediated delivery. The efficiency of the CINV-mediated delivery of plasmid DNA encoding EGFP varied depending on the type of recipient cells. Surprisingly, under experimental conditions, CINVs were unable to deliver both modified and natural short RNA duplexes-small interfering RNA and immunostimulatory RNA-probably due to their poor loading into CINVs. Discussion CINVs demonstrated unique properties for the delivery of therapeutic nucleic acids, especially for antisense oligonucleotide-based therapy.
Collapse
Affiliation(s)
- Anastasiya Oshchepkova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Ivan Chernikov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Svetlana Miroshnichenko
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Olga Patutina
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Oleg Markov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Innokenty Savin
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Yaroslav Staroseletz
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Mariya Meschaninova
- Laboratory of RNA Chemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Pavel Puchkov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow, Russia
| | - Sergey Zhukov
- Laboratory of Nucleic Acids Chemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Maxim Kupryushkin
- Laboratory of Nucleic Acids Chemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Mikhail Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow, Russia
| | - Aleksandra Sen’kova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Valentin Vlassov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Elena Chernolovskaya
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Marina Zenkova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| |
Collapse
|
14
|
Goyal A, Murkute SL, Bhowmik S, Prasad CP, Mohapatra P. Belling the "cat": Wnt/β-catenin signaling and its significance in future cancer therapies. Biochim Biophys Acta Rev Cancer 2024; 1879:189195. [PMID: 39413855 DOI: 10.1016/j.bbcan.2024.189195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
The WNT/β-catenin is among one of the most extensively studied cellular signaling pathways involved in the initiation and progression of several deadly cancers. It is now understood that the WNT/β-catenin signaling, during tumor progression operates in a very complex fashion beyond the earlier assumed simple WNT 'On' or 'Off' mode as it recruits numerous WNT ligands, receptors, transcriptional factors and also cross-talks with other signaling molecules including the noncanonical WNT regulators. WNT/β-catenin signaling molecules are often mutated in different cancers which makes them very challenging to inhibit and sometimes ranks them among the undruggable targets. Furthermore, due to the evolutionary conservation of this pathway, inhibiting WNT/β-catenin has caused significant toxicity in normal cells. These challenges are reflected in clinical trial data, where the use of WNT/β-catenin inhibitors as standalone treatments remains limited. In this review, we have highlighted the crucial functional associations of diverse WNT/β-catenin signaling regulators with cancer progression and the phenotypic switching of tumor cells. Next, we have shed light on the roles of WNT/β-catenin signaling in drug resistance, clonal evolution, tumor heterogeneity, and immune evasion. The present review also focuses on various classes of routine and novel WNT/β-catenin therapeutic regimes while addressing the challenges associated with targeting the regulators of this complex pathway. In the light of multiple case studies on WNT/β-catenin inhibitors, we also highlighted the challenges and opportunities for future clinical trial strategies involving these treatments. Additionally, we have proposed strategies for future WNT/β-catenin-based drug discovery trials, emphasizing the potential of combination therapies and AI/ML-driven prediction approaches. Overall, here we showcased the opportunities, possibilities, and potentialities of WNT/β-catenin signaling modulatory therapeutic regimes as promising precision cancer medicines for the future.
Collapse
Affiliation(s)
- Akansha Goyal
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Satyajit Laxman Murkute
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Sujoy Bhowmik
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology Lab, DR BRA-IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Purusottam Mohapatra
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India.
| |
Collapse
|
15
|
Li S, Wang Y, Li C, Zhou B, Zeng X, Zhu H. Supramolecular nanomedicine in the intelligent cancer therapy: recent advances and future. Front Pharmacol 2024; 15:1490139. [PMID: 39464634 PMCID: PMC11502448 DOI: 10.3389/fphar.2024.1490139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
In recent years, the incidence of cancer has been increasing year by year, and the burden of the disease and the economic burden caused by it has been worsening. Although chemotherapy, immunotherapy, targeted therapy and other therapeutic means continue to progress, they still inevitably have problems such as high toxicity and side effects, susceptibility to drug resistance, and high price. Photothermal therapy and photodynamic therapy have demonstrated considerable advantages in cancer imaging and treatment due to their minimally invasive and selective nature. However, their development has been constrained by challenges related to drug delivery. In recent times, drug delivery systems constructed based on supramolecular chemistry have been the subject of considerable interest, particularly in view of their compatibility with the high permeability and long retention effect of tumors. Furthermore, the advantage of dissociating the active ingredient under pH, light and other stimuli makes them unique in cancer therapy. This paper reviews the current status of supramolecular nanomedicines in cancer therapy, elucidating the challenges faced and providing a theoretical basis for the efficient and precise treatment of malignant tumors.
Collapse
Affiliation(s)
- Shuo Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yujiao Wang
- Division of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Binghao Zhou
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoxi Zeng
- Division of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Sun NY, Kumar S, Kim YS, Varghese D, Mendoza A, Nguyen R, Okada R, Reilly K, Widemann B, Pommier Y, Elloumi F, Dhall A, Patel M, Aber E, Contreras-Burrola C, Kaplan R, Martinez D, Pogoriler J, Hamilton AK, Diskin SJ, Maris JM, Robey RW, Gottesman MM, Rivero JD, Roper N. Identification of DLK1, a Notch ligand, as an immunotherapeutic target and regulator of tumor cell plasticity and chemoresistance in adrenocortical carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617077. [PMID: 39416174 PMCID: PMC11482787 DOI: 10.1101/2024.10.09.617077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Immunotherapeutic targeting of cell surface proteins is an increasingly effective cancer therapy. However, given the limited number of current targets, the identification of new surface proteins, particularly those with biological importance, is critical. Here, we uncover delta-like non-canonical Notch ligand 1 (DLK1) as a cell surface protein with limited normal tissue expression and high expression in multiple refractory adult metastatic cancers including small cell lung cancer (SCLC) and adrenocortical carcinoma (ACC), a rare cancer with few effective therapies. In ACC, ADCT-701, a DLK1 targeting antibody-drug conjugate (ADC), shows potent in vitro activity among established cell lines and a new cohort of patient-derived organoids as well as robust in vivo anti-tumor responses in cell line-derived and patient-derived xenografts. However, ADCT-701 efficacy is overall limited in ACC due to high expression and activity of the drug efflux protein ABCB1 (MDR1, P-glycoprotein). In contrast, ADCT-701 is extremely potent and induces complete responses in DLK1+ ACC and SCLC in vivo models with low or no ABCB1 expression. Genetic deletion of DLK1 in ACC dramatically downregulates ABCB1 and increases ADC payload and chemotherapy sensitivity through NOTCH1-mediated adrenocortical de-differentiation. Single cell RNA-seq of ACC metastatic tumors reveals significantly decreased adrenocortical differentiation in DLK low or negative cells compared to DLK1 positive cells. This works identifies DLK1 as a novel immunotherapeutic target that regulates tumor cell plasticity and chemoresistance in ACC. Our data support targeting DLK1 with an ADC in ACC and neuroendocrine neoplasms in an active first-in-human phase I clinical trial (NCT06041516).
Collapse
Affiliation(s)
- Nai-Yun Sun
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Suresh Kumar
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Yoo Sun Kim
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Diana Varghese
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Rosa Nguyen
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Reona Okada
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Karlyne Reilly
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Brigitte Widemann
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Fathi Elloumi
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Anjali Dhall
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Mayank Patel
- Laboratory of Pathology, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Etan Aber
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | | | - Rosie Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Dan Martinez
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Pogoriler
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amber K. Hamilton
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, and Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sharon J. Diskin
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, and Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - John M. Maris
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, and Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, NCI, Bethesda, MD, USA
| | | | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| | - Nitin Roper
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, Bethesda, MD, USA
| |
Collapse
|
17
|
Parisi C, Laneri F, Fraix A, Sortino S. Multifunctional Molecular Hybrids Photoreleasing Nitric Oxide: Advantages, Pitfalls, and Opportunities. J Med Chem 2024; 67:16932-16950. [PMID: 39009572 DOI: 10.1021/acs.jmedchem.4c01038] [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: 07/17/2024]
Abstract
The multifaceted role nitric oxide (NO) plays in human physiology and pathophysiology has opened new scenarios in biomedicine by exploiting this free radical as an unconventional therapeutic against important diseases. The difficulties in handling gaseous NO and the strict dependence of the biological effects on its doses and location have made the light-activated NO precursors, namely NO photodonors (NOPDs), very appealing by virtue of their precise spatiotemporal control of NO delivery. The covalent integration of NOPDs and additional functional components within the same molecular skeleton through suitable linkers can lead to an intriguing class of multifunctional photoactivatable molecular hybrids. In this Perspective, we provide an overview of the recent advances in these molecular constructs, emphasizing those merging NO photorelease with targeting, fluorescent reporting, and phototherapeutic functionalities. We will highlight the rational design behind synthesizing these molecular hybrids and critically describe the advantages, drawbacks, and opportunities they offer in biomedical research.
Collapse
Affiliation(s)
- Cristina Parisi
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Francesca Laneri
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Aurore Fraix
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Salvatore Sortino
- PhotoChemLab, Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| |
Collapse
|
18
|
Lin BH, Li YC, Murakami M, Wu YS, Huang YH, Hung TH, Ambudkar SV, Wu CP. Epertinib counteracts multidrug resistance in cancer cells by antagonizing the drug efflux function of ABCB1 and ABCG2. Biomed Pharmacother 2024; 180:117542. [PMID: 39388999 DOI: 10.1016/j.biopha.2024.117542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
A significant hurdle in cancer treatment arises from multidrug resistance (MDR), often due to overexpression of ATP-binding cassette (ABC) transporters like ABCB1 and/or ABCG2 in cancer cells. These transporters actively diminish the efficacy of cytotoxic drugs by facilitating ATP hydrolysis-dependent drug efflux and reducing intracellular drug accumulation in cancer cells. Addressing multidrug-resistant cancers poses a significant challenge due to the lack of approved treatments, prompting the exploration of alternative avenues like drug repurposing (also referred to as drug repositioning) of molecularly targeted agents to reverse MDR-mediated by ABCB1 and/or ABCG2 in multidrug-resistant cancer cells. Epertinib, a potent inhibitor of EGFR and HER2 currently in clinical trials for solid tumors, was investigated for its potential to resensitize ABCB1- and ABCG2-overexpressing multidrug-resistant cancer cells to chemotherapeutic agents. Our findings reveal that at sub-toxic, submicromolar concentrations, epertinib restores the sensitivity of multidrug-resistant cancer cells to cytotoxic drugs in a concentration-dependent manner. The results demonstrate that epertinib enhances drug-induced apoptosis in these cancer cells by impeding the drug-efflux function of ABCB1 and ABCG2 without altering their expression. ATPase activity and molecular docking were employed to reveal potential interaction sites between epertinib and the drug-binding pockets of ABCB1 and ABCG2. In summary, our study demonstrates an additional pharmacological capability of epertinib against the activity of ABCB1 and ABCG2. These findings suggest that incorporating epertinib into combination therapy could be advantageous for a specific patient subset with tumors exhibiting high levels of ABCB1 or ABCG2, warranting further exploration.
Collapse
Affiliation(s)
- Bing-Huan Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Yen-Ching Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Megumi Murakami
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan.
| | - Yang-Hui Huang
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan.
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | - Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
19
|
Stojanović Gužvić N, Lüke F, Treitschke S, Coluccio A, Hoffmann M, Feliciello G, Varadarajan AR, Lu X, Weidele K, Botteron C, Materna-Reichelt S, Keil F, Evert K, Weber F, Schamberger T, Althammer M, Grosse J, Hellwig D, Schulz C, Seitz S, Ugocsai P, Schlenska-Lange A, Mayr R, Kaiser U, Dietmaier W, Polzer B, Warfsmann J, Honarnejad K, Pukrop T, Heudobler D, Klein CA, Werno C. Cellular liquid biopsy provides unique chances for disease monitoring, preclinical model generation and therapy adjustment in rare salivary gland cancer patients. Mol Oncol 2024. [PMID: 39367702 DOI: 10.1002/1878-0261.13741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 10/06/2024] Open
Abstract
While cell-free liquid biopsy (cfLB) approaches provide simple and inexpensive disease monitoring, cell-based liquid biopsy (cLB) may enable additional molecular genetic assessment of systemic disease heterogeneity and preclinical model development. We investigated 71 blood samples of 62 patients with various advanced cancer types and subjected enriched circulating tumor cells (CTCs) to organoid culture conditions. CTC-derived tumoroid models were characterized by DNA/RNA sequencing and immunohistochemistry, as well as functional drug testing. Results were linked to molecular features of primary tumors, metastases, and CTCs; CTC enumeration was linked to disease progression. Of 52 samples with positive CTC counts (≥1) from eight different cancer types, only CTCs from two salivary gland cancer (SGC) patients formed tumoroid cultures (P = 0.0005). Longitudinal CTC enumeration of one SGC patient closely reflected disease progression during treatment and revealed metastatic relapse earlier than clinical imaging. Multiomics analysis and functional in vitro drug testing identified potential resistance mechanisms and drug vulnerabilities. We conclude that cLB might add a functional dimension (to the genetic approaches) in the personalized management of rare, difficult-to-treat cancers such as SGC.
Collapse
Affiliation(s)
| | - Florian Lüke
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Steffi Treitschke
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Andrea Coluccio
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Martin Hoffmann
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | | | | | - Xin Lu
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Kathrin Weidele
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Catherine Botteron
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | | | - Felix Keil
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Insitute for Pathology, University of Regensburg, Germany
| | - Katja Evert
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Insitute for Pathology, University of Regensburg, Germany
| | - Florian Weber
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Insitute for Pathology, University of Regensburg, Germany
| | - Thomas Schamberger
- Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Michael Althammer
- Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Jirka Grosse
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Germany
| | - Dirk Hellwig
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Germany
| | - Christian Schulz
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Internal Medicine II, University Hospital Regensburg, Germany
| | - Stephan Seitz
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Obstetrics and Gynecology, University Hospital Regensburg, Germany
| | - Peter Ugocsai
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Department of Obstetrics and Gynecology, University Hospital Regensburg, Germany
| | - Anke Schlenska-Lange
- Department of Oncology and Hematology, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Roman Mayr
- Department of Urology, Caritas St. Josef Medical Center, University of Regensburg, Germany
| | - Ulrich Kaiser
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | | | - Bernhard Polzer
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Jens Warfsmann
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Kamran Honarnejad
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| | - Tobias Pukrop
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, University Hospital Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Christoph A Klein
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
- Experimental Medicine and Therapy Research, University of Regensburg, Germany
| | - Christian Werno
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM-R, Germany
| |
Collapse
|
20
|
Zeng M, Sun S, Feng H, Tan Z, Zhao J, Wu Y, Yuan W, Li Z, Qiu J, Niu M, Gu X. Discovery of novel third generation P-glycoprotein inhibitors bearing an azo moiety with MDR-reversing effect. Eur J Med Chem 2024; 280:116943. [PMID: 39369480 DOI: 10.1016/j.ejmech.2024.116943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
P-glycoprotein (P-gp)-caused multidrug resistance (MDR) is a crucial factor in the cancer chemotherapy failure. Herein, a total of twenty two azo-containing WK-X-34 (WK34, a third generation P-gp inhibitor) derivatives were synthesized as novel P-gp inhibitors. Biological evaluation revealed that compound 7i effectively reversed P-gp-mediated MDR in K562/A02 cells, with a higher reversal fold (RF) value than WK34 (142.79 vs. 64.41). Further investigation indicated that 7i dose-dependently inhibited P-gp function, without affecting its expression. CETSA results illustrated that 7i could obviously improve P-gp stability, suggesting its high affinity with P-gp. Molecular docking analysis revealed that 7i fit well into P-gp's binding pocket, thus displaying potent reversal effect on P-gp-mediated tumor MDR Optical properties evaluation confirmed that azo-containing 7i can undergo reversible changes in the cis and trans configurations under the irradiation of 365 nm and 520 nm wavelength of light. Notably, the configuration change of azo might affect the MDR-reversal potency, and cis-7i has a lower RF value than trans-7i (122.70 vs. 142.79), suggesting that development of photoswitchable P-gp inhibitors might be a novel strategy to reduce the systemic toxicity caused by indiscriminate inhibition of P-gp by traditional inhibitors. Collectively, 7i, as a novel P-gp inhibitor, warranted further investigation.
Collapse
Affiliation(s)
- Meifeng Zeng
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Shuang Sun
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Hao Feng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Zhoupeng Tan
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Jiangyan Zhao
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yiqing Wu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Weiqun Yuan
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Zheng Li
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Jingying Qiu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Mingshan Niu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China; Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, People's Republic of China.
| | - Xiaoke Gu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China.
| |
Collapse
|
21
|
Sahu D, Shi J, Segura Rueda IA, Chatrath A, Dutta A. Development of a polygenic score predicting drug resistance and patient outcome in breast cancer. NPJ Precis Oncol 2024; 8:219. [PMID: 39358487 PMCID: PMC11447244 DOI: 10.1038/s41698-024-00714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
Gene expression profiles of hundreds of cancer cell-lines and the cell-lines' response to drug treatment were analyzed to identify genes whose expression correlated with drug resistance. In the GDSC dataset of 809 cancer cell lines, expression of 36 genes were associated with drug resistance (increased IC50) to many anti-cancer drugs. This was validated in the CTRP dataset of 860 cell lines. A polygenic score derived from the correlation coefficients of the 36 genes in cancer cell lines, UAB36, predicted resistance of cell lines to Tamoxifen. Although the 36 genes were selected from cell line behaviors, UAB36 successfully predicted survival of breast cancer patients in three different cohorts of patients treated with Tamoxifen. UAB36 outperforms two existing predictive gene signatures and is a predictor of outcome of breast cancer patients independent of the known clinical co-variates that affect outcome. This approach should provide promising polygenic biomarkers for resistance in many cancer types against specific drugs.
Collapse
Affiliation(s)
- Divya Sahu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jeffrey Shi
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
| | | | - Ajay Chatrath
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Anindya Dutta
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
22
|
Chen R, Ren Z, Bai L, Hu X, Chen Y, Ye Q, Hu Y, Shi J. Novel antibody-drug conjugates based on DXd-ADC technology. Bioorg Chem 2024; 151:107697. [PMID: 39121594 DOI: 10.1016/j.bioorg.2024.107697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
In recent years, antibody-drug conjugate (ADC) technology, which uses monoclonal antibodies (mAbs) to specifically deliver effective cytotoxic payloads to tumor cells, has become a promising method of tumor targeted therapy. ADCs are a powerful class of biopharmaceuticals that link antibodies targeting specific antigens and small molecule drugs with potent cytotoxicity via a linker, thus enabling selective destruction of cancer cells while minimizing systemic toxicity. DXd is a topoisomerase I inhibitor that induces DNA damage leading to cell cycle arrest, making it an option for ADC payloads. The DXd-ADC technology, developed by Daiichi Sankyo, is a cutting-edge platform that produces a new generation of ADCs with improved therapeutic metrics and has shown significant therapeutic potential in various types of cancer. This review provides a comprehensive assessment of drugs developed with DXd-ADC technology, with a focus on mechanisms of action, pharmacokinetics studies, preclinical data, and clinical outcomes for DS-8201a, U3-1402, DS-1062a, DS-7300a, DS-6157a, and DS-6000a. By integrating existing data, we aim to provide valuable insights into the current therapeutic status and future prospects of these novel agents.
Collapse
Affiliation(s)
- Rong Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Zhiwen Ren
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuefang Hu
- Key Laboratory of Agro-Products Postharvest Handling, Ministry of Agriculture, Academy of Agricultural Planning and Engineering Mara, Beijing 100121, China
| | - Yuchen Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Qiang Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Yuan Hu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
23
|
Li Y, Guo F, Wang W, Lv F, Zhang L, Zhu M, Yang S, Dong S, Zhou M, Li Z, Zhu Z, Yang JM, Zhang Y. Marein, a novel natural product for restoring chemo-sensitivity to cancer cells through competitive inhibition of ABCG2 function. Biochem Pharmacol 2024; 228:116219. [PMID: 38643907 DOI: 10.1016/j.bcp.2024.116219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Abstract
The pivotal roles of ATP-binding cassette (ABC) transporters in drug resistance have been widely appreciated. Here we report that marein, a natural product from Coreopsis tinctoria Nutt, is a potent chemo-sensitizer in drug resistant cancer cells overexpressing ABCG2 transporter. We demonstrate that marein can competitively inhibit efflux activity of ABCG2 protein and increase the intracellular accumulation of the chemotherapeutic drugs that belong to substrate of this transporter. We further show that marein can bind to the conserved amino acid residue F439 of ABCG2, a critical site for drug-substrate interaction. Moreover, marein can significantly sensitize the ABCG2-expressing tumor cells to chemotherapeutic drugs such as topotecan, mitoxantrone, and olaparib. This study reveals a novel role and mechanism of marein in modulating drug resistance, and may have important implications in treatment of cancers that are resistant to chemotherapeutic drugs that belong to the substrates of ABCG2 transporters.
Collapse
Affiliation(s)
- Yixuan Li
- Department of Pharmacy, Taicang TCM Hospital Affiliated to Nanjing University of Chinese Medicine (Taicang Hospital of Traditional Chinese Medicine), Jiangsu, China
| | - Fanfan Guo
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Wenjing Wang
- Department of Pharmacy, The Affiliated Children's Hospital of Soochow University, Jiangsu, China
| | - FangLin Lv
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Lu Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Mingxian Zhu
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Shumin Yang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Shunli Dong
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Mingxuan Zhou
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Zhenyun Li
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Zengyan Zhu
- Department of Pharmacy, The Affiliated Children's Hospital of Soochow University, Jiangsu, China.
| | - Jin-Ming Yang
- Department of Cancer Biology and Toxicology, Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, KY 40506, USA.
| | - Yi Zhang
- Department of Pharmacy, Taicang TCM Hospital Affiliated to Nanjing University of Chinese Medicine (Taicang Hospital of Traditional Chinese Medicine), Jiangsu, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China.
| |
Collapse
|
24
|
Fang H, Zhou Y, Bai X, Che W, Zhang W, Zhang D, Chen Q, Duan W, Nie G, Hou Y. The VEGFA-Induced MAPK-AKT/PTEN/TGFβ Signal Pathway Enhances Progression and MDR in Gastric Cancer. Genes (Basel) 2024; 15:1266. [PMID: 39457390 PMCID: PMC11507385 DOI: 10.3390/genes15101266] [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: 09/06/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Gastric cancer (GC) is a globally frequent cancer, in particular leading in mortality caused by digestive tract cancers in China. Vascular endothelial growth factor A (VEGFA) is excessively expressed in cancers including GC; its involvement in GC development, particularly in multidrug resistance (MDR), and the signal route it affects in GC remain unknown. To explore the roles VEGFA plays during progression and MDR formation in GC, we studied its function in a VEGFA-deleted GC cell platform. METHODS We initially assessed the importance of VEGFA in GC and MDR using database analysis. Then, using CCK8, wound healing, transwell, scanning electron microscopy, immunofluorescence, flow cytometry, and other techniques, the alterations in tumor malignancy-connected cell behaviors and microstructures were photographed and evaluated in a VEGFA-gene-deleted GC cell line (VEGFA-/-SGC7901). Finally, the mechanism of VEGFA in GC progression and MDR was examined by Western blot. RESULTS Database analysis revealed a strong correlation between high VEGFA expression and a poor prognosis for GC. The results showed that VEGFA deletion reduced GC cell proliferation and motility and altered microstructures important for motility, such as the depolymerized cytoskeleton. VEGFA deletion inhibited the growth of pseudopodia/filopodia and suppressed the epithelial-mesenchymal transition (EMT). The occurrence of MDR is induced by overactivation of the MAPK-AKT and TGFβ signaling pathways, while PTEN inhibits these pathways. CONCLUSIONS All findings suggested that VEGFA acts as a cancer enhancer and MDR inducer in GC via the MAPK-AKT/PTEN/TGFβ signal pathway.
Collapse
Affiliation(s)
- Hongming Fang
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| | - Yujuan Zhou
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| | - Xue Bai
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| | - Wanlin Che
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| | - Wenxuan Zhang
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| | - Danying Zhang
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| | - Qingmei Chen
- Guangxi Key Laboratory of Agricultural Resource Chemistry and Biotechnology, 299 Jiao-Yu-Zhong Road, Yulin 537000, China;
| | - Wei Duan
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Melbourne, VIC 3216, Australia;
| | - Guochao Nie
- Guangxi Key Laboratory of Agricultural Resource Chemistry and Biotechnology, 299 Jiao-Yu-Zhong Road, Yulin 537000, China;
| | - Yingchun Hou
- College of Life Sciences, Shaanxi Normal University, 620 West Chang-An Street, Xi’an 710119, China; (H.F.); (Y.Z.); (X.B.); (W.C.); (W.Z.); (D.Z.)
| |
Collapse
|
25
|
Maimaitijiang A, He D, Li D, Li W, Su Z, Fan Z, Li J. Progress in Research of Nanotherapeutics for Overcoming Multidrug Resistance in Cancer. Int J Mol Sci 2024; 25:9973. [PMID: 39337463 PMCID: PMC11432649 DOI: 10.3390/ijms25189973] [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: 07/28/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Chemotherapy has been widely applied in oncotherapy. However, the development of multidrug resistance (MDR) has diminished the effectiveness of anticancer drugs against tumor cells. Such resistance often results in tumor recurrence, metastasis, and patient death. Fortunately, nanoparticle-based drug delivery systems provide a promising strategy by codelivery of multiple drugs and MDR reversal agents and the skillful, flexible, smart modification of drug targets. Such systems have demonstrated the ability to bypass the ABC transporter biological efflux mechanisms due to drug resistance. Hence, how to deliver drugs and exert potential antitumor effects have been successfully explored, applied, and developed. Furthermore, to overcome multidrug resistance, nanoparticle-based systems have been developed due to their good therapeutic effect, low side effects, and high tumor metastasis inhibition. In view of this, we systematically discuss the molecular mechanisms and therapeutic strategies of MDR from nanotherapeutics. Finally, we summarize intriguing ideas and future trends for further research in overcoming MDR.
Collapse
Affiliation(s)
- Ayitila Maimaitijiang
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dongze He
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dingyang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Wenfang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhengding Su
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhongxiong Fan
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
26
|
Ye G, Ye M, Jin X. Roles of clinical application of lenvatinib and its resistance mechanism in advanced hepatocellular carcinoma (Review). Am J Cancer Res 2024; 14:4113-4171. [PMID: 39417171 PMCID: PMC11477829 DOI: 10.62347/ujvp4361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lenvatinib (LEN) is a multi-target TKI, which plays a pivotal role in the treatment of advanced hepatocellular carcinoma (HCC). The inevitable occurrence of drug resistance still prevents curative potential and is deleterious for the prognosis, and a growing body of studies is accumulating, which have devoted themselves to unveiling its underlying resistance mechanism and made some progress. The dysregulation of crucial signaling pathways, non-coding RNA and RNA modifications were proven to be associated with LEN resistance. A range of drugs were found to influence LEN therapeutic efficacy. In addition, the superiority of LEN combination therapy has been shown to potentially overcome the limitations of LEN monotherapy in a series of research, and a range of promising indicators for predicting treatment response and prognosis have been discovered in recent years. In this review, we summarize the latest developments in LEN resistance, the efficacy and safety of LEN combination therapy as well as associated indicators, which may provide new insight into its resistance as well as ideas in the treatment of advanced HCC.
Collapse
Affiliation(s)
- Ganghui Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
- Department of Radiation Oncology, Taizhou Central Hospital (Taizhou University Hospital)Taizhou 318000, Zhejiang, P. R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| |
Collapse
|
27
|
He L, Qian H, Seyiti A, Yang C, Shi N, Chen C, Zhang P, Hou Y. CD133 +/ABCC5 + cervical cancer cells exhibit cancer stem cell properties. Heliyon 2024; 10:e37066. [PMID: 39296204 PMCID: PMC11408061 DOI: 10.1016/j.heliyon.2024.e37066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Objective This study explores the correlation between Forkhead box M1 (FOXM1) and ATP-binding cassette subfamily C member 5 (ABCC5) in relation to paclitaxel resistance in cervical cancer. It aims to identify potential cervical cancer stem cell markers, offering fresh perspectives for developing therapeutic strategies to overcome paclitaxel chemoresistance in cervical cancer. Methods Paclitaxel-resistant Hela cells (Hela/Taxol) were developed by intermittently exposing Hela cells to progressively increasing concentrations of paclitaxel. We assessed the biological properties of both Hela and Hela/Taxol cells using various assays: cell proliferation, clonogenic, cell cycle, apoptosis, scratch, and transwell. To determine which markers better represent tumor stem cells, we analyzed various known and potential stem cell markers in combination. Flow cytometry was employed to measure the proportion of positive markers in both parental and drug-resistant cell lines. Following statistical analysis to establish relative stability, CD133+ABCC5+ cells were sorted for further examination. Subsequent tests included sphere-forming assays and Western blot analysis to detect the presence of the stem cell-specific protein Sox2, aiding in the identification of viable cervical cancer stem cell markers. Results The Hela/Taxol cell line exhibited significantly enhanced proliferation, migration, and invasion capabilities compared to the Hela cell line, alongside a marked reduction in apoptosis rates (P < 0.01). Notably, proportions of CD44+, CD24+CD44+, ABCC5+, CD24+CD44+ABCC5+, CD44+ABCC5+, CD24+CD44+FOXM1+, CD44+FOXM1+, CD133+ABCC5+, and CD133+FOXM1+ were significantly higher (P < 0.05). Furthermore, the size and number of spheres formed byCD133+ABCC5+ cells were greater in the sorted Hela/Taxol line (P < 0.01), with increased expression of the stem cell marker Sox2 (P < 0.001). Conclusion The Hela/Taxol cells demonstrate increased tumoral stemness, suggesting that CD133+ABCC5+ may serve as a novel marker for cervical cancer stem cells.
Collapse
Affiliation(s)
- Lin He
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Hengjun Qian
- Yibin Second People's Hospital, Sichuan, 644002, PR China
| | - Ayinuer Seyiti
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang, 830011, PR China
| | - Chengshaoxiong Yang
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Ning Shi
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Chen Chen
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Pingxu Zhang
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830011, PR China
| | - Youxiang Hou
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang, 830011, PR China
| |
Collapse
|
28
|
Ung J, Kassai M, Tan SF, Loughran TP, Feith DJ, Cabot MC. The Drug Transporter P-Glycoprotein and Its Impact on Ceramide Metabolism-An Unconventional Ally in Cancer Treatment. Int J Mol Sci 2024; 25:9825. [PMID: 39337312 PMCID: PMC11432138 DOI: 10.3390/ijms25189825] [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: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
The tumor-suppressor sphingolipid ceramide is recognized as a key participant in the cytotoxic mechanism of action of many types of chemotherapy drugs, including anthracyclines, Vinca alkaloids, the podophyllotoxin etoposide, taxanes, and the platinum drug oxaliplatin. These drugs can activate de novo synthesis of ceramide or stimulate the production of ceramide via sphingomyelinases to limit cancer cell survival. On the contrary, dysfunctional sphingolipid metabolism, a prominent factor in cancer survival and therapy resistance, blunts the anticancer properties of ceramide-orchestrated cell death pathways, especially apoptosis. Although P-glycoprotein (P-gp) is famous for its role in chemotherapy resistance, herein, we propose alternate interpretations and discuss the capacity of this multidrug transporter as a "ceramide neutralizer", an unwelcome event, highlighting yet another facet of P-gp's versatility in drug resistance. We introduce sphingolipid metabolism and its dysfunctional regulation in cancer, present a summary of factors that contribute to chemotherapy resistance, explain how P-gp "neutralizes" ceramide by hastening its glycosylation, and consider therapeutic applications of the P-gp-ceramide connection in the treatment of cancer.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA;
| | - Miki Kassai
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, The East Carolina Diabetes and Obesity Institute, Greenville, NC 27834, USA;
| | - Su-Fern Tan
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Thomas P. Loughran
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - David J. Feith
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Myles C. Cabot
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, The East Carolina Diabetes and Obesity Institute, Greenville, NC 27834, USA;
| |
Collapse
|
29
|
Wu Y, Zhang J, Tian Y, Chi Shing Cho W, Xu HX, Lin ZX, Xian YF. 20(S)-Ginsenoside Rh2 overcomes gemcitabine resistance in pancreatic cancer by inhibiting LAMC2-Modulated ABC transporters. J Adv Res 2024:S2090-1232(24)00390-4. [PMID: 39270979 DOI: 10.1016/j.jare.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
INTRODUCTION Gemcitabine (GEM) is the first-line drug for pancreatic ductal adenocarcinoma (PDAC), but drug resistance severely restricts its chemotherapeutic efficacy. Laminin subunit γ2 (LAMC2) plays a crucial role in extracellular matrix formation in the development of GEM-resistance. However, the biological function of LAMC2 in GEM resistance and its molecular mechanisms are still unclear. 20(S)-Ginsenoside Rh2 (Rh2), one of the principal active components isolated from Ginseng Radix et Rhizoma, possesses strong anti-tumor effects. However, the effects of Rh2 on overcoming GEM resistance and its action mechanisms remain to be elucidated. OBJECTIVES This study aimed to determine the efficacy of Rh2 on overcoming GEM resistance and to explore its underlying molecular mechanisms. METHODS Clinical study, Western blotting, publicly available databasesand bioinformatic analyses were performed to investigate the protein expression of LAMC2 in the GEM-resistant PDAC patients and the acquired GEM-resistant PDAC cells. Then, the effects of Rh2 on overcoming the GEM resistance in PDAC were evaluated both in vitro and in vivo. Stable silencing or overexpression of LAMC2 in the GEM-resistant PDAC cells were established for validating the role of LAMC2 on Rh2 overcoming the GEM resistance in PDAC. RESULTS The protein expression of LAMC2 was markedly increased in the GEM-resistant PDAC patient biopsies compared to the sensitive cases. The protein expression of LAMC2 was significantly higher in the acquired GEM-resistant PDAC cells than that in their parental cells. Rh2 enhanced the chemosensitivity of GEM in the GEM-resistant PDAC cells, and inhibited the tumor growth of Miapaca-2-GR cell-bearing mice and Krastm4TyjTrp53tm1BrnTg (Pdx1-cre/Esr1*) #Dam/J (KPC) mice. Rh2 effectively reversed the GEM resistance in Miapaca-2-GR and Capan-2-GR cells by inhibiting LAMC2 expression through regulating the ubiquitin-proteasome pathway. Knockdown of LAMC2 enhanced the chemosensitivity of GEM and the effects of Rh2 on overcoming the GEM resistance in PDAC cells and the orthotopic PDAC mouse model. Conversely, LAMC2 overexpression aggravated the chemoresistance of GEM and abolished the effects of Rh2 on overcoming GEM resistance via modulating ATP-binding cassette (ABC) transporters leading to the active GEM efflux. CONCLUSIONS LAMC2 plays an important role in the GEM resistance in PDAC, and Rh2 is a potential adjuvant for overcoming the chemoresistance of GEM in PDAC.
Collapse
Affiliation(s)
- Yulin Wu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Juan Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Yuanyang Tian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China; Department of Pathology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | - Hong-Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China; Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China; The Chinese University of Hong Kong Chinese Medicine Specialty Clinic cum Clinical Teaching and Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China; The Chinese University of Hong Kong Chinese Medicine Specialty Clinic cum Clinical Teaching and Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
30
|
Guo Y, Ashrafizadeh M, Tambuwala MM, Ren J, Orive G, Yu G. P-glycoprotein (P-gp)-driven cancer drug resistance: biological profile, non-coding RNAs, drugs and nanomodulators. Drug Discov Today 2024; 29:104161. [PMID: 39245345 DOI: 10.1016/j.drudis.2024.104161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Drug resistance has compromised the efficacy of chemotherapy. The dysregulation of drug transporters including P-glycoprotein (P-gp) can mediate drug resistance through drug efflux. In this review, we highlight the role of P-gp in cancer drug resistance and the related molecular pathways, including phosphoinositide 3-kinase (PI3K)-Akt, phosphatase and tensin homolog (PTEN) and nuclear factor-κB (NF-κB), along with non-coding RNAs (ncRNAs). Extracellular vesicles secreted by the cells can transport ncRNAs and other proteins to change P-gp activity in cancer drug resistance. P-gp requires ATP to function, and the induction of mitochondrial dysfunction or inhibition of glutamine metabolism can impair P-gp function, thus increasing chemosensitivity. Phytochemicals, small molecules and nanoparticles have been introduced as P-gp inhibitors to increase drug sensitivity in human cancers.
Collapse
Affiliation(s)
- Yang Guo
- Department of Respiratory and Critical Care Medicine, Shenyang Tenth People's Hospital (Shenyang Chest Hospital), No. 11 Beihai Street, Dadong District, Shenyang 110044, Liaoning, China
| | - Milad Ashrafizadeh
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore.
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, No. 163 Shoushan Road, Jiangyin, China.
| |
Collapse
|
31
|
Dong W, Wang W, Cao C. The Evolution of Antibody-Drug Conjugates: Toward Accurate DAR and Multi-specificity. ChemMedChem 2024; 19:e202400109. [PMID: 38758596 DOI: 10.1002/cmdc.202400109] [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: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
Antibody-drug conjugates (ADCs) consist of antibodies, linkers and payloads. They offer targeted delivery of potent cytotoxic drugs to tumor cells, minimizing off-target effects. However, the therapeutic efficacy of ADCs is compromised by heterogeneity in the drug-to-antibody ratio (DAR), which impacts both cytotoxicity and pharmacokinetics (PK). Additionally, the emergence of drug resistance poses significant challenges to the clinical advancement of ADCs. To overcome these limitations, a variety of strategies have been developed, including the design of multi-specific drugs with accurate DAR. This review critically summarizes the current challenges faced by ADCs, categorizing key issues and evaluating various innovative solutions. We provide an in-depth analysis of the latest methodologies for achieving homogeneous DAR and explore design strategies for multi-specific drugs aimed at combating drug resistance. Our discussion offers a current perspective on the advancements made in refining ADC technologies, with an emphasis on enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Wenge Dong
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wanqi Wang
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chan Cao
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
32
|
Salazar PB, Murakami M, Ranganathan N, Durell SR, Ambudkar SV. Mutational analysis reveals the importance of residues of the access tunnel inhibitor site to human P-glycoprotein (ABCB1)-mediated transport. Protein Sci 2024; 33:e5155. [PMID: 39194126 DOI: 10.1002/pro.5155] [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: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024]
Abstract
Human P-glycoprotein (P-gp) utilizes energy from ATP hydrolysis for the efflux of chemically dissimilar amphipathic small molecules and plays an important role in the development of resistance to chemotherapeutic agents in most cancers. Efforts to overcome drug resistance have focused on inhibiting P-gp-mediated drug efflux. Understanding the features distinguishing P-gp inhibitors from substrates is critical. Cryo-electron microscopy has revealed distinct binding patterns, emphasizing the role of the L-site or access tunnel in inhibition. We substituted 5-9 residues of the L-site with alanine to investigate whether the binding of a second inhibitor molecule to the L-site is required for inhibiting drug efflux. We reveal, for the first time, that mutations in the L-site affect the drug efflux activity of P-gp, despite their distance from the substrate-binding pocket (SBP). Surprisingly, after the mutations were introduced, inhibitors such as tariquidar and zosuquidar still inhibited drug efflux by mutant P-gps. Communication between the transmembrane helices (TMHs) and nucleotide-binding domains (NBDs) was evaluated using the ATPase assay, revealing distinct modulation patterns by inhibitors for the mutants, with zosuquidar exhibiting substrate-like stimulation of ATPase. Furthermore, L-site mutations abolished ATP-dependent thermal stabilization. In silico molecular docking studies corroborated the altered inhibitor binding due to mutations in the L-site residues, shedding light on their critical role in substrate transport and inhibitor interactions with P-gp. These findings suggest that inhibitors bind either to the SBP alone, and/or to alternate site(s) when the L-site is disabled by mutagenesis.
Collapse
Affiliation(s)
- Paula B Salazar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Megumi Murakami
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nandhini Ranganathan
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stewart R Durell
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Arakawa Y, Elloumi F, Varma S, Khandagale P, Jo U, Kumar S, Roper N, Reinhold WC, Robey RW, Takebe N, Gottesman MM, Thomas CJ, Boeva V, Berruti A, Abate A, Tamburello M, Sigala S, Hantel C, Weigand I, Wierman ME, Kiseljak-Vassiliades K, Del Rivero J, Pommier Y. A Database Tool Integrating Genomic and Pharmacologic Data from Adrenocortical Carcinoma Cell Lines, PDX, and Patient Samples. CANCER RESEARCH COMMUNICATIONS 2024; 4:2384-2398. [PMID: 39162009 PMCID: PMC11389377 DOI: 10.1158/2767-9764.crc-24-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/07/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Adrenocortical carcinoma (ACC) is a rare and highly heterogeneous disease with a notably poor prognosis due to significant challenges in diagnosis and treatment. Emphasizing on the importance of precision medicine, there is an increasing need for comprehensive genomic resources alongside well-developed experimental models to devise personalized therapeutic strategies. We present ACC_CellMinerCDB, a substantive genomic and drug sensitivity database (available at https://discover.nci.nih.gov/acc_cellminercdb) comprising ACC cell lines, patient-derived xenografts, surgical samples, and responses to more than 2,400 drugs examined by the NCI and National Center for Advancing Translational Sciences. This database exposes shared genomic pathways among ACC cell lines and surgical samples, thus authenticating the cell lines as research models. It also allows exploration of pertinent treatment markers such as MDR-1, SOAT1, MGMT, MMR, and SLFN11 and introduces the potential to repurpose agents like temozolomide for ACC therapy. ACC_CellMinerCDB provides the foundation for exploring larger preclinical ACC models. SIGNIFICANCE ACC_CellMinerCDB, a comprehensive database of cell lines, patient-derived xenografts, surgical samples, and drug responses, reveals shared genomic pathways and treatment-relevant markers in ACC. This resource offers insights into potential therapeutic targets and the opportunity to repurpose existing drugs for ACC therapy.
Collapse
Affiliation(s)
- Yasuhiro Arakawa
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Fathi Elloumi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sudhir Varma
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Prashant Khandagale
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ukhyun Jo
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Suresh Kumar
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nitin Roper
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William C. Reinhold
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Naoko Takebe
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Valentina Boeva
- Department of Computer Science, Institute for Machine Learning, ETH Zurich, Zurich, Switzerland
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Medical Oncology Unit, University of Brescia, Azienda Socio Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, and University of Zurich, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Isabel Weigand
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Margaret E. Wierman
- Department of Medicine-Endocrinology/Metabolism/Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Katja Kiseljak-Vassiliades
- Department of Medicine-Endocrinology/Metabolism/Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
34
|
Das V, Miller JH, Alladi CG, Annadurai N, De Sanctis JB, Hrubá L, Hajdúch M. Antineoplastics for treating Alzheimer's disease and dementia: Evidence from preclinical and observational studies. Med Res Rev 2024; 44:2078-2111. [PMID: 38530106 DOI: 10.1002/med.22033] [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: 03/02/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
As the world population ages, there will be an increasing need for effective therapies for aging-associated neurodegenerative disorders, which remain untreatable. Dementia due to Alzheimer's disease (AD) is one of the leading neurological diseases in the aging population. Current therapeutic approaches to treat this disorder are solely symptomatic, making the need for new molecular entities acting on the causes of the disease extremely urgent. One of the potential solutions is to use compounds that are already in the market. The structures have known pharmacokinetics, pharmacodynamics, toxicity profiles, and patient data available in several countries. Several drugs have been used successfully to treat diseases different from their original purposes, such as autoimmunity and peripheral inflammation. Herein, we divulge the repurposing of drugs in the area of neurodegenerative diseases, focusing on the therapeutic potential of antineoplastics to treat dementia due to AD and dementia. We briefly touch upon the shared pathological mechanism between AD and cancer and drug repurposing strategies, with a focus on artificial intelligence. Next, we bring out the current status of research on the development of drugs, provide supporting evidence from retrospective, clinical, and preclinical studies on antineoplastic use, and bring in new areas, such as repurposing drugs for the prion-like spreading of pathologies in treating AD.
Collapse
Affiliation(s)
- Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - John H Miller
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Charanraj Goud Alladi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Lenka Hrubá
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
35
|
Hsu CY, Faisal A, Jumaa SS, Gilmanova NS, Ubaid M, Athab AH, Mirzaei R, Karampoor S. Exploring the impact of circRNAs on cancer glycolysis: Insights into tumor progression and therapeutic strategies. Noncoding RNA Res 2024; 9:970-994. [PMID: 38770106 PMCID: PMC11103225 DOI: 10.1016/j.ncrna.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/18/2024] [Accepted: 05/04/2024] [Indexed: 05/22/2024] Open
Abstract
Cancer cells exhibit altered metabolic pathways, prominently featuring enhanced glycolytic activity to sustain their rapid growth and proliferation. Dysregulation of glycolysis is a well-established hallmark of cancer and contributes to tumor progression and resistance to therapy. Increased glycolysis supplies the energy necessary for increased proliferation and creates an acidic milieu, which in turn encourages tumor cells' infiltration, metastasis, and chemoresistance. Circular RNAs (circRNAs) have emerged as pivotal players in diverse biological processes, including cancer development and metabolic reprogramming. The interplay between circRNAs and glycolysis is explored, illuminating how circRNAs regulate key glycolysis-associated genes and enzymes, thereby influencing tumor metabolic profiles. In this overview, we highlight the mechanisms by which circRNAs regulate glycolytic enzymes and modulate glycolysis. In addition, we discuss the clinical implications of dysregulated circRNAs in cancer glycolysis, including their potential use as diagnostic and prognostic biomarkers. All in all, in this overview, we provide the most recent findings on how circRNAs operate at the molecular level to control glycolysis in various types of cancer, including hepatocellular carcinoma (HCC), prostate cancer (PCa), colorectal cancer (CRC), cervical cancer (CC), glioma, non-small cell lung cancer (NSCLC), breast cancer, and gastric cancer (GC). In conclusion, this review provides a comprehensive overview of the significance of circRNAs in cancer glycolysis, shedding light on their intricate roles in tumor development and presenting innovative therapeutic avenues.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City, 71710, Taiwan
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, 85004, USA
| | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | - Sally Salih Jumaa
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Nataliya Sergeevna Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia, Moscow
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Aya H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Rasoul Mirzaei
- Venom & Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal & Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Belhadj Z, Akther T, Wang Z, Xie J. Characterization of a deazaflavin analog as a potent inhibitor of multidrug resistance-associated protein 1. Biomed Pharmacother 2024; 178:117167. [PMID: 39032285 DOI: 10.1016/j.biopha.2024.117167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Selective inhibition of overexpressed ATP binding cassette (ABC) transporters is an attractive approach to enhancing the efficacy of chemotherapeutics in multidrug resistant cancers. Previously, we reported that the cancer sensitizing effect of deazaflavin analogs, an important chemotype for developing combination treatments with topoisomerase II (TOP2) poisons, is associated with increased intracellular drug accumulation. Here we report the characterization of ZW-1226, a deazaflavin analog, as a potent inhibitor of multidrug resistance-associated protein 1 (MRP1). Specifically, ZW-1226 inhibited MRP1 with a 16-fold higher potency than the most widely used positive control MK-571 in vesicular transport assay and displayed excellent selectivity indices exceeding 100 over other major ABC transporters, including P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), MRP2 and MRP3. Mechanistically, we revealed that its MRP1 inhibitory action requires the participation of GSH. In chemo-sensitization test, ZW-1226 fully reversed the MRP1-mediated drug resistance to TOP2 poisons etoposide (ETP) and doxorubicin (DOX) in H69AR cells and conferred CC50s comparable to those in the sensitive parental NCI-H69 cells. The sensitization was associated with boosted intracellular accumulation of ETP and DOX and elevated endogenous GSH. Moreover, ZW-1226 showed potential to occupy the leukotriene C4 binding site in molecular docking with bovine MRP1, presumably with the help of GSH. Lastly, ZW-1226 exhibited high tissue to plasma partitions in mice but did not alter ETP distribution to normal tissues, suggesting it could be a viable lead with desirable pharmacokinetic properties to warrant further investigation.
Collapse
Affiliation(s)
- Zakia Belhadj
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thamina Akther
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
37
|
Baeza-Morales A, Medina-García M, Martínez-Peinado P, Pascual-García S, Pujalte-Satorre C, López-Jaén AB, Martínez-Espinosa RM, Sempere-Ortells JM. The Antitumour Mechanisms of Carotenoids: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1060. [PMID: 39334719 PMCID: PMC11428676 DOI: 10.3390/antiox13091060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Carotenoids, known for their antioxidant properties, have garnered significant attention for their potential antitumour activities. This comprehensive review aims to elucidate the diverse mechanisms by which carotenoids exert antitumour effects, focusing on both well-established and novel findings. We explore their role in inducing apoptosis, inhibiting cell cycle progression and preventing metastasis by affecting oncogenic and tumour suppressor proteins. The review also explores the pro-oxidant function of carotenoids within cancer cells. In fact, although their overall contribution to cellular antioxidant defences is well known and significant, some carotenoids can exhibit pro-oxidant effects under certain conditions and are able to elevate reactive oxygen species (ROS) levels in tumoural cells, triggering mitochondrial pathways that would lead to cell death. The final balance between their antioxidant and pro-oxidant activities depends on several factors, including the specific carotenoid, its concentration and the redox environment of the cell. Clinical trials are discussed, highlighting the conflicting results of carotenoids in cancer treatment and the importance of personalized approaches. Emerging research on rare carotenoids like bacterioruberin showcases their superior antioxidant capacity and selective cytotoxicity against aggressive cancer subtypes, such as triple-negative breast cancer. Future directions include innovative delivery systems, novel combinations and personalized treatments, aiming to enhance the therapeutic potential of carotenoids. This review highlights the promising yet complex landscape of carotenoid-based cancer therapies, calling for continued research and clinical exploration.
Collapse
Affiliation(s)
- Andrés Baeza-Morales
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Miguel Medina-García
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Pascual Martínez-Peinado
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Sandra Pascual-García
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Carolina Pujalte-Satorre
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Ana Belén López-Jaén
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Rosa María Martínez-Espinosa
- Biochemistry and Molecular Biology and Edaphology and Agricultural Chemistry Department, Faculty of Sciences, University of Alicante, Ap. 99, E-03080 Alicante, Spain;
- Applied Biochemistry Research Group, Multidisciplinary Institute for Environmental Studies “Ramón Margalef”, University of Alicante, Ap. 99, E-03080 Alicante, Spain
| | - José Miguel Sempere-Ortells
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| |
Collapse
|
38
|
Mariya Vincent D, Mostafa H, Suneer A, Radha Krishnan S, Ong M, Itahana Y, Itahana K, Viswanathan R. Development of Natural-Product-Inspired ABCB1 Inhibitors Through Regioselective Tryptophan C3-Benzylation. Chemistry 2024:e202401782. [PMID: 39190779 DOI: 10.1002/chem.202401782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 08/29/2024]
Abstract
The emergence of drug resistance in cancer cells eventually causing relapse is a serious threat that demands new advances. Upregulation of the ATP-dependent binding cassette (ABC) transporters, such as ABCB1, significantly contributes to the emergence of drug resistance in cancer. Despite more than 30 years of therapeutic discovery, and several generations of inhibitors against P-gp, the search for effective agents that minimize toxicity to human cells, while maintaining efflux pump inhibition is still underway. Leads derived from natural product scaffolds are well-known to be effective in various therapeutic approaches. Inspired by the biosynthetic pathway to Nocardioazine A, a marine alkaloid known to inhibit the P-gp efflux pump in cancer cells, we devised a regioselective pathway to create structurally unique indole-C3-benzyl cyclo-L-Trp-L-Trp diketopiperazines (DKPs). Using bat cells as a model to derive effective ABCB1 inhibitors for targeting human P-gp efflux pumps, we have recently identified exo-C3-N-Dbn-Trp2 (13) as a lead ABCB1 inhibitor. This C3-benzylated lead inhibited ABCB1 better than Verapamil.[21] Additionally, C3-N-Dbn-Trp2 restored chemotherapy sensitivity in drug-resistant human cancer cells and had no adverse effect on cell proliferation in cell cultures. For a clearer structure-activity relationship, we developed a broader screen to test C3-functionalized pyrroloindolines as ABCB1 inhibitors and observed that C3-benzylation is outperforming respective isoprenylated derivatives. Results arising from the molecular docking studies indicate that the interactions at the access tunnel between ABCB1 and the inhibitor result in a powerful predictor for the efficacy of the inhibitor. Based on fluorescence-based assays, we conclude that the most efficacious inhibitor is the p-cyano-derived exo-C3-N-Dbn-Trp2 (33 a), closely followed by the p-nitro substituted analogue. By combining assay results with molecular docking studies, we further correlate that the predictions based on the inhibitor interactions at the access tunnel provide clues about the design of improved ABCB1 inhibitors. As it has been well documented that ABCB1 itself is powerfully engaged in multi-drug resistance, this work lays the foundation for the design of a new class of inhibitors based on the endogenous amino acid-derived cyclo-L-Trp-L-Trp DKP scaffold.
Collapse
Affiliation(s)
- Dona Mariya Vincent
- Departments of Chemistry & Biology, Indian Institute of Science Education and Research, Tirupati, A. P., India
| | - Habib Mostafa
- Departments of Chemistry & Biology, Indian Institute of Science Education and Research, Tirupati, A. P., India
| | - Anza Suneer
- Departments of Chemistry & Biology, Indian Institute of Science Education and Research, Tirupati, A. P., India
| | | | - Mingmin Ong
- Programme in Cancer & Stem Cell Biology, Duke-NUS Medical School, 169857, Singapore, Singapore
| | - Yoko Itahana
- Programme in Cancer & Stem Cell Biology, Duke-NUS Medical School, 169857, Singapore, Singapore
| | - Koji Itahana
- Programme in Cancer & Stem Cell Biology, Duke-NUS Medical School, 169857, Singapore, Singapore
| | - Rajesh Viswanathan
- Departments of Chemistry & Biology, Indian Institute of Science Education and Research, Tirupati, A. P., India
| |
Collapse
|
39
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2024. [PMID: 39180410 DOI: 10.1002/med.22077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
40
|
van Gelder M, Li Y, Wander DPA, Berlin I, Overkleeft HS, van der Zanden SY, Neefjes JJC. Novel N,N-Dimethyl-idarubicin Analogues Are Effective Cytotoxic Agents for ABCB1-Overexpressing, Doxorubicin-Resistant Cells. J Med Chem 2024; 67:13802-13812. [PMID: 39088428 PMCID: PMC11345819 DOI: 10.1021/acs.jmedchem.4c00614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Anthracyclines comprise one of the most effective anticancer drug classes. Doxorubicin, daunorubicin, epirubicin, and idarubicin have been in clinical use for decades, but their application remains complicated by treatment-related toxicities and drug resistance. We previously demonstrated that the combination of DNA damage and histone eviction exerted by doxorubicin drives its associated adverse effects. However, whether the same properties dictate drug resistance is unclear. In the present study, we evaluate a library of 40 anthracyclines on their cytotoxicity, intracellular uptake, and subcellular localization in K562 wildtype versus ABCB1-transporter-overexpressing, doxorubicin-resistant cells. We identify several highly potent cytotoxic anthracyclines. Among these, N,N-dimethyl-idarubicin and anthracycline (composed of the idarubicin aglycon and the aclarubicin trisaccharide) stand out, due to their histone eviction-mediated cytotoxicity toward doxorubicin-resistant cells. Our findings thus uncover understudied anthracycline variants warranting further investigation in the quest for safer and more effective anticancer agents that circumvent cellular export by ABCB1.
Collapse
Affiliation(s)
- Merle
A. van Gelder
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 CZ Leiden, The Netherlands
| | - Yufeng Li
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 CZ Leiden, The Netherlands
| | - Dennis P. A. Wander
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 CZ Leiden, The Netherlands
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Ilana Berlin
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 CZ Leiden, The Netherlands
| | - Hermen S. Overkleeft
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Sabina Y. van der Zanden
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 CZ Leiden, The Netherlands
| | - Jacques J. C. Neefjes
- Department
of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, Einthovenweg 20, 2333 CZ Leiden, The Netherlands
| |
Collapse
|
41
|
Kruschel RD, Barbosa MG, Almeida MJ, Xavier CPR, Vasconcelos MH, McCarthy FO. Discovery of Potent Isoquinolinequinone N-Oxides to Overcome Cancer Multidrug Resistance. J Med Chem 2024; 67:13909-13924. [PMID: 39093920 PMCID: PMC11345829 DOI: 10.1021/acs.jmedchem.4c00705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
Multidrug resistance (MDR) of human tumors has resulted in an immediate need to develop appropriate new drugs. This work outlines the development of 20 potent IQQ N-oxide derivatives in two isomeric families, both exhibiting nanomolar GI50 against human tumor cell lines. Preliminary NCI-60 tumor screening sees the C(6) isomers achieve a mean GI50 > 2 times lower than the corresponding C(7) isomers. MDR evaluation of nine selected compounds reveals that each presents lower GI50 concentrations in two MDR tumor cell lines. Four of the series display nanomolar GI50 values against MDR cells, having selectivity ratios up to 2.7 versus the sensitive (parental) cells. The most potent compound 25 inhibits the activity of drug efflux pumps in MDR cells, causes significant ROS accumulation, and potently inhibits cell proliferation, causing alterations in the cell cycle profile. Our findings are confirmed by 3D spheroid models, providing new candidates for studies against MDR cancers.
Collapse
Affiliation(s)
- Ryan D. Kruschel
- School
of Chemistry, Analytical and Biological Chemistry Research Facility, University College Cork, Cork T12 K8AF, Ireland
| | - Mélanie
A. G. Barbosa
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
- FFUP−Faculty
of Pharmacy of the University of Porto, 4050-313 Porto Portugal
| | - Maria João Almeida
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
| | - Cristina P. R. Xavier
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
| | - M. Helena Vasconcelos
- i3S−Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto Portugal
- Cancer
Drug Resistance Group, IPATIMUP−Institute of Molecular Pathology
and Immunology, University of Porto, 4200-135 Porto Portugal
- FFUP−Faculty
of Pharmacy of the University of Porto, 4050-313 Porto Portugal
| | - Florence O. McCarthy
- School
of Chemistry, Analytical and Biological Chemistry Research Facility, University College Cork, Cork T12 K8AF, Ireland
| |
Collapse
|
42
|
Marangoni-Iglecias L, Rojo-Tolosa S, Márquez-Pete N, Cura Y, Moreno-Toro N, Membrive-Jiménez C, Sánchez-Martin A, Pérez-Ramírez C, Jiménez-Morales A. Precision Medicine in Childhood Cancer: The Influence of Genetic Polymorphisms on Vincristine-Induced Peripheral Neuropathy. Int J Mol Sci 2024; 25:8797. [PMID: 39201483 PMCID: PMC11354794 DOI: 10.3390/ijms25168797] [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: 07/23/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer is the leading cause of disease-related death among children. Vincristine (VCR), a key component of childhood cancer treatment protocols, is associated with the risk of peripheral neuropathy (PN), a condition that may be reversible upon drug discontinuation but can also leave lasting sequelae. Single nucleotide polymorphism (SNP) in genes involved in VCR pharmacokinetics and pharmacodynamics have been investigated in relation to an increased risk of PN. However, the results of these studies have been inconsistent. A retrospective cohort study was conducted to investigate the potential association of drug transporter genes from the ATP-binding cassette (ABC) family and the centrosomal protein 72 (CEP72) gene with the development of PN in 88 Caucasian children diagnosed with cancer and treated with VCR. Genotyping was performed using real-time PCR techniques for the following SNPs: ABCB1 rs1128503, ABCC1 rs246240, ABCC2 rs717620, and CEP72 rs924607. The results indicated that age at diagnosis (OR = 1.33; 95% CI = 1.07-1.75) and the ABCC1 rs246240 G allele (OR = 12.48; 95% CI = 2.26-100.42) were associated with vincristine-induced peripheral neuropathy (VIPN). No association was found between this toxicity and CEP72 rs924607. Our study provides insights that may contribute to optimizing childhood cancer therapy in the future by predicting the risk of VIPN.
Collapse
Affiliation(s)
- Luciana Marangoni-Iglecias
- Clinical Analysis Laboratory Unit, Hospital Universitário Maria Aparecida Pedrossian HUMAP-UFMS, Av. Sen. Filinto Müler, 355, Vila Ipiranga, Campo Grande 79080-190, Brazil;
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
| | - Susana Rojo-Tolosa
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
- Biosanitary Research Institute ibs.GRANADA, Avda. de Madrid 15, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18011 Granada, Spain
- Pneumology Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain
| | - Noelia Márquez-Pete
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
- Biosanitary Research Institute ibs.GRANADA, Avda. de Madrid 15, 18012 Granada, Spain
| | - Yasmín Cura
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18011 Granada, Spain
| | - Noelia Moreno-Toro
- Pediatric Service, Santa Ana Regional Hospital, Av. Enrique Martín Cuevas, s/n, Motril, 18600 Granada, Spain;
| | - Cristina Membrive-Jiménez
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
- Biosanitary Research Institute ibs.GRANADA, Avda. de Madrid 15, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18011 Granada, Spain
| | - Almudena Sánchez-Martin
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
| | - Cristina Pérez-Ramírez
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
- Biosanitary Research Institute ibs.GRANADA, Avda. de Madrid 15, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18011 Granada, Spain
| | - Alberto Jiménez-Morales
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain (Y.C.); (C.M.-J.); (A.S.-M.); (C.P.-R.); (A.J.-M.)
- Biosanitary Research Institute ibs.GRANADA, Avda. de Madrid 15, 18012 Granada, Spain
| |
Collapse
|
43
|
Kurre D, Dang PX, Le LT, Gadkari VV, Alam A. Structural insight into binding site access and ligand recognition by human ABCB1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607598. [PMID: 39185192 PMCID: PMC11343101 DOI: 10.1101/2024.08.12.607598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
ABCB1 is a broad-spectrum efflux pump central to cellular drug handling and multidrug resistance in humans. However, its mechanisms of poly-specific substrate recognition and transport remain poorly resolved. Here we present cryo-EM structures of lipid embedded human ABCB1 in its apo, substrate-bound, inhibitor-bound, and nucleotide-trapped states at 3.4-3.9 Å resolution without using stabilizing antibodies or mutations and each revealing a distinct conformation. The substrate binding site is located within one half of the molecule and, in the apo state, is obstructed by transmembrane helix (TM) 4. Substrate and inhibitor binding are distinguished by major differences in TM arrangement and ligand binding chemistry, with TM4 playing a central role in all conformational transitions. Our data offer fundamental new insights into the role structural asymmetry, secondary structure breaks, and lipid interactions play in ABCB1 function and have far-reaching implications for ABCB1 inhibitor design and predicting its substrate binding profiles.
Collapse
Affiliation(s)
- Devanshu Kurre
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
| | - Phuoc X. Dang
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
- Current Address: Department of Pharmacy - Inpatient, Mayo Clinic, Rochester, Minnesota 55901, United States
| | - Le T.M. Le
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
- Current Address: Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55901, United States
| | - Varun V. Gadkari
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Amer Alam
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, United States
| |
Collapse
|
44
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
45
|
Lu Q, Ambudkar SV, Yang DH. Editorial: ABC transporters and drug resistance. Drug Resist Updat 2024; 77:101135. [PMID: 39178712 DOI: 10.1016/j.drup.2024.101135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Affiliation(s)
- Qisi Lu
- The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola 11501, USA.
| |
Collapse
|
46
|
La Banca V, De Domenico S, Nicolai S, Gatti V, Scalera S, Maugeri M, Mauriello A, Montanaro M, Pahnke J, Candi E, D’Amico S, Peschiaroli A. ABCC1 Is a ΔNp63 Target Gene Overexpressed in Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:8741. [PMID: 39201428 PMCID: PMC11354449 DOI: 10.3390/ijms25168741] [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: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
The transcription factor ΔNp63 plays a pivotal role in maintaining the integrity of stratified epithelial tissues by regulating the expression of distinct target genes involved in lineage specification, cell stemness, cell proliferation and differentiation. Here, we identified the ABC transporter subfamily member ABCC1 as a novel ΔNp63 target gene. We found that in immortalized human keratinocytes and in squamous cell carcinoma (SCC) cells, ∆Np63 induces the expression of ABCC1 by physically occupying a p63-binding site (p63 BS) located in the first intron of the ABCC1 gene locus. In cutaneous SCC and during the activation of the keratinocyte differentiation program, ∆Np63 and ABCC1 levels are positively correlated raising the possibility that ABCC1 might be involved in the regulation of the proliferative/differentiative capabilities of squamous tissue. However, we did not find any gross alteration in the structure and morphology of the epidermis in humanized hABCC1 knock-out mice. Conversely, we found that the genetic ablation of ABCC1 led to a marked reduction in inflammation-mediated proliferation of keratinocytes, suggesting that ABCC1 might be involved in the regulation of keratinocyte proliferation upon inflammatory/proliferative signals. In line with these observations, we found a significant increase in ABCC1 expression in squamous cell carcinomas (SCCs), a tumor type characterized by keratinocyte hyper-proliferation and a pro-inflammatory tumor microenvironment. Collectively, these data uncover ABCC1 as an additional ∆Np63 target gene potentially involved in those skin diseases characterized by dysregulation of proliferation/differentiation balance.
Collapse
Affiliation(s)
- Veronica La Banca
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
| | - Sara De Domenico
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
| | - Sara Nicolai
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| | - Veronica Gatti
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| | - Stefano Scalera
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.S.); (M.M.)
| | - Marcello Maugeri
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.S.); (M.M.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
| | - Manuela Montanaro
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab/Section of Neuropathology Research, Department of Pathology (PAT), Medical Faculty/Clinical Medicine (KlinMed), Clinics for Laboratory Medicine (KLM), University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway;
- Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany
- Department of Pharmacology, The Faculty of Medicine and Life Sciences, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Tel Aviv 6997801, Israel
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00166 Rome, Italy
| | - Silvia D’Amico
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| | - Angelo Peschiaroli
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| |
Collapse
|
47
|
Akman M, Monteleone C, Doronzo G, Godel M, Napoli F, Merlini A, Campani V, Nele V, Balmas E, Chontorotzea T, Fontana S, Digiovanni S, Barbu FA, Astanina E, Jafari N, Salaroglio IC, Kopecka J, De Rosa G, Mohr T, Bertero A, Righi L, Novello S, Scagliotti GV, Bussolino F, Riganti C. TFEB controls sensitivity to chemotherapy and immuno-killing in non-small cell lung cancer. J Exp Clin Cancer Res 2024; 43:219. [PMID: 39107857 PMCID: PMC11304671 DOI: 10.1186/s13046-024-03142-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND In non-small cell lung cancer (NSCLC) the efficacy of chemo-immunotherapy is affected by the high expression of drug efflux transporters as ABCC1 and by the low expression of ABCA1, mediating the isopentenyl pyrophosphate (IPP)-dependent anti-tumor activation of Vγ9Vδ2 T-lymphocytes. In endothelial cells ABCA1 is a predicted target of the transcription factor EB (TFEB), but no data exists on the correlation between TFEB and ABC transporters involved in the chemo-immuno-resistance in NSCLC. METHODS The impact of TFEB/ABCC1/ABCA1 expression on NSCLC patients' survival was analyzed in the TCGA-LUAD cohort and in a retrospective cohort of our institution. Human NSCLC cells silenced for TFEB (shTFEB) were analyzed for ABC transporter expression, chemosensitivity and immuno-killing. The chemo-immuno-sensitizing effects of nanoparticles encapsulating zoledronic acid (NZ) on shTFEB tumors and on tumor immune-microenvironment were evaluated in Hu-CD34+ mice by single-cell RNA-sequencing. RESULTS TFEBlowABCA1lowABCC1high and TFEBhighABCA1highABCC1low NSCLC patients had the worst and the best prognosis, respectively, in the TCGA-LUAD cohort and in a retrospective cohort of patients receiving platinum-based chemotherapy or immunotherapy as first-line treatment. By silencing shTFEB in NSCLC cells, we demonstrated that TFEB was a transcriptional inducer of ABCA1 and a repressor of ABCC1. shTFEB cells had also a decreased activity of ERK1/2/SREBP2 axis, implying reduced synthesis and efflux via ABCA1 of cholesterol and its intermediate IPP. Moreover, TFEB silencing reduced cholesterol incorporation in mitochondria: this event increased the efficiency of OXPHOS and the fueling of ABCC1 by mitochondrial ATP. Accordingly, shTFEB cells were less immuno-killed by the Vγ9Vδ2 T-lymphocytes activated by IPP and more resistant to cisplatin. NZ, which increased IPP efflux but not OXPHOS and ATP production, sensitized shTFEB immuno-xenografts, by reducing intratumor proliferation and increasing apoptosis in response to cisplatin, and by increasing the variety of anti-tumor infiltrating cells (Vγ9Vδ2 T-lymphocytes, CD8+T-lymphocytes, NK cells). CONCLUSIONS This work suggests that TFEB is a gatekeeper of the sensitivity to chemotherapy and immuno-killing in NSCLC, and that the TFEBlowABCA1lowABCC1high phenotype can be predictive of poor response to chemotherapy and immunotherapy. By reshaping both cancer metabolism and tumor immune-microenvironment, zoledronic acid can re-sensitize TFEBlow NSCLCs, highly resistant to chemo- and immunotherapy.
Collapse
Affiliation(s)
- Muhlis Akman
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Ciro Monteleone
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Gabriella Doronzo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- IRCCS Candiolo Cancer Institute, Candiolo, Italy
| | - Martina Godel
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Francesca Napoli
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | - Alessandra Merlini
- Thoracic Oncology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | - Virginia Campani
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Valeria Nele
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Elisa Balmas
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Tatiana Chontorotzea
- Center for Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Simona Fontana
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Sabrina Digiovanni
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Francesca Alice Barbu
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | | | - Niloufar Jafari
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Iris Chiara Salaroglio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Joanna Kopecka
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Thomas Mohr
- Center for Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Luisella Righi
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | - Silvia Novello
- Thoracic Oncology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | | | - Federico Bussolino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- IRCCS Candiolo Cancer Institute, Candiolo, Italy
| | - Chiara Riganti
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy.
| |
Collapse
|
48
|
Yuan Z, Lei Y, Wan B, Yang M, Jiang Y, Tian C, Wang Z, Wang W. Cadmium exposure elicited dynamic RNA m 6A modification and epi-transcriptomic regulation in the Pacific whiteleg shrimp Litopenaeus vannamei. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101307. [PMID: 39126882 DOI: 10.1016/j.cbd.2024.101307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent post-transcriptional RNA modification in eukaryotic organisms, but its roles in the regulation of physiological resistance of marine crustaceans to heavy metal pollutants are poorly understood. In this study, the transcriptome-wide m6A RNA methylation profiles and dynamic m6A changes induced by acute Cd2+ exposure in the the pacific whiteleg shrimp Litopenaeus vannamei were comprehensively analyzed. Cd2+ toxicity caused a significant reduction in global RNA m6A methylation level, with major m6A regulators including the m6A methyltransferase METTL3 and the m6A binding protein YTHDF2 showing declined expression. Totally, 11,467 m6A methylation peaks from 6415 genes and 17,291 peaks within 7855 genes were identified from the Cd2+ exposure group and the control group, respectively. These m6A peaks were predominantly enriched in the 3' untranslated region (UTR) and around the start codon region of the transcripts. 7132 differentially expressed genes (DEGs) and 7382 differentially m6A-methylated genes (DMGs) were identified. 3186 genes showed significant changes in both gene expression and m6A methylation levels upon cadmium exposure, and they were related to a variety of biological processes and gene pathways. Notably, an array of genes associated with antioxidation homeostasis, transmembrane transporter activity and intracellular detoxification processes were significantly enriched, demonstrating that m6A modification may mediate the physiological responses of shrimp to cadmium toxicity via regulating ROS balance, Cd2+ transport and toxicity mitigation. The study would contribute to a deeper understanding of the evolutionary and functional significance of m6A methylation to the physiological resilience of decapod crustaceans to heavy metal toxicants.
Collapse
Affiliation(s)
- Zhixiang Yuan
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yiguo Lei
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Boquan Wan
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Miao Yang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yue Jiang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Changxu Tian
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhongduo Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China
| | - Wei Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang 524088, China.
| |
Collapse
|
49
|
Lentzas A, de Gooijer MC, Zuidema S, Meurs A, Çitirikkaya CH, Venekamp N, Beijnen JH, van Tellingen O. ATP-binding cassette transporter inhibitor potency and substrate drug affinity are critical determinants of successful drug delivery enhancement to the brain. Fluids Barriers CNS 2024; 21:62. [PMID: 39103921 DOI: 10.1186/s12987-024-00562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Pharmacotherapy for brain diseases is severely compromised by the blood-brain barrier (BBB). ABCB1 and ABCG2 are drug transporters that restrict drug entry into the brain and their inhibition can be used as a strategy to boost drug delivery and pharmacotherapy for brain diseases. METHODS We employed elacridar and tariquidar in mice to explore the conditions for effective inhibition at the BBB. Abcg2;Abcb1a/b knockout (KO), Abcb1a/b KO, Abcg2 KO and wild-type (WT) mice received a 3 h i.p. infusion of a cocktail of 8 typical substrate drugs in combination with elacridar or tariquidar at a range of doses. Abcg2;Abcb1a/b KO mice were used as the reference for complete inhibition, while single KO mice were used to assess the potency to inhibit the remaining transporter. Brain and plasma drug levels were measured by LC-MS/MS. RESULTS Complete inhibition of ABCB1 at the BBB is achieved when the elacridar plasma level reaches 1200 nM, whereas tariquidar requires at least 4000 nM. Inhibition of ABCG2 is more difficult. Elacridar inhibits ABCG2-mediated efflux of weak but not strong ABCG2 substrates. Strikingly, tariquidar does not enhance the brain uptake of any ABCG2-subtrate drug. Similarly, elacridar, but not tariquidar, was able to inhibit its own brain efflux in ABCG2-proficient mice. The plasma protein binding of elacridar and tariquidar was very high but similar in mouse and human plasma, facilitating the translation of mouse data to humans. CONCLUSIONS This work shows that elacridar is an effective pharmacokinetic-enhancer for the brain delivery of ABCB1 and weaker ABCG2 substrate drugs when a plasma concentration of 1200 nM is exceeded.
Collapse
Affiliation(s)
- Aristeidis Lentzas
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M1 3WE, UK
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Stefanie Zuidema
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Amber Meurs
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Ceren H Çitirikkaya
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Nikkie Venekamp
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
- Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
50
|
Gil-Gonzalo R, Durante-Salmerón DA, Pouri S, Doncel-Pérez E, Alcántara AR, Aranaz I, Acosta N. Chitosan-Coated Liposome Formulations for Encapsulation of Ciprofloxacin and Etoposide. Pharmaceutics 2024; 16:1036. [PMID: 39204381 PMCID: PMC11359655 DOI: 10.3390/pharmaceutics16081036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer and bacterial infections rank among the most significant global health threats. accounting for roughly 25 million fatalities each year. This statistic underscores the urgent necessity for developing novel drugs, enhancing current treatments, and implementing systems that boost their bioavailability to achieve superior therapeutic outcomes. Liposomes have been recognised as effective carriers; nonetheless, they encounter issues with long-term stability and structural integrity, which limit their pharmaceutical applicability. Chitosomes (chitosan-coated liposomes) are generally a good alternative to solve these issues. This research aims to demonstrate the effective individual encapsulation of ciprofloxacin (antibacterial, hydrophilic) and etoposide (anticancer, hydrophobic), within chitosomes to create more effective drug delivery systems (oral administration for ciprofloxacin, parenteral administration for etoposide). Thus, liposomes and chitosomes were prepared using the thin-film hydration technique and were characterised through ATR-FTIR, Dynamic Light Scattering (DLS), zeta potential, and release profiling. In both cases, the application of chitosomes enhanced long-term stability in size and surface charge. Chitosome-encapsulated ciprofloxacin formulations exhibited a slower and sustained release profile, while the combined effect of etoposide and chitosan showed heightened efficacy against the glioblastoma cell line U373. Therefore, coating liposomes with chitosan improved the encapsulation system's properties, resulting in a promising method for drug delivery.
Collapse
Affiliation(s)
- Rubén Gil-Gonzalo
- Pluridisciplinar Institute, Complutense University of Madrid, Paseo Juan XXIII, 1, E-28040 Madrid, Spain; (R.G.-G.); (S.P.)
| | - D. Alonzo Durante-Salmerón
- Department of Chemistry in Pharmaceutical Science, Pharmacy Faculty, Complutense University of Madrid, Plaza de Ramón y Cajal s/n, E-28040 Madrid, Spain; (D.A.D.-S.); (A.R.A.)
| | - Saeedeh Pouri
- Pluridisciplinar Institute, Complutense University of Madrid, Paseo Juan XXIII, 1, E-28040 Madrid, Spain; (R.G.-G.); (S.P.)
| | - Ernesto Doncel-Pérez
- Neural Regeneration Group, Hospital Nacional de Parapléjicos (SESCAM), E-45071 Toledo, Spain;
| | - Andrés R. Alcántara
- Department of Chemistry in Pharmaceutical Science, Pharmacy Faculty, Complutense University of Madrid, Plaza de Ramón y Cajal s/n, E-28040 Madrid, Spain; (D.A.D.-S.); (A.R.A.)
| | - Inmaculada Aranaz
- Pluridisciplinar Institute, Complutense University of Madrid, Paseo Juan XXIII, 1, E-28040 Madrid, Spain; (R.G.-G.); (S.P.)
- Department of Chemistry in Pharmaceutical Science, Pharmacy Faculty, Complutense University of Madrid, Plaza de Ramón y Cajal s/n, E-28040 Madrid, Spain; (D.A.D.-S.); (A.R.A.)
| | - Niuris Acosta
- Pluridisciplinar Institute, Complutense University of Madrid, Paseo Juan XXIII, 1, E-28040 Madrid, Spain; (R.G.-G.); (S.P.)
- Department of Chemistry in Pharmaceutical Science, Pharmacy Faculty, Complutense University of Madrid, Plaza de Ramón y Cajal s/n, E-28040 Madrid, Spain; (D.A.D.-S.); (A.R.A.)
| |
Collapse
|