1
|
Marques AVL, Ruginsk BE, Prado LDO, de Lima DE, Daniel IW, Moure VR, Valdameri G. The association of ABC proteins with multidrug resistance in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119878. [PMID: 39571941 DOI: 10.1016/j.bbamcr.2024.119878] [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: 08/26/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024]
Abstract
Multidrug resistance (MDR) poses one of the primary challenges for cancer treatment, especially in cases of metastatic disease. Various mechanisms contribute to MDR, including the overexpression of ATP-binding cassette (ABC) proteins. In this context, we reviewed the literature to establish a correlation between the overexpression of ABC proteins and MDR in cancer, considering both in vitro and clinical studies. Initially, we presented an overview of the seven subfamilies of ABC proteins, along with the subcellular localization of each protein. Subsequently, we identified a panel of 20 ABC proteins (ABCA1-3, ABCA7, ABCB1-2, ABCB4-6, ABCC1-5, ABCC10-11, ABCE1, ABCF2, ABCG1, and ABCG2) associated with MDR. We also emphasize the significance of drug sequestration by certain ABC proteins into intracellular compartments. Among the anticancer drugs linked to MDR, 29 were definitively identified as substrates for at least one of the three most crucial ABC transporters: ABCB1, ABCC1, and ABCG2. We further discussed that the most commonly used drugs in standard regimens for mainly breast cancer, lung cancer, and acute lymphoblastic leukemia could be subject to MDR mediated by ABC transporters. Collectively, these insights will aid in conducting new studies aimed at a deeper understanding of the clinical MDR mediated by ABC proteins and in designing more effective pharmacological treatments to enhance the objective response rate in cancer patients.
Collapse
Affiliation(s)
- Andrezza Viviany Lourenço Marques
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Bruna Estelita Ruginsk
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Larissa de Oliveira Prado
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Diogo Eugênio de Lima
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Isabelle Watanabe Daniel
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Vivian Rotuno Moure
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil.
| | - Glaucio Valdameri
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil.
| |
Collapse
|
2
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [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: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
3
|
Li X, Zheng W, Han L, He ZJ, Kang JC. A new aglycone derivative from the saprophytic fungus Tubeufia rubra. Nat Prod Res 2024:1-7. [PMID: 39688321 DOI: 10.1080/14786419.2024.2424391] [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: 03/20/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024]
Abstract
A new aglycone derivative (1) and five known compounds (2-6) have been isolated from a saprophytic fungus Tubeufia rubra for the first time. Their structures and absolute configurations were determined by nuclear magnetic resonance, high resolution mass spectrometry data and electronic circular dichroism. The bioactivities of all compounds were evaluated by cell analysis. Compound 5 showed significant toxicity to human cancer A549 cells, and the IC50 value was 32. 89 μg/mL. Compound 6 could reverse the multidrug resistance of A549/DDP cells, and the reversal value was 2.92. With the extension of the action time, the IC50 value of compound 6 decreased from 424.72 μg/mL at 24 h to 9.45 μg/mL at 72 h.
Collapse
Affiliation(s)
- Xin Li
- Engineering Research Center of the Utilization for Characteristic Bio-Pharmaceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang, P. R. China
- College of Pharmacy, Guizhou University, Guiyang, P. R. China
| | - Wen Zheng
- College of Pharmacy, Guizhou University, Guiyang, P. R. China
| | - Long Han
- College of Pharmacy, Guizhou University, Guiyang, P. R. China
| | - Zhang-Jiang He
- College of Pharmacy, Guizhou University, Guiyang, P. R. China
| | - Ji-Chuan Kang
- Engineering Research Center of the Utilization for Characteristic Bio-Pharmaceutical Resources in Southwest, Ministry of Education, Guizhou University, Guiyang, P. R. China
- College of Pharmacy, Guizhou University, Guiyang, P. R. China
| |
Collapse
|
4
|
Siciliano AC, Forciniti S, Onesto V, Iuele H, Cave DD, Carnevali F, Gigli G, Lonardo E, Del Mercato LL. A 3D Pancreatic Cancer Model with Integrated Optical Sensors for Noninvasive Metabolism Monitoring and Drug Screening. Adv Healthc Mater 2024; 13:e2401138. [PMID: 38978424 DOI: 10.1002/adhm.202401138] [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: 03/26/2024] [Revised: 06/06/2024] [Indexed: 07/10/2024]
Abstract
A distinct feature of pancreatic ductal adenocarcinoma (PDAC) is a prominent tumor microenvironment (TME) with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. The dynamic crosstalk between cancer cells and the dense stromal compartment leads to spatially and temporally heterogeneous metabolic alterations, such as acidic pH that contributes to drug resistance in PDAC. Thus, monitoring the extracellular pH metabolic fluctuations within the TME is crucial to predict and to quantify anticancer drug efficacy. Here, a simple and reliable alginate-based 3D PDAC model embedding ratiometric optical pH sensors and cocultures of tumor (AsPC-1) and stromal cells for simultaneously monitoring metabolic pH variations and quantify drug response is presented. By means of time-lapse confocal laser scanning microscopy (CLSM) coupled with a fully automated computational analysis, the extracellular pH metabolic variations are monitored and quantified over time during drug testing with gemcitabine, folfirinox, and paclitaxel, commonly used in PDAC therapy. In particular, the extracellular acidification is more pronounced after drugs treatment, resulting in increased antitumor effect correlated with apoptotic cell death. These findings highlight the importance of studying the influence of cellular metabolic mechanisms on tumor response to therapy in 3D tumor models, this being crucial for the development of personalized medicine approaches.
Collapse
Affiliation(s)
- Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
5
|
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] [MESH Headings] [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
|
6
|
Kim EH, Ryu Y, Choi J, Park D, Lee JW, Chi SG, Kim SH, Yang Y. Targeting miR-21 to Overcome P-glycoprotein Drug Efflux in Doxorubicin-Resistant 4T1 Breast Cancer. Biomater Res 2024; 28:0095. [PMID: 39434899 PMCID: PMC11491560 DOI: 10.34133/bmr.0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/17/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Acquired resistance to chemotherapy is a major challenge in the treatment of triple-negative breast cancer (TNBC). Despite accumulated evidence showing microRNA-21 (miR-21) as a vital regulator of tumor progression, the role of miR-21 in modulating the multidrug resistance of TNBC remains obscure. In this study, we demonstrate that miR-21 affects chemoresistance in 4T1 TNBC cells in response to doxorubicin (DOX) by regulating the P-glycoprotein (P-gp) drug efflux pump. Overexpression of miR-21 in the 4T1 cells markedly reduced their sensitivity to DOX, impeding DOX-promoted cell death. We employed anti-miR-21 oligonucleotide conjugated with a PD-L1-binding peptide (P21) for targeted delivery to 4T1 tumor cells. The selective down-regulation of miR-21 in 4T1 TNBC led to the reversal of P-gp-mediated DOX resistance by up-regulating phosphatase and tensin homolog (PTEN). Our study highlights that miR-21 is a key regulator of drug efflux pumps in TNBC, and targeting miR-21 could enhance DOX sensitivity, offering a potential therapeutic option for patients with DOX-resistant TNBC.
Collapse
Affiliation(s)
- Eun Hye Kim
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Life Sciences,
Korea University, Seoul 02841, Republic of Korea
| | - Youngri Ryu
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Life Sciences,
Korea University, Seoul 02841, Republic of Korea
| | - Jiwoong Choi
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Daeho Park
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Life Sciences,
Korea University, Seoul 02841, Republic of Korea
| | - Jong Won Lee
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology,
Korea University, Seoul 02841, Republic of Korea
| | - Sung-Gil Chi
- Department of Life Sciences,
Korea University, Seoul 02841, Republic of Korea
| | - Sun Hwa Kim
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology,
Korea University, Seoul 02841, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Division,
Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School,
University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
7
|
Bravo-Alfaro DA, Montalvo-González E, Zapien-Macias JM, Sampieri-Moran JM, García HS, Luna-Bárcenas G. Annonaceae acetogenins: A potential treatment for gynecological and breast cancer. Fitoterapia 2024; 178:106187. [PMID: 39147170 DOI: 10.1016/j.fitote.2024.106187] [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: 04/23/2024] [Revised: 08/05/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Breast and gynecological cancers are major health concerns due to their increasing incidence rates, and in some cases, their low survival probability. In recent years, multiple compounds of natural origin have been analyzed as alternative treatments for this disease. For instance, Acetogenins are plant secondary metabolites from the Annonaceae family, and its potential anticancer activity has been reported against a wide range of cancer cells both in vitro and in vivo. Several studies have demonstrated promising results of Acetogenins' antitumor capacity, given their selective activity of cellular inhibition at low concentrations. This review outlines the origin, structure, and antineoplastic activities in vitro and in vivo of Acetogenins from Annonaceae against breast cancer and gynecological cancers reported to date. Here, we also provide a systematic summary of the activity and possible mechanisms of action of Acetogenins against these types of cancer and provide references for developing future therapies based on Acetogenins and nanotechnologies.
Collapse
Affiliation(s)
- Diego A Bravo-Alfaro
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Epigmenio González 500 Fracc. San Pablo, Querétaro, Qro 76130, Mexico
| | - Efigenia Montalvo-González
- Laboratorio Integral de Investigación en Alimentos, Tecnológico Nacional de México-Instituto Tecnológico de Tepic. Av. Tecnológico 2595 Fracc. Lagos del Country, Tepic, Nayarit 63175, Mexico
| | - J Martin Zapien-Macias
- Horticultural Sciences Department, University of Florida, Institute of Food and Agricultural Sciences, Gainesville, FL 32611, United States of America
| | - Jessica M Sampieri-Moran
- Unidad de Investigación y Desarrollo de Alimentos, Tecnológico Nacional de México/Instituto Tecnológico de Veracruz, M.A. de Quevedo 2779, Col. Formando Hogar, Veracruz, Ver 91897, Mexico
| | - Hugo S García
- Unidad de Investigación y Desarrollo de Alimentos, Tecnológico Nacional de México/Instituto Tecnológico de Veracruz, M.A. de Quevedo 2779, Col. Formando Hogar, Veracruz, Ver 91897, Mexico.
| | - Gabriel Luna-Bárcenas
- Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Epigmenio González 500 Fracc. San Pablo, Querétaro, Qro 76130, Mexico.
| |
Collapse
|
8
|
Shahpouri P, Mehralitabar H, Kheirabadi M, Kazemi Noureini S. Potential suppression of multidrug-resistance-associated protein 1 by coumarin derivatives: an insight from molecular docking and MD simulation studies. J Biomol Struct Dyn 2024; 42:9184-9200. [PMID: 37667877 DOI: 10.1080/07391102.2023.2250456] [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: 04/24/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023]
Abstract
Human MRP1 protein plays a vital role in cancer multidrug resistance. Coumarins show promising pharmacological properties. Virtual screening, ADMET, molecular docking and molecular dynamics (MD) simulations were utilized as pharmacoinformatic tools to identify potential MRP1 inhibitors among coumarin derivatives. Using in silico ADMET, 50 hits were further investigated for their selectivity toward the nucleotide-binding domains (NBDs) of MRP1 using molecular docking. Accordingly, coumarin, its symmetrical ketone derivative Lig. No. 4, and Reversan were candidates for focused docking study with the NBDs domains compared with ATP. The result indicates that Lig. No. 4, with the best binding score, interacts with NBDs via hydrogen bonds with residues: GLN713, LYS684, GLY683, CYS682 in NBD1, and GLY1432, GLY771, SER769 and GLN1374 in NBD2, which mostly overlap with ATP binding residues. Moreover, doxorubicin (Doxo) was docked to the transmembrane domains (TMDs) active site of MRP1. Doxo interaction with TMDs was subjected to MD simulation in the NBDs free and occupied with Lig. No. 4 states. The results showed that Doxo interacts more strongly with TMD residues in inward facing feature of TMDs helices. However, when Lig. No. 4 exists in NBDs, Doxo interactions are different, and TMD helices show more outward-facing conformation. This result may suggest a partial competitive inhibition mechanism for the Lig. No. 4 on MRP1 compared with ATP. So, it may inhibit active complex formation by interfering with ATP entrance to NBDs and locking MRP1 conformation in outward-facing mode. This study suggests a valuable coumarin derivative that can be further investigated for potent MRP1 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Parisa Shahpouri
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Havva Mehralitabar
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mitra Kheirabadi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | | |
Collapse
|
9
|
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
|
10
|
Kaehler M, von Bubnoff N, Cascorbi I, Gorantla SP. Molecular biomarkers of leukemia: convergence-based drug resistance mechanisms in chronic myeloid leukemia and myeloproliferative neoplasms. Front Pharmacol 2024; 15:1422565. [PMID: 39104388 PMCID: PMC11298451 DOI: 10.3389/fphar.2024.1422565] [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: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Leukemia represents a diverse group of hematopoietic neoplasms that can be classified into different subtypes based on the molecular aberration in the affected cell population. Identification of these molecular classification is required to identify specific targeted therapeutic approaches for each leukemic subtype. In general, targeted therapy approaches achieve good responses in some leukemia subgroups, however, resistance against these targeted therapies is common. In this review, we summarize molecular drug resistance biomarkers in targeted therapies in BCR::ABL1-driven chronic myeloid leukemia (CML) and JAK2-driven myeloproliferative neoplasms (MPNs). While acquisition of secondary mutations in the BCR::ABL1 kinase domain is the a common mechanism associated with TKI resistance in CML, in JAK2-driven MPNs secondary mutations in JAK2 are rare. Due to high prevalence and lack of specific therapy approaches in MPNs compared to CML, identification of crucial pathways leading to inhibitor persistence in MPN model is utterly important. In this review, we focus on different alternative signaling pathways activated in both, BCR::ABL1-mediated CML and JAK2-mediated MPNs, by combining data from in vitro and in vivo-studies that could be used as potential biomarkers of drug resistance. In a nutshell, some common similarities, especially activation of PDGFR, Ras, PI3K/Akt signaling pathways, have been demonstrated in both leukemias. In addition, induction of the nucleoprotein YBX1 was shown to be involved in TKI-resistant JAK2-mediated MPN, as well as TKI-resistant CML highlighting deubiquitinating enzymes as potential biomarkers of TKI resistance. Taken together, whole exome sequencing of cell-based or patients-derived samples are highly beneficial to define specific resistance markers. Additionally, this might be helpful for the development of novel diagnostic tools, e.g., liquid biopsy, and novel therapeutic agents, which could be used to overcome TKI resistance in molecularly distinct leukemia subtypes.
Collapse
Affiliation(s)
- Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sivahari Prasad Gorantla
- Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
11
|
Rijwan, Arjmand F, Tabassum S. Repurposing the antihistamine drug bilastine as an anti-cancer metallic drug entity: synthesis and single-crystal X-ray structure analysis of metal-based bilastine and phen [Co(II), Cu(II) and Zn(II)] tailored anticancer chemotherapeutic agents against resistant cancer cells. Dalton Trans 2024; 53:10126-10141. [PMID: 38817206 DOI: 10.1039/d4dt00426d] [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: 06/01/2024]
Abstract
Bilastine (BLA), 2-(4-(2-(4-(1-(2-ethoxyethyl)-1H-benzo[d]imidazole-2-yl)-piperidin-1-yl)-ethyl)-phenyl)-2-methylpropanoic acid, is an active antihistamine drug. With the idea of repurposing drugs from the existing pool of 'active' pharmaceutical ingredients, the therapeutic potency of bilastine as an anticancer agent was investigated via the tailored synthesis of a metal-based anticancer drug formulation of the type [BLA(phen)2M(II)]+·X-, where M = Co, Cu, and Zn and X- = NO3 and ClO4. The synthesized metal-based chemotherapeutics derived from the bilastine drug that acts as a ligand were thoroughly characterized using spectroscopic techniques, namely, UV-vis, FT-IR, and EPR (in the case of 1 and 2); 1H-NMR and 13C-NMR (in the case of 3); ESI-MS and single-crystal X-ray diffraction studies. Comprehensive biological studies (DNA binding, cleavage, and cytotoxic activity) using various biophysical and gel electrophoretic methods were carried out to validate their potential as anticancer agents. The cytotoxic activity of 'therapeutically promising' copper(II)-based drug candidate 2 was evaluated against MCF-7, MBA-MD-231, HeLa, HepG2, and Mia-PaCa-2 cancer cells via an SRB assay, and the results demonstrated 2 as a potent anticancer agent at low nanomolar concentrations against all tested cancer cells, preferably with a much superior anticancer efficacy against human pancreatic cancer cells.
Collapse
Affiliation(s)
- Rijwan
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India.
| | - Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India.
| | - Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University, Aligarh, UP 202002, India.
| |
Collapse
|
12
|
Yang Q, To KKW, Hu G, Fu K, Yang C, Zhu S, Pan C, Wang F, Luo K, Fu L. BI-2865, a pan-KRAS inhibitor, reverses the P-glycoprotein induced multidrug resistance in vitro and in vivo. Cell Commun Signal 2024; 22:325. [PMID: 38872211 PMCID: PMC11170860 DOI: 10.1186/s12964-024-01698-4] [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: 03/15/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Multidrug resistance (MDR) limits successful cancer chemotherapy. P-glycoprotein (P-gp), BCRP and MRP1 are the key triggers of MDR. Unfortunately, no MDR modulator was approved by FDA to date. Here, we will investigate the effect of BI-2865, a pan-KRAS inhibitor, on reversing MDR induced by P-gp, BCRP and MRP1 in vitro and in vivo, and its reversal mechanisms will be explored. METHODS The cytotoxicity of BI-2865 and its MDR removal effect in vitro were tested by MTT assays, and the corresponding reversal function in vivo was assessed through the P-gp mediated KBv200 xenografts in mice. BI-2865 induced alterations of drug discharge and reservation in cells were estimated by experiments of Flow cytometry with fluorescent doxorubicin, and the chemo-drug accumulation in xenografts' tumor were analyzed through LC-MS. Mechanisms of BI-2865 inhibiting P-gp substrate's efflux were analyzed through the vanadate-sensitive ATPase assay, [125I]-IAAP-photolabeling assay and computer molecular docking. The effects of BI-2865 on P-gp expression and KRAS-downstream signaling were detected via Western blotting, Flow cytometry and/or qRT-PCR. Subcellular localization of P-gp was visualized by Immunofluorescence. RESULTS We found BI-2865 notably fortified response of P-gp-driven MDR cancer cells to the administration of chemo-drugs including paclitaxel, vincristine and doxorubicin, while such an effect was not observed in their parental sensitive cells and BCRP or MRP1-driven MDR cells. Importantly, the mice vivo combination study has verified that BI-2865 effectively improved the anti-tumor action of paclitaxel without toxic injury. In mechanism, BI-2865 prompted doxorubicin accumulating in carcinoma cells by directly blocking the efflux function of P-gp, which more specifically, was achieved by BI-2865 competitively binding to the drug-binding sites of P-gp. What's more, at the effective MDR reversal concentrations, BI-2865 neither varied the expression and location of P-gp nor reduced its downstream AKT or ERK1/2 signaling activity. CONCLUSIONS This study uncovered a new application of BI-2865 as a MDR modulator, which might be used to effectively, safely and specifically improve chemotherapeutic efficacy in the clinical P-gp mediated MDR refractory cancers.
Collapse
MESH Headings
- Humans
- Animals
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Multiple/drug effects
- Mice
- Cell Line, Tumor
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Xenograft Model Antitumor Assays
- Mice, Nude
- Doxorubicin/pharmacology
- Mice, Inbred BALB C
- Female
Collapse
Affiliation(s)
- Qihong Yang
- People's Hospital of Longhua, Shenzhen, 518109, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Guilin Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shuangli Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kewang Luo
- People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
13
|
Phaisal W, Albitar O, Chariyavilaskul P, Jantarabenjakul W, Wacharachaisurapol N, Ghadzi SMS, Zainal H, Harun SN. Genetic and clinical predictors of rifapentine and isoniazid pharmacokinetics in paediatrics with tuberculosis infection. J Antimicrob Chemother 2024; 79:1270-1278. [PMID: 38661209 DOI: 10.1093/jac/dkae059] [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: 07/19/2023] [Accepted: 02/20/2024] [Indexed: 04/26/2024] Open
Abstract
OBJECTIVES Twelve weekly doses of rifapentine and isoniazid (3HP regimen) are recommended for TB preventive therapy in children with TB infection. However, they present with variability in the pharmacokinetic profiles. The current study aimed to develop a pharmacokinetic model of rifapentine and isoniazid in 12 children with TB infection using NONMEM. METHODS Ninety plasma and 41 urine samples were collected at Week 4 of treatment. Drug concentrations were measured using a validated HPLC-UV method. MassARRAY® SNP genotyping was used to investigate genetic factors, including P-glycoprotein (ABCB1), solute carrier organic anion transporter B1 (SLCO1B1), arylacetamide deacetylase (AADAC) and N-acetyl transferase (NAT2). Clinically relevant covariates were also analysed. RESULTS A two-compartment model for isoniazid and a one-compartment model for rifapentine with transit compartment absorption and first-order elimination were the best models for describing plasma and urine data. The estimated (relative standard error, RSE) of isoniazid non-renal clearance was 3.52 L·h-1 (23.1%), 2.91 L·h-1 (19.6%), and 2.58 L·h-1 (20.0%) in NAT2 rapid, intermediate and slow acetylators. A significant proportion of the unchanged isoniazid was cleared renally (2.7 L·h-1; 8.0%), while the unchanged rifapentine was cleared primarily through non-renal routes (0.681 L·h-1; 3.6%). Participants with the ABCB1 mutant allele had lower bioavailability of rifapentine, while food prolonged the mean transit time of isoniazid. CONCLUSIONS ABCB1 mutant allele carriers may require higher rifapentine doses; however, this must be confirmed in larger trials. Food did not affect overall exposure to isoniazid and only delayed absorption time.
Collapse
Affiliation(s)
- Weeraya Phaisal
- Center for Medical Diagnostic Laboratories, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Orwa Albitar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Penang, Malaysia
| | - Pajaree Chariyavilaskul
- Center for Medical Diagnostic Laboratories, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Watsamon Jantarabenjakul
- Center of Excellence for Paediatric Infectious Diseases and Vaccines, Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Division of Infectious Diseases, Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Noppadol Wacharachaisurapol
- Center for Medical Diagnostic Laboratories, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Hadzliana Zainal
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Penang, Malaysia
| | - Sabariah Noor Harun
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Penang, Malaysia
| |
Collapse
|
14
|
Beier AMK, Ebersbach C, Siciliano T, Scholze J, Hofmann J, Hönscheid P, Baretton GB, Woods K, Guezguez B, Dubrovska A, Markowitsch SD, Thomas C, Puhr M, Erb HHH. Targeting the glutamine metabolism to suppress cell proliferation in mesenchymal docetaxel-resistant prostate cancer. Oncogene 2024; 43:2038-2050. [PMID: 38750263 PMCID: PMC11196217 DOI: 10.1038/s41388-024-03059-4] [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: 01/14/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024]
Abstract
Docetaxel (DX) serves as a palliative treatment option for metastatic prostate cancer (PCa). Despite initial remission, acquired DX resistance is inevitable. The mechanisms behind DX resistance have not yet been deciphered, but a mesenchymal phenotype is associated with DX resistance. Mesenchymal phenotypes have been linked to metabolic rewiring, obtaining most ATP production by oxidative phosphorylation (OXPHOS) powered substantially by glutamine (Gln). Likewise, Gln is known to play an essential role in modulating bioenergetic, redox homeostasis and autophagy. Herein, investigations of Gln deprivation on DX-sensitive and -resistant (DR) PCa cells revealed that the DR cell sub-lines were susceptible to Gln deprivation. Mechanistically, Gln deprivation reduced OXPHOS and ATP levels, causing a disturbance in cell cycle progression. Genetic and chemical inhibition of the Gln-metabolism key protein GLS1 could validate the Gln deprivation results, thereby representing a valid therapeutic target. Moreover, immunohistological investigation of GLS1 revealed a high-expressing GLS1 subgroup post-docetaxel failure, exhibiting low overall survival. This subgroup presents an intriguing opportunity for targeted therapy focusing on glutamine metabolism. Thus, these findings highlight a possible clinical rationale for the chemical inhibition of GLS1 as a therapeutic strategy to target mesenchymal DR PCa cells, thereby delaying accelerated tumour progression.
Collapse
Affiliation(s)
| | - Celina Ebersbach
- Department of Urology, Technische Universität Dresden, Dresden, Germany
| | - Tiziana Siciliano
- Department of Urology, Technische Universität Dresden, Dresden, Germany
| | - Jana Scholze
- Department of Urology, Technische Universität Dresden, Dresden, Germany
| | - Jörg Hofmann
- Department of Urology, Technische Universität Dresden, Dresden, Germany
| | - Pia Hönscheid
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Gustavo B Baretton
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Kevin Woods
- IIIrd Department of Medicine - Hematology & Oncology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Borhane Guezguez
- IIIrd Department of Medicine - Hematology & Oncology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Dresden, Germany
| | - Sascha D Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Mainz, Germany
| | - Christian Thomas
- Department of Urology, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Martin Puhr
- Medical University of Innsbruck, Department of Urology, 6020, Innsbruck, Austria
| | - Holger H H Erb
- Department of Urology, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Sun L, Li Z, Lan J, Wu Y, Zhang T, Ding Y. Better together: nanoscale co-delivery systems of therapeutic agents for high-performance cancer therapy. Front Pharmacol 2024; 15:1389922. [PMID: 38831883 PMCID: PMC11144913 DOI: 10.3389/fphar.2024.1389922] [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: 02/22/2024] [Accepted: 04/22/2024] [Indexed: 06/05/2024] Open
Abstract
Combination therapies can enhance the sensitivity of cancer to drugs, lower drug doses, and reduce side effects in cancer treatment. However, differences in the physicochemical properties and pharmacokinetics of different therapeutic agents limit their application. To avoid the above dilemma and achieve accurate control of the synergetic ratio, a nanoscale co-delivery system (NCDS) has emerged as a prospective tool for combined therapy in cancer treatment, which is increasingly being used to co-load different therapeutic agents. In this study, we have summarized the mechanisms of therapeutic agents in combination for cancer therapy, nanoscale carriers for co-delivery, drug-loading strategies, and controlled/targeted co-delivery systems, aiming to give a general picture of these powerful approaches for future NCDS research studies.
Collapse
Affiliation(s)
- Liyan Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ya Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Furtado A, Duarte AC, Costa AR, Gonçalves I, Santos CRA, Gallardo E, Quintela T. Circadian ABCG2 Expression Influences the Brain Uptake of Donepezil across the Blood-Cerebrospinal Fluid Barrier. Int J Mol Sci 2024; 25:5014. [PMID: 38732233 PMCID: PMC11084460 DOI: 10.3390/ijms25095014] [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: 03/21/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Donepezil (DNPZ) is a cholinesterase inhibitor used for the management of Alzheimer's disease (AD) and is dependent on membrane transporters such as ABCG2 to actively cross brain barriers and reach its target site of action in the brain. Located in the brain ventricles, the choroid plexus (CP) forms an interface between the cerebrospinal fluid (CSF) and the bloodstream, known as the blood-CSF barrier (BCSFB). Historically, the BCSFB has received little attention as a potential pathway for drug delivery to the central nervous system (CNS). Nonetheless, this barrier is presently viewed as a dynamic transport interface that limits the traffic of molecules into and out of the CNS through the presence of membrane transporters, with parallel activity with the BBB. The localization and expression of drug transporters in brain barriers represent a huge obstacle for drug delivery to the brain and a major challenge for the development of therapeutic approaches to CNS disorders. The widespread interest in understanding how circadian clocks modulate many processes that define drug delivery in order to predict the variability in drug safety and efficacy is the next bridge to improve effective treatment. In this context, this study aims at characterizing the circadian expression of ABCG2 and DNPZ circadian transport profile using an in vitro model of the BCSFB. We found that ABCG2 displays a circadian pattern and DNPZ is transported in a circadian way across this barrier. This study will strongly impact on the capacity to modulate the BCSFB in order to control the penetration of DNPZ into the brain and improve therapeutic strategies for the treatment of AD according to the time of the day.
Collapse
Affiliation(s)
- André Furtado
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Universityof Beira Interior, Avenida Infante Dom Henrique, 6200-506 Covilhã, Portugal
| | - Ana Catarina Duarte
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Universityof Beira Interior, Avenida Infante Dom Henrique, 6200-506 Covilhã, Portugal
| | - Ana R. Costa
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Universityof Beira Interior, Avenida Infante Dom Henrique, 6200-506 Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Universityof Beira Interior, Avenida Infante Dom Henrique, 6200-506 Covilhã, Portugal
| | - Cecília R. A. Santos
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Universityof Beira Interior, Avenida Infante Dom Henrique, 6200-506 Covilhã, Portugal
| | - Eugenia Gallardo
- Laboratório de Fármaco-Toxicologia-UBIMedical, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Universityof Beira Interior, Avenida Infante Dom Henrique, 6200-506 Covilhã, Portugal
- Faculty of Health Sciences, Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal
| |
Collapse
|
17
|
Li LB, Yang LX, Liu L, Liu FR, Li AH, Zhu YL, Wen H, Xue X, Tian ZX, Sun H, Li PC, Zhao XG. Targeted inhibition of the HNF1A/SHH axis by triptolide overcomes paclitaxel resistance in non-small cell lung cancer. Acta Pharmacol Sin 2024; 45:1060-1076. [PMID: 38228910 PMCID: PMC11053095 DOI: 10.1038/s41401-023-01219-y] [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: 07/17/2023] [Accepted: 12/17/2023] [Indexed: 01/18/2024] Open
Abstract
Paclitaxel resistance is associated with a poor prognosis in non-small cell lung cancer (NSCLC) patients, and currently, there is no promising drug for paclitaxel resistance. In this study, we investigated the molecular mechanisms underlying the chemoresistance in human NSCLC-derived cell lines. We constructed paclitaxel-resistant NSCLC cell lines (A549/PR and H460/PR) by long-term exposure to paclitaxel. We found that triptolide, a diterpenoid epoxide isolated from the Chinese medicinal herb Tripterygium wilfordii Hook F, effectively enhanced the sensitivity of paclitaxel-resistant cells to paclitaxel by reducing ABCB1 expression in vivo and in vitro. Through high-throughput sequencing, we identified the SHH-initiated Hedgehog signaling pathway playing an important role in this process. We demonstrated that triptolide directly bound to HNF1A, one of the transcription factors of SHH, and inhibited HNF1A/SHH expression, ensuing in attenuation of Hedgehog signaling. In NSCLC tumor tissue microarrays and cancer network databases, we found a positive correlation between HNF1A and SHH expression. Our results illuminate a novel molecular mechanism through which triptolide targets and inhibits HNF1A, thereby impeding the activation of the Hedgehog signaling pathway and reducing the expression of ABCB1. This study suggests the potential clinical application of triptolide and provides promising prospects in targeting the HNF1A/SHH pathway as a therapeutic strategy for NSCLC patients with paclitaxel resistance. Schematic diagram showing that triptolide overcomes paclitaxel resistance by mediating inhibition of the HNF1A/SHH/ABCB1 axis.
Collapse
Affiliation(s)
- Ling-Bing Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Ling-Xiao Yang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Lei Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Fan-Rong Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Alex H Li
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10010, USA
| | - Yi-Lin Zhu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Hao Wen
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Zhong-Xian Tian
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Hong Sun
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10010, USA
| | - Pei-Chao Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
| | - Xiao-Gang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
| |
Collapse
|
18
|
Iyer KK, Poel D, Miggelenbrink A, Kerkhof W, Janssen J, Bakkerus L, de Jong L, van den Hombergh E, Nagtegaal ID, Tauriello DVF, van Erp NP, Verheul HMW. High-dose short-term osimertinib treatment is effective in patient-derived metastatic colorectal cancer organoids. BJC REPORTS 2024; 2:29. [PMID: 39516561 PMCID: PMC11523998 DOI: 10.1038/s44276-024-00042-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/11/2023] [Accepted: 01/20/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Most tyrosine kinase inhibitors (TKIs) have failed in clinical trials for metastatic colorectal cancer (mCRC). To leverage the additional lower-affinity targets that most TKIs have, high-dose regimens that trigger efficacy are explored. Here, we studied unprecedented drug exposure-response relationships in vitro using mCRC patient-derived tumour organoids (PDTOs). METHODS We investigated the cytotoxic anti-tumour effect of high-dose, short-term (HDST) TKI treatment on 5 PDTOs. Sunitinib, cediranib and osimertinib were selected based on favourable physicochemical and pharmacokinetic properties. Intra-tumoroid TKI concentrations were measured using a clinically validated LC/MS-MS method. Cell death was determined using an enzyme activity assay, immunofluorescent staining and western blotting. RESULTS Most PDTOs tested were sensitive to sunitinib and cediranib, but all to osimertinib. Furthermore, HDST osimertinib treatment effectively blocks organoid growth. This treatment led to markedly elevated intra-tumoroid TKI concentrations, which correlated with PDTO sensitivity. Mechanistically, HDST osimertinib treatment induced apoptosis in treated PDTOs. CONCLUSION Our work provides a better understanding of TKI exposure vs response and can be used to determine patient-specific sensitivity. Additionally, these results may guide both mechanistic elucidation in organotypic translational models and the translation of target drug exposure to clinical dosing strategies. Moreover, HDST osimertinib treatment warrants clinical exploration for mCRC.
Collapse
Affiliation(s)
- Kirti K Iyer
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Medical Biosciences, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dennis Poel
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Medical Biosciences, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Anne Miggelenbrink
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Medical Biosciences, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Wouter Kerkhof
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Medical Biosciences, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jorien Janssen
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Lotte Bakkerus
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Loek de Jong
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Erik van den Hombergh
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Daniele V F Tauriello
- Department of Medical Biosciences, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Medical Oncology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands.
- Department of Medical Oncology, Erasmus Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
Manoharan JP, Palanisamy H, Vidyalakshmi S. Overcoming multi drug resistance mediated by ABC transporters by a novel acetogenin- annonacin from Annona muricata L. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117598. [PMID: 38113989 DOI: 10.1016/j.jep.2023.117598] [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: 06/20/2023] [Revised: 11/28/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Multi-Drug Resistance (MDR), mediated by P-glycoprotein (P-gp) is one of the barriers to successful chemotherapy in colon cancer patients. Annona muricata L. (A.muricata), commonly known as soursop/Graviola, is a medicinal plant that has been traditionally used in treating diverse diseases including cancer. Phytochemicals of A.muricata (Annonaceous Acetogenins-AGEs) have been well-reported for their anti-cancer effects on various cancers. AIM OF THE STUDY The study aimed to examine the effect of AGEs in reversing MDR in colorectal cancer cells. METHODS Based on molecular docking and molecular dynamic simulation, the stability of annonacin upon P-gp was investigated. Further in vitro studies were carried in oxaliplatin-resistant human colon cancer cells (SW480R) to study the biological effect of annonacin, in reversing drug resistance in these cells. RESULTS Molecular docking and simulation studies have indicated that annonacin stably interacted at the drug binding site of P-gp. In vitro analysis showed that annonacin was able to significantly reduce the expression of P-gp by 2.56 folds. It also induced apoptosis in the drug-resistant colon cancer cells. Moreover, the intracellular accumulation of P-gp substrate (calcein-AM) was observed to increase in resistant cells upon treatment with annonacin. CONCLUSION Our findings suggest that annonacin could inhibit the efflux of chemotherapeutic drugs mediated by P-gp and thereby help in reversing MDR in colon cancer cells. Further in vivo studies are required to decipher the underlying mechanism of annonacin in treating MDR cancers.
Collapse
Affiliation(s)
- Jeevitha Priya Manoharan
- Department of Biotechnology, PSG College of Technology, Coimbatore, Tamil Nadu, India; Department of Biomedical Engineering, Sri Ramakrishna Engineering College, Coimbatore, Tamil Nadu, India.
| | - Hema Palanisamy
- Department of Biotechnology, PSG College of Technology, Coimbatore, Tamil Nadu, India.
| | | |
Collapse
|
20
|
Zou JY, Chen QL, Luo XC, Damdinjav D, Abdelmohsen UR, Li HY, Battulga T, Chen HB, Wang YQ, Zhang JY. Natural products reverse cancer multidrug resistance. Front Pharmacol 2024; 15:1348076. [PMID: 38572428 PMCID: PMC10988293 DOI: 10.3389/fphar.2024.1348076] [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: 12/01/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Cancer stands as a prominent global cause of death. One of the key reasons why clinical tumor chemotherapy fails is multidrug resistance (MDR). In recent decades, accumulated studies have shown how Natural Product-Derived Compounds can reverse tumor MDR. Discovering novel potential modulators to reduce tumor MDR by Natural Product-Derived Compounds has become a popular research area across the globe. Numerous studies mainly focus on natural products including flavonoids, alkaloids, terpenoids, polyphenols and coumarins for their MDR modulatory activity. Natural products reverse MDR by regulating signaling pathways or the relevant expressed protein or gene. Here we perform a deep review of the previous achievements, recent advances in the development of natural products as a treatment for MDR. This review aims to provide some insights for the study of multidrug resistance of natural products.
Collapse
Affiliation(s)
- Jia-Yu Zou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qi-Lei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xiao-Ci Luo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Usama Ramadan Abdelmohsen
- Deraya Center for Scientific Research, Deraya University, New Minia, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Hong-Yan Li
- Ministry of Education Engineering Research Center of Tibetan Medicine Detection Technology, Xizang Minzu University, Xianyang, China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Yu-Qing Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Affiliated TCM Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian-Ye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
21
|
Wang P, Min S, Chen C, Hu J, Wei D, Wang X. Phytohemagglutinin from Phaseolus vulgaris enhances the lung cancer cell chemotherapy sensitivity by changing cell membrane permeability. J Nat Med 2024; 78:355-369. [PMID: 38265611 DOI: 10.1007/s11418-023-01772-0] [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: 07/17/2023] [Accepted: 12/08/2023] [Indexed: 01/25/2024]
Abstract
Chemotherapy is still a prevalent strategy for clinical lung cancer treatment. However, the inevitable emerged drug resistance has become a great hurdle to therapeutic effect. Studies have demonstrated that the primary cause of drug resistance is a decrease in the chemotherapeutic medicine concentration. Several lectins have been confirmed to be effective as chemotherapy adjuvants, enhancing the anti-tumor effects of chemotherapy drugs. Here, we combined phytohemagglutinin (PHA), which has been reported possess anti-tumor effects, with chemotherapy drugs Cisplatin (DDP) and Adriamycin (ADM) on lung cancer cells to detect the sensitivities of PHA as a chemotherapy adjuvant. Our results demonstrated that the PHA significantly enhanced the sensitivity of lung cancer cells to DDP and ADM, and Western blot showed that PHA combined with DDP or ADM enhance cytotoxic effects by inhibiting autophagy and promoting apoptosis. More importantly, we found PHA enhanced the chemotherapeutic drugs cytotoxicity by changing the cell membrane to increase the intracellular chemotherapeutic drugs concentration. Besides, the combination of PHA and ADM increased the ADM concentration in the multidrug-resistant strain A549-R cells and achieved the drug sensitization effect. Our results suggest that PHA combined with chemotherapy can be applied in the treatment of lung cancer cells and lung cancer multidrug-resistant strains, and provide a novel strategy for clinical tumor chemotherapy and a new idea to solve the problem of drug resistance in clinical lung cancer.
Collapse
Affiliation(s)
- Peipei Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Shitong Min
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Dapeng Wei
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xia Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [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: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
23
|
Gao Y, Deng H, Zhao Y, Li M, Wang L, Zhang Y. Gene Expression of Abcc2 and Its Regulation by Chicken Xenobiotic Receptor. TOXICS 2024; 12:55. [PMID: 38251011 PMCID: PMC10818656 DOI: 10.3390/toxics12010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
Membrane transporter multidrug resistance-associated protein 2 (MRP2/Abcc2) exhibits high pharmaco-toxicological relevance because it exports multiple cytotoxic compounds from cells. However, no detailed information about the gene expression and regulation of MRP2 in chickens is yet available. Here, we sought to investigate the expression distribution of Abcc2 in different tissues of chicken and then determine whether Abcc2 expression is induced by chicken xenobiotic receptor (CXR). The bioinformatics analyses showed that MRP2 transporters have three transmembrane structural domains (MSDs) and two highly conserved nucleotide structural domains (NBDs), and a close evolutionary relationship with turkeys. Tissue distribution analysis indicated that Abcc2 was highly expressed in the liver, kidney, duodenum, and jejunum. When exposed to metyrapone (an agonist of CXR) and ketoconazole (an antagonist of CXR), Abcc2 expression was upregulated and downregulated correspondingly. We further confirmed that Abcc2 gene regulation is dependent on CXR, by overexpressing and interfering with CXR, respectively. We also demonstrated the induction of Abcc2 expression and the activity of ivermectin, with CXR being a likely mediator. Animal experiments demonstrated that metyrapone and ivermectin induced Abcc2 in the liver, kidney, and duodenum of chickens. Together, our study identified the gene expression of Abcc2 and its regulation by CXR in chickens, which may provide novel targets for the reasonable usage of veterinary drugs.
Collapse
Affiliation(s)
- Yanhong Gao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Huacheng Deng
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Yuying Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Mei Li
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Liping Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Yujuan Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| |
Collapse
|
24
|
Damiani D, Tiribelli M. ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target? Biomedicines 2024; 12:111. [PMID: 38255216 PMCID: PMC10813371 DOI: 10.3390/biomedicines12010111] [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: 11/20/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Despite the progress in the knowledge of disease pathogenesis and the identification of many molecular markers as potential targets of new therapies, the cure of acute myeloid leukemia remains challenging. Disease recurrence after an initial response and the development of resistance to old and new therapies account for the poor survival rate and still make allogeneic stem cell transplantation the only curative option. Multidrug resistance (MDR) is a multifactorial phenomenon resulting from host-related characteristics and leukemia factors. Among these, the overexpression of membrane drug transporter proteins belonging to the ABC (ATP-Binding Cassette)-protein superfamily, which diverts drugs from their cellular targets, plays an important role. Moreover, a better understanding of leukemia biology has highlighted that, at least in cancer, ABC protein's role goes beyond simple drug transport and affects many other cell functions. In this paper, we summarized the current knowledge of ABCG2 (formerly Breast Cancer Resistance Protein, BCRP) in acute myeloid leukemia and discuss the potential ways to overcome its efflux function and to revert its ability to confer stemness to leukemia cells, favoring the persistence of leukemia progenitors in the bone marrow niche and justifying relapse also after therapy intensification with allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| |
Collapse
|
25
|
Shen L, Zhou P, Wang YM, Zhu Z, Yuan Q, Cao S, Li J. Supramolecular nanoparticles based on elastin-like peptides modified capsid protein as drug delivery platform with enhanced cancer chemotherapy efficacy. Int J Biol Macromol 2024; 256:128107. [PMID: 38007030 DOI: 10.1016/j.ijbiomac.2023.128107] [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: 10/24/2023] [Revised: 11/11/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
Cancer, a prevalent disease posing significant threats to human health and longevity, necessitates effective therapeutic interventions. Chemotherapy has emerged as a primary strategy following surgical procedures for combating most malignancies. Despite the considerable efficacy of conventional chemotherapeutic agents against cancer cells, their utility is hindered by profound challenges such as multidrug resistance and deleterious toxic side effects, thereby limiting their systemic application. To tackle these challenges, we have devised a promising nanomedicine platform based on a plant virus. Specifically, we have selected the cowpea melanoma mottled virus (CCMV) as our nano-delivery system owing to its monodisperse and homogeneous size, as well as its intrinsic ability for controlled self-assembly. Leveraging the potential of this platform, we have engineered CCMV-based nanoparticles functionalized with elastin-like peptides (ELPs) at their N-terminal region. The target protein, CP-ELP, was expressed via E.coli, enabling encapsulation of the model drug DOX upon structural domain modification of the protein. The resulting nanoparticles exhibit uniform size distribution, facilitating efficient internalization by tumor cells and subsequent intracellular drug release, leading to enhanced antitumor efficacy. In addition, EVLP@DOX nanoparticles were found to activate immune response of tumor microenvironment in vivo, which further inhibiting tumor growth. Our designed nanoparticles have also demonstrated remarkable therapeutic effectiveness and favorable biological safety profiles in both murine melanoma and colorectal cancer models.
Collapse
Affiliation(s)
- Luxuan Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Min Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhixiong Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuqin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jianshu Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology & Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
26
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [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: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
27
|
Burgueño-Rodríguez G, Méndez Y, Olano N, Schelotto M, Castillo L, Soler AM, da Luz J. Pharmacogenetics of pediatric acute lymphoblastic leukemia in Uruguay: adverse events related to induction phase drugs. Front Pharmacol 2023; 14:1278769. [PMID: 38044950 PMCID: PMC10690766 DOI: 10.3389/fphar.2023.1278769] [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: 08/16/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
In Uruguay, the pediatric acute lymphoblastic leukemia (ALL) cure rate is 82.2%, similar to those reported in developed countries. However, many patients suffer adverse effects that could be attributed, in part, to genetic variability. This study aims to identify genetic variants related to drugs administered during the induction phase and analyze their contribution to adverse effects, considering individual genetic ancestry. Ten polymorphisms in five genes (ABCB1, CYP3A5, CEP72, ASNS, and GRIA1) related to prednisone, vincristine, and L-asparaginase were genotyped in 200 patients. Ancestry was determined using 45 ancestry informative markers (AIMs). The sample ancestry was 69.2% European, 20.1% Native American, and 10.7% African, but with high heterogeneity. Mucositis, Cushing syndrome, and neurotoxicity were the only adverse effects linked with genetic variants and ancestry. Mucositis was significantly associated with ASNS (rs3832526; 3R/3R vs. 2R carriers; OR: = 6.88 [1.88-25.14], p = 0.004) and CYP3A5 (non-expressors vs. expressors; OR: 4.55 [1.01-20.15], p = 0.049) genes. Regarding Cushing syndrome, patients with the TA genotype (rs1049674, ASNS) had a higher risk of developing Cushing syndrome than those with the TT genotype (OR: 2.60 [1.23-5.51], p = 0.012). Neurotoxicity was significantly associated with ABCB1 (rs9282564; TC vs. TT; OR: 4.25 [1.47-12.29], p = 0.007). Moreover, patients with <20% Native American ancestry had a lower risk of developing neurotoxicity than those with ≥20% (OR: 0.312 [0.120-0.812], p = 0.017). This study shows the importance of knowing individual genetics to improve the efficacy and safety of acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Gabriela Burgueño-Rodríguez
- Laboratorio de Genética Molecular Humana, Departamento de Ciencias Biológicas, CENUR Litoral Norte-Sede Salto, Universidad de la República, Salto, Uruguay
- Red Latinoamericana de Implementación y Validación de Guías Clínicas Farmacogenómicas (RELIVAF-CYTED), Santiago, Chile
| | - Yessika Méndez
- Servicio Hemato Oncológico Pediátrico (SHOP), Centro Hospitalario Pereira Rossell (CHPR), Montevideo, Uruguay
| | - Natalia Olano
- Servicio Hemato Oncológico Pediátrico (SHOP), Centro Hospitalario Pereira Rossell (CHPR), Montevideo, Uruguay
| | - Magdalena Schelotto
- Servicio Hemato Oncológico Pediátrico (SHOP), Centro Hospitalario Pereira Rossell (CHPR), Montevideo, Uruguay
| | - Luis Castillo
- Servicio Hemato Oncológico Pediátrico (SHOP), Centro Hospitalario Pereira Rossell (CHPR), Montevideo, Uruguay
| | - Ana María Soler
- Laboratorio de Genética Molecular Humana, Departamento de Ciencias Biológicas, CENUR Litoral Norte-Sede Salto, Universidad de la República, Salto, Uruguay
- Red Latinoamericana de Implementación y Validación de Guías Clínicas Farmacogenómicas (RELIVAF-CYTED), Santiago, Chile
| | - Julio da Luz
- Laboratorio de Genética Molecular Humana, Departamento de Ciencias Biológicas, CENUR Litoral Norte-Sede Salto, Universidad de la República, Salto, Uruguay
- Red Latinoamericana de Implementación y Validación de Guías Clínicas Farmacogenómicas (RELIVAF-CYTED), Santiago, Chile
| |
Collapse
|
28
|
Guo Q, Li J, Mao J, Chen W, Yang M, Yang Y, Hua Y, Qiu L. Hollow MIL-125 Nanoparticles Loading Doxorubicin Prodrug and 3-Methyladenine for Reversal of Tumor Multidrug Resistance. J Funct Biomater 2023; 14:546. [PMID: 37998115 PMCID: PMC10671911 DOI: 10.3390/jfb14110546] [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: 09/08/2023] [Revised: 10/13/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Multidrug resistance (MDR) is a key factor in chemotherapy failure and tumor recurrence. The inhibition of drug efflux and autophagy play important roles in MDR therapy. Herein, a multifunctional delivery system (HA-MIL-125@DVMA) was prepared for synergistically reverse tumor MDR. Tumor-targeted hollow MIL-125-Ti nanoparticles were used to load the doxorubicin-vitamin E succinate (DV) prodrug and 3-methyladenine (3-MA) to enhance reverse MDR effects. The pH-sensitive DV can kill tumor cells and inhibit P-gp-mediated drug efflux, and 3-MA can inhibit autophagy. HA-MIL-125@DVMA had uniformly distributed particle size and high drug-load content. The nanoparticles could effectively release the drugs into tumor microenvironment due to the rapid hydrazone bond-breaking under low pH conditions, resulting in a high cumulative release rate. In in vitro cellular experiments, the accumulation of HA-MIL-125@DVMA and HA-MIL-125@DV in MCF-7/ADR cells was significantly higher than that in the control groups. Moreover, the nanoparticles significantly inhibited drug efflux in the cells, ensuring the accumulation of the drugs in cell cytoplasm and causing drug-resistant cells' death. Importantly, HA-MIL-125@DVMA effectively inhibited tumor growth without changes in body weight in tumor-bearing mice. In summary, the combination of the acid-sensitive prodrug DV and autophagy inhibitor 3-MA in a HA-MIL-125 nanocarrier can enhance the antitumor effect and reverse tumor MDR.
Collapse
Affiliation(s)
- Qingfeng Guo
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214122, China;
| | - Jie Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Jing Mao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Meiyang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Yang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Yuming Hua
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214122, China;
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| |
Collapse
|
29
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
30
|
Chen JY, Sung CJ, Chen SC, Hsiang YP, Hsu YC, Teng YN. Redefine the role of d-α-Tocopheryl polyethylene glycol 1000 succinate on P-glycoprotein, multidrug resistance protein 1, and breast cancer resistance protein mediated cancer multidrug resistance. Eur J Pharm Sci 2023; 190:106579. [PMID: 37689120 DOI: 10.1016/j.ejps.2023.106579] [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: 04/21/2023] [Revised: 07/22/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Cancer drug resistance is an ever-changing problem that most patients need to face in their later stages of treatment, especially the multidrug resistant (MDR) type. The drug efflux transporters, including P-glycoprotein (P-gp), multidrug resistance protein 1 (MRP1), and breast cancer resistance protein (BCRP), play the crucial roles in this sophisticated battle. In recent decades, researchers try to find potential inhibitors to impede the drug efflux function of above transporters. d-α-Tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS) is a prevalently used excipient in the formulation design. In the present study, the modulatory effects and mechanisms of vitamin E TPGS on the efflux transporters were investigated. And the cancer MDR reversing ability of vitamin E TPGS was evaluated as well. Stable-cloned transporter over-expressed cell lines were used for mechanisms study, while several types of MDR cancer cell lines were adopted as reversing evaluation models. The results exhibited that vitamin E TPGS significantly inhibited the efflux function of P-gp, MRP1, and BCRP under non-cytotoxic concentrations, but not influencing the protein expression levels. Through efflux assay and molecular docking, vitamin E TPGS was found to be an uncompetitive, non-competitive, and competitive inhibitor on chemotherapeutic drug doxorubicin efflux in P-gp, MRP1, and BCRP over-expressing cell lines, respectively. Furthermore, the basal ATPase activity of three transporters were significantly inhibited by vitamin E TPGS at 10 μM. And the cell membrane fluidity of P-gp over-expressing cell line was enhanced by 22.58% with 5 μM vitamin E TPGS treatment, compared to the parental Flp-In™-293 cell line (without P-gp). The resistance reversing ability of vitamin E TPGS was prominent in MCF-7/DOX MDR breast cancer cell line, which over-expressed P-gp, MRP1, and BCRP. These significant results suggested that vitamin E TPGS is a promising modulator on transporters mediated cancer MDR. Vitamin E TPGS is not an inert excipient, but possesses MDR-reversing pharmacological effects, and deserves a re-purposing application on the future combinatorial regimen design for MDR cancer treatment.
Collapse
Affiliation(s)
- Jing-Yi Chen
- Department of Medical Laboratory Science, College of medical science and technology, I-Shou University, 8 Yida Road, Kaohsiung 82445, Taiwan, R.O.C; School of Medicine for International Students, College of Medicine, I-Shou University, 8 Yida Road, Kaohsiung 82445, Taiwan, R.O.C
| | - Chieh-Ju Sung
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, No.1 Jen Ai road section 1, Taipei 100233, Taiwan, R.O.C
| | - Ssu-Chi Chen
- School of Medicine, College of Medicine, I-Shou University, 8 Yida Road, Kaohsiung 82445, Taiwan, R.O.C
| | - Yi-Ping Hsiang
- Department of Pharmacy, E-Da Hospital, 1 Yida Road, Kaohsiung 82445, Taiwan, R.O.C
| | - Yung-Chia Hsu
- Department of Pharmacy, E-Da Cancer Hospital, 21 Yida Road, Kaohsiung 82445, Taiwan, R.O.C
| | - Yu-Ning Teng
- School of Medicine, College of Medicine, I-Shou University, 8 Yida Road, Kaohsiung 82445, Taiwan, R.O.C; Department of Pharmacy, E-Da Cancer Hospital, 21 Yida Road, Kaohsiung 82445, Taiwan, R.O.C.
| |
Collapse
|
31
|
Hsieh CY, Lin CC, Chang WC. Taxanes in the Treatment of Head and Neck Squamous Cell Carcinoma. Biomedicines 2023; 11:2887. [PMID: 38001888 PMCID: PMC10669519 DOI: 10.3390/biomedicines11112887] [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: 09/21/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Taxanes, particularly docetaxel (DTX), has been widely used for combination therapy of head and neck squamous cell carcinoma (HNSCC). For locally advanced unresectable HNSCC, DTX combined with cisplatin and 5-fluorouracil as a revolutionary treatment revealed an advantage in the improvement of patient outcome. In addition, DTX plus immune check inhibitors (ICIs) showed low toxicity and an increased response of patients with recurrent or metastatic HNSCC (R/M HNSCC). Accumulated data indicate that taxanes not only function as antimitotics but also impair diverse oncogenic signalings, including angiogenesis, inflammatory response, ROS production, and apoptosis induction. However, despite an initial response, the development of resistance remains a major obstacle to treatment response. Taxane resistance could result from intrinsic mechanisms, such as enhanced DNA/RNA damage repair, increased drug efflux, and apoptosis inhibition, and extrinsic effects, such as angiogenesis and interactions between tumor cells and immune cells. This review provides an overview of taxanes therapy applied in different stages of HNSCC and describe the mechanisms of taxane resistance in HNSCC. Through a detailed understanding, the mechanisms of resistance may help in developing the potential therapeutic methods and the effective combination strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan;
| | - Ching-Chan Lin
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan;
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
32
|
Budagaga Y, Sabet Z, Zhang Y, Novotná E, Hanke I, Rozkoš T, Hofman J. Tazemetostat synergistically combats multidrug resistance by the unique triple inhibition of ABCB1, ABCC1, and ABCG2 efflux transporters in vitro and ex vivo. Biochem Pharmacol 2023; 216:115769. [PMID: 37634597 DOI: 10.1016/j.bcp.2023.115769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
ATP-binding cassette (ABC) drug efflux transporters and drug metabolizing enzymes play crucial roles in pharmacokinetic drug-drug interactions and multidrug tumor resistance (MDR). Tazemetostat (EPZ-6438, Tazverik) is a novel epigenetic drug that has been recently approved for the therapy of advanced epithelioid sarcoma and follicular lymphoma. Additionally, this medication is currently being clinically tested to treat several other cancers such as non-small cell lung cancer (NSCLC). This study aimed to investigate the inhibitory effects of tazemetostat on selected ABC transporters/cytochrome P450 3A4 (CYP3A4) enzyme to comprehensively explore its role in MDR. First, our accumulation and molecular docking studies showed that tazemetostat is a unique triple inhibitor of ABCB1, ABCC1, and ABCG2 transporters. In contrast, tazemetostat exhibited only low level of interaction with the CYP3A4 isozyme. Drug combination assays confirmed that tazemetostat is a multipotent MDR modulator able to synergize with various conventional chemotherapeutics in vitro. Subsequent caspase activity assays and microscopic staining of apoptotic nuclei proved that the effective induction of apoptosis is behind the observed synergies. Notably, a potent MDR-modulatory capacity of tazemetostat was recorded in primary ex vivo NSCLC explants generated from patients' biopsies. On the contrary, its possible position of pharmacokinetic MDR's victim was excluded in comparative proliferation assays. Finally, tested drug has not been identified as an inducer of resistant phenotype in NSCLC cell lines. In conclusion, we demonstrated that tazemetostat is a unique multispecific chemosensitizer, which has strong potential to overcome limitations seen in the era of traditional MDR modulators.
Collapse
Affiliation(s)
- Youssif Budagaga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Ziba Sabet
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Yu Zhang
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Eva Novotná
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Ivo Hanke
- Department of Cardiac Surgery, Faculty of Medicine, Charles University in Hradec Králové and University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Rozkoš
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine and University Hospital in Hradec Králové, Czech Republic, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic.
| |
Collapse
|
33
|
Ismailov ZB, Belykh ES, Chernykh AA, Udoratina AM, Kazakov DV, Rybak AV, Kerimova SN, Velegzhaninov IO. Systematic review of comparative transcriptomic studies of cellular resistance to genotoxic stress. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 792:108467. [PMID: 37657754 DOI: 10.1016/j.mrrev.2023.108467] [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: 01/07/2023] [Revised: 08/19/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
The development of resistance by tumor cells to various types of therapy is a significant problem that decreases the effectiveness of oncology treatments. For more than two decades, comparative transcriptomic studies of tumor cells with different sensitivities to ionizing radiation and chemotherapeutic agents have been conducted in order to identify the causes and mechanisms underlying this phenomenon. However, the results of such studies have little in common and often contradict each other. We have assumed that a systematic analysis of a large number of such studies will provide new knowledge about the mechanisms of development of therapeutic resistance in tumor cells. Our comparison of 123 differentially expressed gene (DEG) lists published in 98 papers suggests a very low degree of consistency between the study results. Grouping the data by type of genotoxic agent and tumor type did not increase the similarity. The most frequently overexpressed genes were found to be those encoding the transport protein ABCB1 and the antiviral defense protein IFITM1. We put forward a hypothesis that the role played by the overexpression of the latter in the development of resistance may be associated not only with the stimulation of proliferation, but also with the limitation of exosomal communication and, as a result, with a decrease in the bystander effect. Among down regulated DEGs, BNIP3 was observed most frequently. The expression of BNIP3, together with BNIP3L, is often suppressed in cells resistant to non-platinum genotoxic chemotherapeutic agents, whereas it is increased in cells resistant to ionizing radiation. These observations are likely to be mediated by the binary effects of these gene products on survival, and regulation of apoptosis and autophagy. The combined data also show that even such obvious mechanisms as inhibition of apoptosis and increase of proliferation are not universal but show multidirectional changes.
Collapse
Affiliation(s)
- Z B Ismailov
- Institute of Biology of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia
| | - E S Belykh
- Institute of Biology of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia
| | - A A Chernykh
- Institute of Physiology of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, 50 Pervomaiskaya St., Syktyvkar 167982, Russia
| | - A M Udoratina
- Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, Nizhny Novgorod 603022, Russia
| | - D V Kazakov
- Institute of Physics and Mathematics of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, 4 Oplesnina St., Syktyvkar 167982, Russia
| | - A V Rybak
- Institute of Biology of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia
| | - S N Kerimova
- State Medical Institution Komi Republican Oncology Center, 46 Nyuvchimskoe highway, Syktyvkar 167904, Russia
| | - I O Velegzhaninov
- Institute of Biology of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia.
| |
Collapse
|
34
|
Sun X, Zhao P, Lin J, Chen K, Shen J. Recent advances in access to overcome cancer drug resistance by nanocarrier drug delivery system. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:390-415. [PMID: 37457134 PMCID: PMC10344729 DOI: 10.20517/cdr.2023.16] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023]
Abstract
Cancer is currently one of the most intractable diseases causing human death. Although the prognosis of tumor patients has been improved to a certain extent through various modern treatment methods, multidrug resistance (MDR) of tumor cells is still a major problem leading to clinical treatment failure. Chemotherapy resistance refers to the resistance of tumor cells and/or tissues to a drug, usually inherent or developed during treatment. Therefore, an urgent need to research the ideal drug delivery system to overcome the shortcoming of traditional chemotherapy. The rapid development of nanotechnology has brought us new enlightenments to solve this problem. The novel nanocarrier provides a considerably effective treatment to overcome the limitations of chemotherapy or other drugs resulting from systemic side effects such as resistance, high toxicity, lack of targeting, and off-target. Herein, we introduce several tumor MDR mechanisms and discuss novel nanoparticle technology applied to surmount cancer drug resistance. Nanomaterials contain liposomes, polymer conjugates, micelles, dendrimers, carbon-based, metal nanoparticles, and nucleotides which can be used to deliver chemotherapeutic drugs, photosensitizers, and small interfering RNA (siRNA). This review aims to elucidate the advantages of nanomedicine in overcoming cancer drug resistance and discuss the latest developments.
Collapse
Affiliation(s)
- Xiangyu Sun
- Medicines and Equipment Department, Beijing Chaoyang Emergency Medical Rescuing Center, Chaoyang District, Beijing 100026, China
| | - Ping Zhao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Meg Centre, Guangzhou 510006, Guangdong, China
| | - Jierou Lin
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Meg Centre, Guangzhou 510006, Guangdong, China
| | - Kun Chen
- Beijing Chaoyang Emergency Medical Rescuing Center, Chaoyang District, Beijing 100026, China
| | - Jianliang Shen
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China
| |
Collapse
|
35
|
Yu CP, Wang YR, Hou YC, Hsieh MT, Li PY, Kuo SC, Lin SP. Two curcumin analogs inhibited the function and protein expression of breast cancer resistance protein: in vitro and in vivo studies. Xenobiotica 2023; 53:454-464. [PMID: 37728540 DOI: 10.1080/00498254.2023.2260886] [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: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023]
Abstract
1. Two curcumin analogs, (1E,6E)-1,7-bis(3,5-diethyl-4-hydroxyphenyl)hepta-1,6-diene-3,5- dione (N17) and its prodrug ((1E,6E)-3,5-dioxohepta-1,6-diene-1,7-diyl)bis(2,6-diethyl-4,1- phenylene)bis(3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate) (N17'), were evaluated as breast cancer resistance protein (BCRP) inhibitors.2. MDCKII-BCRP and MDCKII-WT were used to evaluate the modulation effects of N17 and N17' on BCRP and to explore the relevant mechanism. Sprague-Dawley rats were orally administered rosuvastatin (ROS), a probe substrate of BCRP, without and with N17' (100 mg/kg) to investigate the effect of N17' on ROS pharmacokinetics.3. In cell studies, N17 and N17' were substrates of BCRP, and they decreased the activity and protein expression of BCRP. In rat study, N17' increased the systemic exposure of ROS by 218% (p = 0.058).4. N17 and N17' are potential BCRP inhibitors and will be promising candidates for overcoming the BCRP-mediated multidrug resistance.
Collapse
Affiliation(s)
- Chung-Ping Yu
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Yi-Ru Wang
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Yu-Chi Hou
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Min-Tsang Hsieh
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
- ResearchCenter for Chinese Herbal Medicine, China Medical University, Taichung, Taiwan, ROC
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Pei-Ying Li
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
| | - Sheng-Chu Kuo
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
- Chinese Medicinal Research and Development Center, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Shiuan-Pey Lin
- School of Pharmacy, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
36
|
Mineiro R, Santos C, Gonçalves I, Lemos M, Cavaco JEB, Quintela T. Regulation of ABC transporters by sex steroids may explain differences in drug resistance between sexes. J Physiol Biochem 2023:10.1007/s13105-023-00957-1. [PMID: 36995571 DOI: 10.1007/s13105-023-00957-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/09/2023] [Indexed: 03/31/2023]
Abstract
Drug efficacy is dependent on the pharmacokinetics and pharmacodynamics of therapeutic agents. Tight junctions, detoxification enzymes, and drug transporters, due to their localization on epithelial barriers, modulate the absorption, distribution, and the elimination of a drug. The epithelial barriers which control the pharmacokinetic processes are sex steroid hormone targets, and in this way, sex hormones may also control the drug transport across these barriers. Thus, sex steroids contribute to sex differences in drug resistance and have a relevant impact on the sex-related efficacy of many therapeutic drugs. As a consequence, for the further development and optimization of therapeutic strategies, the sex of the individuals must be taken into consideration. Here, we gather and discuss the evidence about the regulation of ATP-binding cassette transporters by sex steroids, and we also describe the signaling pathways by which sex steroids modulate ATP-binding cassette transporters expression, with a focus in the most important ATP-binding cassette transporters involved in multidrug resistance.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Cecília Santos
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Manuel Lemos
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - José Eduardo B Cavaco
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique. 6200-506, Covilhã, Portugal.
- UDI-IPG-Unidade de Investigação Para o Desenvolvimento Do Interior, Instituto Politécnico da Guarda, Guarda, Portugal.
| |
Collapse
|
37
|
Samavarchi Tehrani S, Esmaeili F, Shirzad M, Goodarzi G, Yousefi T, Maniati M, Taheri-Anganeh M, Anushiravani A. The critical role of circular RNAs in drug resistance in gastrointestinal cancers. Med Oncol 2023; 40:116. [PMID: 36917431 DOI: 10.1007/s12032-023-01980-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Nowadays, drug resistance (DR) in gastrointestinal (GI) cancers, as the main reason for cancer-related mortality worldwide, has become a serious problem in the management of patients. Several mechanisms have been proposed for resistance to anticancer drugs, including altered transport and metabolism of drugs, mutation of drug targets, altered DNA repair system, inhibited apoptosis and autophagy, cancer stem cells, tumor heterogeneity, and epithelial-mesenchymal transition. Compelling evidence has revealed that genetic and epigenetic factors are strongly linked to DR. Non-coding RNA (ncRNA) interferences are the most crucial epigenetic alterations explored so far, and among these ncRNAs, circular RNAs (circRNAs) are the most emerging members known to have unique properties. Due to the absence of 5' and 3' ends in these novel RNAs, the two ends are covalently bonded together and are generated from pre-mRNA in a process known as back-splicing, which makes them more stable than other RNAs. As far as the unique structure and function of circRNAs is concerned, they are implicated in proliferation, migration, invasion, angiogenesis, metastasis, and DR. A clear understanding of the molecular mechanisms responsible for circRNAs-mediated DR in the GI cancers will open a new window to the management of GI cancers. Hence, in the present review, we will describe briefly the biogenesis, multiple features, and different biological functions of circRNAs. Then, we will summarize current mechanisms of DR, and finally, discuss molecular mechanisms through which circRNAs regulate DR development in esophageal cancer, pancreatic cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fataneh Esmaeili
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Moein Shirzad
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmood Maniati
- Department of English, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Amir Anushiravani
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Sharma P, Singh N, Sharma S. Genetic variations in ABC transporter genes as a predictive biomarker for toxicity in North Indian lung cancer patients undergoing platinum-based doublet chemotherapy. J Biochem Mol Toxicol 2023; 37:e23269. [PMID: 36507589 DOI: 10.1002/jbt.23269] [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: 05/13/2022] [Revised: 09/26/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
ATP-binding cassette (ABC) transporters are expressed in various human tissues and play a vital role in the efflux of various chemotherapeutic drugs. The current study has assessed genetic variants of ABCB1, ABCC1, ABCC2, and ABCG2 genes in 407 lung cancer patients undergoing platinum-based doublet chemotherapy. The association of ABCB1 (C1236 T, C3435 T, and G2677 T/A), ABCC1 (G3173 A and G2168 A),ABCC2 (G4544 A), and ABCG2 (C421 A) polymorphisms with chemotherapy-induced adverse events were assessed, and statistical analysis was conducted. Our data showed that patients harboring heterozygous (GA) genotype for ABCC1 G3173 A had an increased risk of developing leukopenia (odds ratio [OR] = 1.88, p = 0.04) and anemia (adjusted odds ratio [AOR] = 2.70, p = 0.03). For ABCC2 G4544 A polymorphism, patients harboring one copy of the mutant (GA) allele showed an increased risk of developing anemia (OR = 4.24, p = 0.03). After adjusting with various confounding factors, the heterozygous (GA) genotype showed a 5.63-fold increased risk of developing anemia (AOR = 5.63, p = 0.03). The ABCB1 G2677 A (OR = 0.37, p = 0.008) and ABCC1 G3173 A (OR = 0.54, p = 0.04) polymorphism showed a lower incidence of developing nephrotoxicity. In ABCG2 C421 A polymorphism, patients harboring heterozygous (CA) genotype had a lower incidence of having diarrhea (OR = 0.25, p = 0.04). An increased risk of having diarrhea was observed in the heterozygous genotype (GA) for ABCC1 G3173 A polymorphism (AOR = 2.78, p = 0.04). An increased risk of liver injury was found in the patients carrying heterozygous genotype of the ABCC1 G3173 A (OR = 2.06, p = 0.02) and ABCB1 C1236 T (OR = 1.85, p = 0.01). This study demonstrates the role of polymorphic variations in ABCB1, ABCC1, ABCC2, and ABCG2 in predicting hematological, nephrotoxicity, gastrointestinal, and hepatotoxicity.
Collapse
Affiliation(s)
- Parul Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, India
| |
Collapse
|
39
|
ATP-binding cassette efflux transporters and MDR in cancer. Drug Discov Today 2023; 28:103537. [PMID: 36801375 DOI: 10.1016/j.drudis.2023.103537] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/27/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Of the many known multidrug resistance (MDR) mechanisms, ATP-binding cassette (ABC) transporters expelling drug molecules out of cells is a major factor limiting the efficacy of present-day anticancer drugs. In this review, we highlights updated information on the structure, function, and regulatory mechanisms of major MDR-related ABC transporters, such as P-glycoprotein (P-gp), multidrug resistance protein 1 (MRP1), and breast cancer resistance protein (BCRP), and the effect of modulators on their functions. We also provide focused information on different modulators of ABC transporters that could be utilized against the emerging MDR crisis in cancer treatment. Finally, we discuss the importance of ABC transporters as therapeutic targets in light of future strategic planning for translating ABC transporter inhibitors into clinical practice.
Collapse
|
40
|
Anti-colon cancer effects of Spirulina polysaccharide and its mechanism based on 3D models. Int J Biol Macromol 2023; 228:559-569. [PMID: 36581031 DOI: 10.1016/j.ijbiomac.2022.12.244] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/03/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Spirulina polysaccharides (PSP) possess significant biological properties. However, it is still a lack of investigation on the anti-colorectal cancer effect and mechanism. In this study, PSP showed significant effects on LoVo cell spheroids with an IC50 value of 0.1943 mg/mL. The analysis of transcriptomics and metabolomics indicated the impact of PSP on LoVo spheroid cells through involvement in the two pathways of "glycine, serine, and threonine metabolism" and "ABC transporters". And, the q-PCR data further verified the pointed mechanism of PSP on colon cancer (CC) by regulating the expression levels of relevant genes in the synthesis pathways of serine and glycine in tumor cells. Furthermore, the anti-colon cancer effects of PSP were verified via other human colon cancer cell lines HCT116 and HT29 spheroids (IC50 = 0.0646 mg/mL and 0.2213 mg/mL, respectively), and three patient-derived organoids (PDOs) with IC50 values ranging from 3.807 to 7.788 mg/mL. In addition, this study found that a mild concentration of PSP cannot enhance the anti-tumor effect of 5-Fu. And a significant inhibition was found of PSP in 5-Fu resistance organoids. These results illustrated that PSP could be a treatment or supplement for 5-Fu resistant colorectal cancer (CRC).
Collapse
|
41
|
Ahmad S, Gupta D, Ahmed T, Islam A. Designing of new tetrahydro-β-carboline-based ABCG2 inhibitors using 3D-QSAR, molecular docking, and DFT tools. J Biomol Struct Dyn 2023; 41:14016-14027. [PMID: 36752362 DOI: 10.1080/07391102.2023.2176361] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023]
Abstract
Human ATP-binding cassette superfamily G member 2 (ABCG2) protein is a member of the ABC transporter family, which is responsible for multidrug resistance (MDR) in cancerous cells. MDR reduces the effectiveness of chemotherapy in breast cancer, which is one of the leading causes of death in women globally. MDR in cancer cells is one of the immediate signs of progression of resistance; thus, various anticancer drugs can be designed. To reduce MDR, we utilized the tetrahydro-β-carboline (THβC) compound library. We accomplished a three-dimensional quantitative structure-activity relationship (3D-QSAR), scaffold hopping to design a new library of compounds of THβC, and further molecular docking, induced-fit docking (IFD), molecular mechanics energies combined with generalized born and surface area continuum solvation (MM-GBSA), drug-like features, ADMET properties, and density functional theory (DFT) studies were performed. From these studies, the best 3D-QSAR model (r2 = 0.99, q2 = 0.92) was found, and the necessity of electrostatic, steric, and hydrophobic field effects were determined that could modulate bioactivity. Moreover, based on electrostatic, steric, and hydrophobic field notations, new THβC derivatives (3409) were designed. These findings might provide new insight for researchers to perform in vitro and in vivo studies for better antagonists against MDR in treating breast cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shahnawaz Ahmad
- School of Biotechnology, College of Engineering and Technology, IFTM University, Moradabad, Uttar Pradesh, India
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi, India
| | - Tanzeel Ahmed
- School of Biotechnology, College of Engineering and Technology, IFTM University, Moradabad, Uttar Pradesh, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
42
|
QSAR modeling and molecular docking studies of 2-oxo-1, 2-dihydroquinoline-4- carboxylic acid derivatives as p-glycoprotein inhibitors for combating cancer multidrug resistance. Heliyon 2023; 9:e13020. [PMID: 36747956 PMCID: PMC9898678 DOI: 10.1016/j.heliyon.2023.e13020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023] Open
Abstract
Multidrug resistance (MDR) proteins related to the ATP-binding cassette family are found in a very wide range of human tumors and result in therapeutic failure. The overexpression of efflux pumps such as ABCB1 is one of the mechanisms of MDR. This paper aims to develop a reliable quantitative structure-activity relationship (QSAR) model that best describes the correlation between the activity and the molecular structures in order to predict the inhibitory biological activity towards ABCB1. In this regard, a series of quinoline derivatives of 18 compounds were analyzed using different linear and non-linear machine learning (ML) regression methods including k-nearest neighbors (KNN), decision tree (DT), back propagation neural networks (BPNN) and gradient boosting-based (GB) methods. Their aim is to explain the origin of the activity of these investigated compounds and therefore, design new quinoline derivatives with higher effect on ABCB1. A total of 16 ML predictive models were developed on different number of 2D and 3D descriptors and were evaluated using the coefficient of determination (R2) and the root mean squared error (RMSE) statistical metrics. Among all developed models, A GB-based model in particular catboost achieved the highest predictive quality, with one descriptor, expressed by R2 and RMSE of 95% and 0.283 respectively. Molecular docking studies against the target crystal structure of the outward-facing p-glycoprotein (6C0V) revealed significant binding affinities via both hydrophobic and H-bond interactions with the relevant compounds. The 17 has shown the highest binding energy of -9.22 kcal/mol. Therefore, it can suggest that 17 may prove to be a valuable potential lead structure for the design and synthesis of more potent P-glycoprotein inhibitors for combination used with anti-cancer drugs for cancer multidrug resistance management.
Collapse
|
43
|
Repurposing Antidepressants and Phenothiazine Antipsychotics as Efflux Pump Inhibitors in Cancer and Infectious Diseases. Antibiotics (Basel) 2023; 12:antibiotics12010137. [PMID: 36671340 PMCID: PMC9855052 DOI: 10.3390/antibiotics12010137] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Multidrug resistance (MDR) is a major obstacle in the therapy of infectious diseases and cancer. One of the major mechanisms of MDR is the overexpression of efflux pumps (EPs) that are responsible for extruding antimicrobial and anticancer agents. EPs have additional roles of detoxification that may aid the development of bacterial infection and the progression of cancer. Therefore, targeting EPs may be an attractive strategy to treat bacterial infections and cancer. The development and discovery of a new drug require a long timeline and may come with high development costs. A potential alternative to reduce the time and costs of drug development is to repurpose already existing drugs. Antidepressants and antipsychotic agents are widely used in clinical practice in the treatment of psychiatric disorders and some somatic diseases. Antidepressants and antipsychotics have demonstrated various beneficial activities that may be utilized in the treatment of infections and cancer. This review aims to provide a brief overview of antibacterial and anticancer effects of selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs) and phenothiazine antipsychotics, while focusing on EPs. However, it should be noted that the antimicrobial activity of a traditionally non-antibiotic drug may have clinical implications regarding dysbiosis and bacterial MDR.
Collapse
|
44
|
Voci S, Gagliardi A, Ambrosio N, Salvatici MC, Fresta M, Cosco D. Gliadin Nanoparticles Containing Doxorubicin Hydrochloride: Characterization and Cytotoxicity. Pharmaceutics 2023; 15:pharmaceutics15010180. [PMID: 36678809 PMCID: PMC9860592 DOI: 10.3390/pharmaceutics15010180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Doxorubicin hydrochloride (DOX) is a well-known antitumor drug used as first line treatment for many types of malignancies. Despite its clinical relevance, the administration of the compound is negatively affected by dose-dependent off-target toxicity phenomena. Nanotechnology has helped to overcome these important limitations by improving the therapeutic index of the bioactive and promoting the translation of novel nanomedicines into clinical practice. Herein, nanoparticles made up of wheat gliadin and stabilized by polyoxyethylene (2) oleyl ether were investigated for the first time as carriers of DOX. The encapsulation of the compound did not significantly affect the physico-chemical features of the gliadin nanoparticles (GNPs), which evidenced a mean diameter of ~180 nm, a polydispersity index < 0.2 and a negative surface charge. The nanosystems demonstrated great stability regarding temperature (25−50 °C) and were able to retain high amounts of drug, allowing its prolonged and sustained release for up to a week. In vitro viability assay performed against breast cancer cells demonstrated that the nanoencapsulation of DOX modulated the cytotoxicity of the bioactive as a function of the incubation time with respect to the free form of the drug. The results demonstrate the potential use of GNPs as carriers of hydrophilic antitumor compounds.
Collapse
Affiliation(s)
- Silvia Voci
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S Venuta”, 88100 Catanzaro, Italy
| | - Agnese Gagliardi
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S Venuta”, 88100 Catanzaro, Italy
| | - Nicola Ambrosio
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S Venuta”, 88100 Catanzaro, Italy
| | - Maria Cristina Salvatici
- Institute of Chemistry of Organometallic Compounds (ICCOM)-Electron Microscopy Centre (Ce.M.E.), National Research Council (CNR), Via Madonna del Piano n. 10, Sesto Fiorentino, 50019 Florence, Italy
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S Venuta”, 88100 Catanzaro, Italy
| | - Donato Cosco
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S Venuta”, 88100 Catanzaro, Italy
- Correspondence: ; Tel.: +39-0961-369-4119
| |
Collapse
|
45
|
A New ABCB1 Inhibitor Enhances the Anticancer Effect of Doxorubicin in Both In Vitro and In Vivo Models of NSCLC. Int J Mol Sci 2023; 24:ijms24020989. [PMID: 36674503 PMCID: PMC9861803 DOI: 10.3390/ijms24020989] [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: 12/16/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
In tumors, the multi drug resistance phenomenon may occur through the efflux of chemotherapeutic drugs out of cancer cells, impeding their accumulation, and eventually reducing their toxicity. This process is mediated by transporters overexpressed in the plasma membranes of tumor cells, among which is the P-glycoprotein/multidrug resistance 1/ATP-binding cassette B1 (P-gp/MDR1/ABCB1). The aim of this study was to explore the effect of a new molecule, called AIF-1, on ABCB1 activity. In a cellular model of non-small cell lung cancer (NSCLC), AIF-1 significantly inhibited ABCB1 activity, which was evaluated by the fluorimetric measurement of the intracellular accumulation of calcein. AIF-1 also significantly increased the intracellular content of doxorubicin, which was evaluated by confocal microscopy and LC-MS/MS analysis. This effect translated to higher cytotoxicity of doxorubicin and reduced cellular proliferation. Finally, in a murine xenograft model, the tumor volume increased by 267% and 148% on average in mice treated with vehicle and doxorubicin alone, respectively. After the co-administration of doxorubicin with AIF-1, tumor volume increased by only 13.4%. In conclusion, these results suggest enhancement of the efficacy of the chemotherapeutic drug doxorubicin by AIF-1, laying the basis for the future development of new ABCB1 inhibitors for tumor treatment.
Collapse
|
46
|
Kaehler M, Cascorbi I. Molecular Mechanisms of Tyrosine Kinase Inhibitor Resistance in Chronic Myeloid Leukemia. Handb Exp Pharmacol 2023; 280:65-83. [PMID: 36882601 DOI: 10.1007/164_2023_639] [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: 03/09/2023]
Abstract
The hematopoietic neoplasm chronic myeloid leukemia (CML) is a rare disease caused by chromosomal reciprocal translocation t(9;22)(q34:q11) with subsequent formation of the BCR-ABL1 fusion gene. This fusion gene encodes a constitutively active tyrosine kinase, which results in malignant transformation of the cells. Since 2001, CML can be effectively treated using tyrosine kinase inhibitors (TKIs) such as imatinib, which prevent phosphorylation of downstream targets by blockade of the BCR-ABL kinase. Due to its tremendous success, this treatment became the role model of targeted therapy in precision oncology. Here, we review the mechanisms of TKI resistance focusing on BCR-ABL1-dependent and -independent mechanisms. These include the genomics of the BCR-ABL1, TKI metabolism and transport and alternative signaling pathways.
Collapse
Affiliation(s)
- Meike Kaehler
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
47
|
Wu J, Feng J, Zhang Q, He Y, Xu C, Wang C, Li W. Epigenetic regulation of stem cells in lung cancer oncogenesis and therapy resistance. Front Genet 2023; 14:1120815. [PMID: 37144123 PMCID: PMC10151750 DOI: 10.3389/fgene.2023.1120815] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/22/2023] [Indexed: 05/06/2023] Open
Abstract
Epigenetics plays an important role in regulating stem cell signaling, as well as in the oncogenesis of lung cancer and therapeutic resistance. Determining how to employ these regulatory mechanisms to treat cancer is an intriguing medical challenge. Lung cancer is caused by signals that cause aberrant differentiation of stem cells or progenitor cells. The different pathological subtypes of lung cancer are determined by the cells of origin. Additionally, emerging studies have demonstrated that the occurrence of cancer treatment resistance is connected to the hijacking of normal stem cell capability by lung cancer stem cells, especially in the processes of drug transport, DNA damage repair, and niche protection. In this review, we summarize the principles of the epigenetic regulation of stem cell signaling in relation to the emergence of lung cancer and resistance to therapy. Furthermore, several investigations have shown that the tumor immune microenvironment in lung cancer affects these regulatory pathways. And ongoing experiments on epigenetics-related therapeutic strategies provide new insight for the treatment of lung cancer in the future.
Collapse
Affiliation(s)
- Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiaming Feng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Qiran Zhang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yazhou He
- Department of oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuan Xu
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
- *Correspondence: Weimin Li, ; Chengdi Wang,
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, Med-X Center for Manufacturing, Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
- *Correspondence: Weimin Li, ; Chengdi Wang,
| |
Collapse
|
48
|
Lukanov T, Ivanova M, Yankova P, Al Hadra B, Mihaylova A, Genova M, Svinarov D, Naumova E. Impact of CYP3A7, CYP2D6 and ABCC2/ABCC3 polymorphisms on tacrolimus steady state concentrations in Bulgarian kidney transplant recipients. BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2022.2081517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Tsvetelin Lukanov
- Department of Clinical Immunology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
- Department of Clinical Immunology and Stem Cell Bank, University Hospital Alexandrovska, Sofia, Bulgaria
| | - Milena Ivanova
- Department of Clinical Immunology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Petya Yankova
- Department of Clinical Immunology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Bushra Al Hadra
- Department of Clinical Immunology and Stem Cell Bank, University Hospital Alexandrovska, Sofia, Bulgaria
| | - Anastasiya Mihaylova
- Department of Clinical Immunology and Stem Cell Bank, University Hospital Alexandrovska, Sofia, Bulgaria
| | - Marianka Genova
- Department of Clinical Laboratory & Clinical Pharmacology, University Hospital Alexandrovska, Sofia, Bulgaria
- Department of Clinical Laboratory, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Dobrin Svinarov
- Department of Clinical Laboratory & Clinical Pharmacology, University Hospital Alexandrovska, Sofia, Bulgaria
- Department of Clinical Laboratory, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Elisaveta Naumova
- Department of Clinical Immunology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
49
|
Linke D, Donix L, Peitzsch C, Erb HHH, Dubrovska A, Pfeifer M, Thomas C, Fuessel S, Erdmann K. Comprehensive Evaluation of Multiple Approaches Targeting ABCB1 to Resensitize Docetaxel-Resistant Prostate Cancer Cell Lines. Int J Mol Sci 2022; 24:ijms24010666. [PMID: 36614114 PMCID: PMC9820728 DOI: 10.3390/ijms24010666] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Docetaxel (DTX) is a mainstay in the treatment of metastatic prostate cancer. Failure of DTX therapy is often associated with multidrug resistance caused by overexpression of efflux membrane transporters of the ABC family such as the glycoprotein ABCB1. This study investigated multiple approaches targeting ABCB1 to resensitize DTX-resistant (DTXR) prostate cancer cell lines. In DU145 DTXR and PC-3 DTXR cells as well as age-matched parental controls, the expression of selected ABC transporters was analyzed by quantitative PCR, Western blot, flow cytometry and immunofluorescence. ABCB1 effluxing activity was studied using the fluorescent ABCB1 substrate rhodamine 123. The influence of ABCB1 inhibitors (elacridar, tariquidar), ABCB1-specific siRNA and inhibition of post-translational glycosylation on DTX tolerance was assessed by cell viability and colony formation assays. In DTXR cells, only ABCB1 was highly upregulated, which was accompanied by a strong effluxing activity and additional post-translational glycosylation of ABCB1. Pharmacological inhibition and siRNA-mediated knockdown of ABCB1 completely resensitized DTXR cells to DTX. Inhibition of glycosylation with tunicamycin affected DTX resistance partially in DU145 DTXR cells, which was accompanied by a slight intracellular accumulation and decreased effluxing activity of ABCB1. In conclusion, DTX resistance can be reversed by various strategies with small molecule inhibitors representing the most promising and feasible approach.
Collapse
Affiliation(s)
- Dinah Linke
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Lukas Donix
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Claudia Peitzsch
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), 01307 Dresden, Germany
| | - Holger H. H. Erb
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
| | - Anna Dubrovska
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
| | - Manuel Pfeifer
- Institute of Legal Medicine, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Christian Thomas
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Susanne Fuessel
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
- Correspondence: ; Tel.: +49-351-458-14544
| | - Kati Erdmann
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
| |
Collapse
|
50
|
Murugaiyaa Pandiyan S, Shanmugaraj P, Manoharan JP, Vidyalakshmi S. A network pharmacological approach to reveal the multidrug resistance reversal and associated mechanisms of acetogenins against colorectal cancer. J Biomol Struct Dyn 2022; 40:13527-13546. [PMID: 34669561 DOI: 10.1080/07391102.2021.1990130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multidrug Resistance (MDR) in tumors is caused by the over-expression of ATP Binding Cassette transporter proteins such as Multidrug Resistance Protein 1 and Breast Cancer Resistance Protein 1. This in silico study focuses on identifying a MDR inhibitor among acetogenins (AGEs) of Annona muricata and also aims at predicting colorectal cancer (CRC) core targets of AGEs through a network pharmacological approach. Twenty-four AGEs were initially screened for their ADME properties. Molecular interaction studies were performed with the two proteins MRP1 and BCRP1. As the structure of MRP1 was not available, an inward-facing conformation of MRP1 was modeled. A Protein-protein interaction network was constructed for the correlating targets of CRC. KEGG pathway and Gene Ontology analysis were performed for the predicted CRC targets. We identified four lead AGEs: Muricatocin B, Annonacinone, Annonacin A and Annomuricin E having a higher binding affinity towards MDR proteins. MD simulation studies performed with the three lead AGEs and the MDR proteins showed that MRP1(DBD): Annomuricin E complex was stable throughout the simulation. Our analysis revealed ABCG2, ERBB2, STAT3, AR, SRC and ABCC1 as CRC targets of the lead molecules. The top 10 signaling pathways and functions of correlative CRC targets were also predicted. We conclude that the identified lead molecules might act as competitive inhibitors for reversing MDR in CRC. Additionally, network pharmacological studies established the correlative CRC targets and their mechanisms of action. Further experimental studies are needed to validate our findings. Communicated by Ramaswamy H. Sarma.
Collapse
|