1
|
Sarı ZB, Sarı ME, Aytar EC, Gümrükçüoğlu A, Torunoğlu EI, Ozdemir-Sanci T, Demirel G, Durmaz A. Flow Cytometry and Gene Expression Modulation by Euphorbia rigida Methanol Extract in A549 Lung Cancer Cells: Induction of Apoptosis Through Bax, Caspase-9, and Bcl-2 Pathways. Adv Biol (Weinh) 2025:e00136. [PMID: 40432491 DOI: 10.1002/adbi.202500136] [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: 02/13/2025] [Revised: 05/06/2025] [Indexed: 05/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) remains a major cause of cancer-related mortality. This study investigates the cytotoxic effects of Euphorbia rigida extract on A549 NSCLC cells and its potential as a therapeutic agent. Cellular morphology was observed microscopically, and cell viability was evaluated using dose-dependent proliferation assays. Apoptosis-related gene expression-including Bax, Bcl-2, and Caspase-9-was analyzed via quantitative PCR (qPCR). Chromatographic methods identified bioactive flavonoids, and molecular docking assessed their binding to cancer-related proteins. Additionally, absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles were evaluated. The extract induced apoptotic morphological changes such as cell shrinkage and loss of intercellular contact. A dose-dependent reduction in A549 viability was observed, with an IC50 of 0.5 mg mL-1. Gene expression indicated activation of the intrinsic mitochondrial apoptotic pathway, with increased Bax and Caspase-9 and decreased Bcl-2 expression. Flow cytometry using Annexin V-allophycocyanin (V-APC) staining revealed selective cytotoxicity: significant apoptosis in A549 cells while preserving viability in BEAS-2B normal lung epithelial cells. Identified flavonoids included quercetin, apigenin, and myricetin, which showed strong binding affinities in docking studies. ADMET profiling supported their drug-likeness. These findings highlight E. rigida potential in NSCLC treatment via apoptosis induction and selective cytotoxicity.
Collapse
Affiliation(s)
- Zeynep Betül Sarı
- Faculty of Medicine, Department of Basic Medical Sciences, Medical Biology, Ankara Yıldırım Beyazıt University, Ankara, 06010, Türkiye
| | - Muhammed Emin Sarı
- Faculty of Medicine, Department of Medical Biology, Necmettin Erbakan University, Konya, 42090, Türkiye
| | - Erdi Can Aytar
- Faculty of Agriculture, Department of Horticulture, Usak University, Uşak, 64200, Türkiye
| | - Abidin Gümrükçüoğlu
- Medicinal-Aromatic Plants Application and Research Center, Artvin Çoruh University, Artvin, 08000, Türkiye
| | - Emine Incilay Torunoğlu
- Faculty of Medicine, Department of Medical Biochemistry, Necmettin Erbakan University, Konya, 42090, Türkiye
| | - Tuba Ozdemir-Sanci
- Faculty of Medicine, Department of Histology and Embriyology, Ankara Yildirim Beyazit University, Ankara, 06010, Türkiye
| | - Gamze Demirel
- Selçuk University - Akşehir Kadir Yallagöz School of Health - Department of Nutrition and Dietetics, Konya, 42560, Türkiye
| | - Alper Durmaz
- Ali Nihat Gökyiğit Botanical Garden Application and Research Center, Artvin Çoruh University, Artvin, 08000, Türkiye
| |
Collapse
|
2
|
Chan SJW, Xiao Z, Soh WWM, Zhu JY, Lopez-Garcia F, Deng H, Bazan GC. Membrane Intercalation of a Conjugated Oligoelectrolyte Photosensitizer Enables Efficient Anticancer Photodynamic Therapy. Adv Healthc Mater 2025:e2501300. [PMID: 40411856 DOI: 10.1002/adhm.202501300] [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: 03/24/2025] [Revised: 05/07/2025] [Indexed: 05/26/2025]
Abstract
Photodynamic therapy (PDT) complements traditional chemotherapeutic methods by leveraging an external optical stimulus to target and eradicate tumor cells. Photosensitizers with favorable photophysical properties are, however, often hampered by poor biodistribution and inadequate cellular uptake, highlighting the need for novel molecular design strategies. Conjugated oligoelectrolytes (COEs) are a class of optical reporters that readily incorporate within cells due to their lipid bilayer mimicking molecular topology. Herein, a COE-derived photosensitizer, COE-PP, which features a central porphyrin core flanked by ionic pendant groups is disclosed. COE-PP has a solubility in aqueous media of >20 mm, yet the membrane-mimicking molecular topology enables high cellular uptake and prolonged retention of the COE in vitro and in vivo of up to 21 days. COE-PP is an effective phototheranostic agent for simultaneous tumor imaging and eradication, with the generation of reactive oxygen species (ROS) upon illumination. The membrane localization of COE-PP promotes cell death during PDT by ROS and through downstream pathways involving lysosomal membrane permeabilization. These synergistic effects enable effective treatment of COE-containing tumors. From a broad design perspective, the molecular architecture of COE-PP demonstrates the potential of utilizing lipid bilayer-mimicking molecular topologies to design phototheranostic molecules that offer spatiotemporal control for therapeutic interventions.
Collapse
Affiliation(s)
- Samuel Jun Wei Chan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Zhaolin Xiao
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Targeted Tracer Research and Development Laboratory Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wilson Wee Mia Soh
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Ji-Yu Zhu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
| | - Fernando Lopez-Garcia
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
| | - Hui Deng
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Targeted Tracer Research and Development Laboratory Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Guillermo Carlos Bazan
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
| |
Collapse
|
3
|
Muhammed Y, De Sabatino M, Lazenby RA. The Heterogeneity in the Response of A549 Cells to Toyocamycin Observed Using Hopping Scanning Ion Conductance Microscopy. J Phys Chem B 2025; 129:4904-4916. [PMID: 40338629 DOI: 10.1021/acs.jpcb.4c08793] [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: 05/09/2025]
Abstract
Scanning ion conductance microscopy (SICM) is a noninvasive topographic mapping technique used in imaging live cells, unlike electron microscopy and certain applications of fluorescence microscopy, which can disrupt cell integrity. In this study, we used SICM to track the morphological changes of the same A549 cells to uncover the cell-to-cell heterogeneity in their response to the drug. We found that toyocamycin (TOY) induced rapid reorganization of the actin cytoskeleton in A549 cells, causing them to become circular, irregular, or ellipsoidal in shape. Mapping of the dynamic changes in morphology revealed membrane blebbing and a significant decrease in volume over time. Using high-throughput SICM, we mapped the morphology of multiple single cells treated with TOY, which revealed that A549 showed characteristics of apoptosis and necrosis. The drug treatment does not significantly change the average root-mean-square (RMS) roughness of the cells. However, the drug leads to an increase in membrane height, possibly indicating early apoptotic changes. Plotting the individual RMS roughness of the cells showed a cell with an increase in roughness and the presence of pores, which is also an indication of necrosis behavior. Our results demonstrate that SICM is an effective technique for revealing the evolution of heterogeneity in single cells in their responses to anticancer drugs over time.
Collapse
Affiliation(s)
- Yusuf Muhammed
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Mia De Sabatino
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Robert A Lazenby
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
4
|
Chan BA, Youlden DR, Guan T, Lehman M, Windsor M, Bolton A, Dunn N, Cossio D, Philpot S, Sanmugarajah J. Setting the Benchmark: Patterns of Care and Outcomes for Early-stage Non-small Cell Lung Cancer in Queensland, Australia, 2011-2017. Asia Pac J Clin Oncol 2025. [PMID: 40308036 DOI: 10.1111/ajco.14177] [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] [Indexed: 05/02/2025]
Abstract
AIM Treatment paradigms for early-stage non-small cell lung cancer (NSCLC) are evolving rapidly. Our aim was to document baseline patterns of care and outcomes at the population level immediately prior to the introduction of immunotherapy. METHODS Data were obtained from the Queensland Oncology Repository. The study cohort comprised Queensland residents diagnosed with a non-metastatic primary NSCLC between 2011 and 2017, with follow-up on treatment and mortality to December 31, 2022. Poisson regression was used to determine patient and clinical characteristics associated with receiving different treatment modalities within 1 year of diagnosis. Variations in 5-year observed survival were assessed using flexible parametric modelling. RESULTS A total of 4445 people were included, of whom 30% were treated with surgery only, 15% with surgery plus chemotherapy and/or radiotherapy and 44% with chemotherapy and/or radiotherapy only. The remaining 10% did not receive any recorded treatment. People in outer regional/remote areas had lower rates of radiotherapy (relative likelihood [RL] = 0.87, 95% confidence interval [CI] 0.78-0.97) and chemotherapy (RL = 0.89, 95% CI 0.81-0.98) than those in major cities, but there were no significant differences by First Nations status or socio-economic status. Five-year observed survival varied from 63% (95% CI 60%-65%) for stage I to 41% (38%-45%) for stage II and 20% (18%-22%) for stage III. The treatment modality significantly affected survival irrespective of stage at diagnosis (all p < 0.001). CONCLUSION Monitoring treatment outcomes for early-stage NSCLC at the population level is crucial for optimizing patient care, resource allocation and informing consumer choice. Emerging approaches involving immunotherapy are expected to further improve outcomes.
Collapse
Affiliation(s)
- Bryan A Chan
- Centre for Bioinnovation, University of the Sunshine Coast, Sunshine Coast, Australia
- School of Medicine and Dentistry, Griffith University, Gold Coast, Australia
- The University of Queensland, Brisbane, Australia
- Sunshine Coast University Hospital, Queensland Health, Sunshine Coast, Australia
- Queensland Cancer Control Safety and Quality Partnership, Lung Cancer Sub-committee, Queensland Health, Brisbane, Australia
| | - Danny R Youlden
- Cancer Alliance Queensland, Metro South Health, Brisbane, Australia
| | - Tracey Guan
- Cancer Alliance Queensland, Metro South Health, Brisbane, Australia
| | - Margot Lehman
- The University of Queensland, Brisbane, Australia
- Queensland Cancer Control Safety and Quality Partnership, Lung Cancer Sub-committee, Queensland Health, Brisbane, Australia
- Princess Alexandra Hospital, Queensland Health, Brisbane, Australia
| | - Morgan Windsor
- Queensland Cancer Control Safety and Quality Partnership, Lung Cancer Sub-committee, Queensland Health, Brisbane, Australia
- The Prince Charles Hospital, Queensland Health, Brisbane, Australia
| | - Alison Bolton
- Queensland Cancer Control Safety and Quality Partnership, Lung Cancer Sub-committee, Queensland Health, Brisbane, Australia
| | - Nathan Dunn
- Cancer Alliance Queensland, Metro South Health, Brisbane, Australia
| | - Danica Cossio
- Cancer Alliance Queensland, Metro South Health, Brisbane, Australia
| | - Shoni Philpot
- Queensland Cancer Control Safety and Quality Partnership, Lung Cancer Sub-committee, Queensland Health, Brisbane, Australia
- Cancer Alliance Queensland, Metro South Health, Brisbane, Australia
| | - Jasotha Sanmugarajah
- School of Medicine and Dentistry, Griffith University, Gold Coast, Australia
- Queensland Cancer Control Safety and Quality Partnership, Lung Cancer Sub-committee, Queensland Health, Brisbane, Australia
- Gold Coast University Hospital, Queensland Health, Gold Coast, Australia
| |
Collapse
|
5
|
Varghese R, Ramamoorthy S. Deciphering the effects of bixin on pulmonary alveolar adenocarcinoma migration and proliferation via targeting BAX/BCL-2 and Cyclin D1. Sci Rep 2025; 15:15109. [PMID: 40301461 PMCID: PMC12041254 DOI: 10.1038/s41598-025-96788-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 03/29/2025] [Indexed: 05/01/2025] Open
Abstract
There is a tremendous upsurge in lung cancer incidences due to changing lifestyles and other environmental risk factors. Unfortunately, the use of clinical therapeutics is causing serious side effects and drug-resistant tumors. Taking account of the severity of lung cancer malignancy and the pressing need for natural therapeutics, we investigated the anticancer potential of bixin in A549, pulmonary alveolar adenocarcinoma cell lines meticulously for the first time. Bixin is an apocarotenoid present in the seed arils of Bixa orellana known for its remarkable coloring utilities and high medicinal value. Here, we identified the cytotoxic and anti-migratory nature of bixin through MTT and scratch assay. Bixin also induced characteristic apoptotic morphological changes in cells which were distinguished through 4',6-diamidino-2-phenylindole (DAPI), and Acridine orange/Ethidium bromide (AO/EB) labeling. Bixin induced the mitochondrion-associated intrinsic apoptosis in A549 cells as evidenced in mitochondrial membrane potential assay, apoptosis assay, cell cycle analysis, and caspase assays. The relative gene expression studies proved that the bixin upregulated BAX, and downregulated BCL-2 and Cyclin D1. The in-silico analyses, molecular docking and molecular dynamics simulation underlined the interaction features of bixin and targeted proteins.
Collapse
Affiliation(s)
- Ressin Varghese
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
6
|
Wu Q, Qin Z, Sun P, Liu F, Ge Y, Wang P. Determination of Antioxidant, Cytotoxicity, and Anti-human Lung Cancer Properties of Silver Nanoparticles Green-Formulated by Foeniculum vulgare Extract Combined with Radiotherapy. Biol Trace Elem Res 2025; 203:2139-2148. [PMID: 39107456 DOI: 10.1007/s12011-024-04332-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/29/2024] [Indexed: 03/20/2025]
Abstract
The current investigation involved the silver nanoparticles green synthesis utilizing the aqueous extract derived from the Foeniculum vulgare leaves (AgNPs@FV). The effectiveness of these newly developed nanoparticles in conjunction with radiotherapy was evaluated on lung cancer cells. The synthesized AgNPs@FV underwent characterization through various analytical techniques such as energy dispersive X-ray (EDX), field emission-scanning electron microscopy (FE-SEM), X-ray diffraction (XRD), and ultraviolet-visible (UV-Vis) spectrophotometry. The efficacy of AgNPs@FV in conjunction with radiotherapy against human lung cancer was assessed through the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. The AgNPs@FV exhibited a spherical morphology ranging in size from 10.16 to 42.74 nm. The EDX diagram of nanoparticles shows energy signals at 3.02 and 2.64 keV, which are attributed to Ag Lβ and Ag Lα, respectively. During the antioxidant evaluation, AgNPs@FV and butylated hydroxytoluene (BHT) displayed IC50 values of 166 and 59 µg/mL, respectively. The cells treated with AgNPs@FV in conjunction with radiotherapy were evaluated using the MTT assay over 48 h to determine cytotoxicity and anti-human lung cancer characteristics on normal (human umbilical vein endothelial cell (HUVEC)) and lung cancer cells and exhibited IC50 values of 211, 166, and 296 µg/mL against NCI-H2126, NCI-H1299, and NCI-H1437, respectively. Furthermore, the malignant lung cell viability decreased when treated with a combination of AgNPs@FV and radiotherapy. Based on the aforementioned findings, it is possible that the newly developed AgNPs@FV could serve as a novel chemotherapeutic medication or adjunct for addressing lung cancer following the completion of clinical trials involving human subjects.
Collapse
Affiliation(s)
- Qian Wu
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, No.216 Guanshan Road, Hongshan District, Wuhan, 430074, Hubei, China
| | - Zifu Qin
- Department of Health, Vertigo Examination Room, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan City, 250013, Shandong, China
| | - Pei Sun
- Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, No. 105, Jiefang Road, Jinan City, 250013, Shandong, China
| | - Fang Liu
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, No.216 Guanshan Road, Hongshan District, Wuhan, 430074, Hubei, China
| | - Yin Ge
- Physical Examination Center, Wuhan Third Hospital, Tongren Hospital of Wuhan University, No.216 Guanshan Road, Hongshan District, Wuhan, 430074, Hubei, China
| | - Pengbo Wang
- Department of Radiotherapy, Yantaishan Hospital, No.91, Jiefang Road, Zhifu District, Yantai, 264001, Shandong, China.
| |
Collapse
|
7
|
Abdalla BA, Ali RM, Kakamad FH, Ahmed HK, Ali RM, Abdullah AM, Yasseen HA, Fattah FH, Hama Rashid DJ, Karim SO, Mahmood YM, Hasan SJ, Hamasaeed AG. Role of dasatinib in the management of lung cancer: A meta-analysis of clinical trials. Biomed Rep 2025; 22:51. [PMID: 39926044 PMCID: PMC11803369 DOI: 10.3892/br.2025.1929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025] Open
Abstract
Lung cancer, particularly non-small cell lung cancer (NSCLC), the leading cause of cancer-related deaths worldwide, has prompted extensive research into innovative treatments, including targeted therapies. The present meta-analysis aims to evaluate the efficacy of dasatinib, both as monotherapy and in combination with other therapies, for the treatment of lung cancer. Adhering to the PRISMA guidelines, a meticulous, thorough review of clinical trials was conducted across reputable databases, including Google Scholar, PubMed/MEDLINE, and EMBASE, focusing on studies published in English up to May 5th, 2024. Inclusion criteria were restricted to randomized clinical trials (RCTs) assessing the efficacy of dasatinib, either as a standalone therapy or in combination with other treatments. The search identified 55 studies, of which nine RCTs met the inclusion criteria: four phase II, three phase I, and two phase I/II. A total of 234 patients participated, with 107 receiving dasatinib alone and 127 undergoing combination therapy. Histological analyses revealed that 79.1% of patients had non-small cell lung cancer, with adenocarcinoma being the predominant subtype (63.8%), followed by squamous cell carcinoma (22.1%). Treatment responses varied, with 52.4% of the patients in the dasatinib alone group experiencing progressive disease, while 38.3% achieved stable disease; by contrast, 29.6% of patients in the combination therapy showed progressive disease. Adverse events, including anemia and fatigue, were more prevalent with combination therapies. Dasatinib treatment shows potential for improving overall survival with fewer adverse events compared with combination therapies in patients with lung cancer; however, large-scale clinical trials are essential to confirm its efficacy as a standalone treatment.
Collapse
Affiliation(s)
- Berun A. Abdalla
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- Kscien Organization for Scientific Research (Middle East Office), Sulaymaniyah, Kurdistan 46001, Iraq
| | - Rebaz M. Ali
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- Department of Oncology, Hiwa Hospital, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Fahmi H. Kakamad
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- Kscien Organization for Scientific Research (Middle East Office), Sulaymaniyah, Kurdistan 46001, Iraq
- College of Medicine, University of Sulaimani, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Harem K. Ahmed
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Rawa M. Ali
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- Hospital for Treatment of Victims of Chemical Weapons, Halabja 46018, Iraq
| | - Ari M. Abdullah
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- Department of Pathology, Sulaymaniyah Teaching Hospital, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Hadeel A. Yasseen
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- College of Medicine, University of Sulaimani, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Fattah H. Fattah
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- College of Medicine, University of Sulaimani, Sulaymaniyah, Kurdistan 46001, Iraq
| | | | - Sanaa O. Karim
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- College of Nursing, University of Sulaimani, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Yousif M. Mahmood
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Sabah J. Hasan
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
| | - Ahmed G. Hamasaeed
- Scientific Affairs Department, Smart Health Tower, Sulaymaniyah, Kurdistan 46001, Iraq
- Faculty of Medical Sciences, School of Pharmacy, University of Sulaimani, Sulaymaniyah, Kurdistan 46001, Iraq
| |
Collapse
|
8
|
Smolarz B, Łukasiewicz H, Samulak D, Piekarska E, Kołaciński R, Romanowicz H. Lung Cancer-Epidemiology, Pathogenesis, Treatment and Molecular Aspect (Review of Literature). Int J Mol Sci 2025; 26:2049. [PMID: 40076671 PMCID: PMC11900952 DOI: 10.3390/ijms26052049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Lung cancer is one of the most common malignant cancers in most countries and is the leading cause of death among cancer diseases worldwide. Despite constant progress in diagnosis and therapy, survival rates of patients diagnosed with lung cancer remain unsatisfactory. Numerous epidemiological and experimental studies conducted as early as the 1970s confirm that the most important risk factor for the development of lung cancer is long-term smoking, which remains valid to this day. In the paper, the authors present the latest data on the epidemiology, pathogenesis, treatment and molecular aspects of this cancer. In the last decade, many molecular alterations that are effective in the development of lung cancer have been discovered. In adenocarcinoma, tyrosine kinase inhibitors were developed for EGFR mutations and ALK and ROS1 translocations and were approved for use in the treatment of advanced stage adenocarcinomas. In the case of squamous cell carcinoma, the evaluation of these mutations is not yet being used in clinical practice. In addition, there are ongoing studies concerning many potential therapeutic molecular targets, such as ROS, MET, FGFR, DDR-2 and RET. Constant progress in diagnostic and therapeutic methods gives rise to hopes for an improved prognosis in patients with lung cancer.
Collapse
Affiliation(s)
- Beata Smolarz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
| | - Honorata Łukasiewicz
- Faculty of Medicine and Health Sciences, Department of Nursing, The President Stanisław Wojciechowski Calisia University, 62-800 Kalisz, Poland;
| | - Dariusz Samulak
- Department of Obstetrics and Gynecology and Gynecological Oncology, Regional Hospital in Kalisz, 62-800 Kalisz, Poland;
- Department of Obstetrics, The President Stanisław Wojciechowski Calisia University, 62-800 Kalisz, Poland
| | - Ewa Piekarska
- Regional Hospital in Kalisz, 62-800 Kalisz, Poland; (E.P.); (R.K.)
| | | | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
| |
Collapse
|
9
|
Yang Q, Zhao D, Ju L, Cao P, Wei J, Liu Z. Brigatinib can inhibit proliferation and induce apoptosis of human immortalized keratinocyte cells. Front Pharmacol 2025; 16:1524277. [PMID: 40041486 PMCID: PMC11876137 DOI: 10.3389/fphar.2025.1524277] [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: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Background Brigatinib is approved in multiple countries for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC). Despite its superior efficacy, the dermal toxicities caused by brigatinib cannot be overlooked. However, its underlying mechanism remains unknown. Methods The effects of brigatinib on the proliferation ability of human immortalized keratinocyte (HaCaT) cells were evaluated using Cell Counting Kit-8 (CCK-8) proliferation, colony formation, and 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays. The effects of brigatinib on apoptosis were detected using Annexin FITC/PI and Acridine Orange (AO) staining assays. Cell cycle was assessed with flow cytometry. An analysis of transcriptome by RNA sequencing procedures (RNA-seq) was performed to reveal the key regulatory genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to find out the biological function and related signal pathways. The expressions of amphiregulin, epiregulin and transforming growth factor alpha (TGFA) and the protein levels of Phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) and Cleaved-Caspase three were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot assay. Results Brigatinib inhibits cell proliferation with an IC50 value of 2.9 μmol/L and significantly increases apoptosis rates. Transcriptome sequencing (RNA-seq) indicates that brigatinib could significantly downregulate the expression of amphiregulin, epiregulin and TGFA. In addition, we demonstrated that brigatinib reduced the protein expression of amphiregulin, epiregulin, TGFA, PI3K, AKT and phosphorylated AKT (p-AKT). Conclusion This study confirms the inhibition of HaCaT cells growth and progression by brigatinib and highlights the potential value of the PI3K/AKT pathway as a therapeutic target for brigatinib-induced dermal toxicities.
Collapse
Affiliation(s)
- Qi Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dan Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Linjie Ju
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jifu Wei
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixian Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
von Itzstein MS, Burns TF, Dowell JE, Horn L, Camidge DR, York SJ, Eaton KD, Kyle K, Fattah F, Liu J, Mu-Mosley H, Gupta A, Nadeem U, Gao A, Zhang S, Gerber DE. Phase I/II Trial of Exportin 1 Inhibitor Selinexor plus Docetaxel in Previously Treated, Advanced KRAS-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res 2025; 31:639-648. [PMID: 39651955 PMCID: PMC11832340 DOI: 10.1158/1078-0432.ccr-24-1722] [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: 07/30/2024] [Revised: 09/20/2024] [Accepted: 12/03/2024] [Indexed: 02/18/2025]
Abstract
PURPOSE Patients with Kirsten rat sarcoma viral oncogene (KRAS)-mutant non-small cell lung cancer (NSCLC) have limited therapeutic options. Based on the activity of nuclear export inhibition in preclinical models, we evaluated this strategy in previously treated, advanced KRAS-mutant NSCLC. PATIENTS AND METHODS The primary outcomes of this multicenter phase I/II dose-escalation trial of selinexor plus docetaxel were safety and tolerability. Selinexor was started 1 week before docetaxel to permit monotherapy pharmacodynamic assessment. RESULTS Among 40 enrolled patients, the median age was 66 years, 55% were female, and 85% were White. The MTD was selinexor 60 mg orally weekly plus docetaxel 75 mg/m2 every 3 weeks. The most common adverse events were nausea (73%, 8% grade ≥3), fatigue (70%, 5% grade ≥3), neutropenia (65%, 60% grade ≥3), and diarrhea (58%, 10% grade ≥3). Of 32 efficacy-evaluable patients, 7 (22%) had partial responses and 18 (56%) had stable disease. Outcomes were not associated with KRAS mutation type but were significantly better in cases with wild-type TP53 (42%), including response and disease control rates (27% and 80% vs. 9% and 27%, respectively; P = 0.03) and progression-free survival (median 7.4 vs. 1.8 months; HR, 0.2; 95% confidence interval, 0.07-0.67; P = 0.003). After selinexor initiation and prior to docetaxel administration, serum lactate dehydrogenase levels increased an average of 51 U/L in TP53-altered cases and decreased an average of 48 U/L in TP53 wild-type cases (P = 0.06). CONCLUSIONS Selinexor plus docetaxel was relatively well tolerated in patients with advanced KRAS-mutant NSCLC. The regimen has promising efficacy in TP53 wild-type cases, in which selinexor monotherapy may also have activity.
Collapse
Affiliation(s)
- Mitchell S. von Itzstein
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Timothy F. Burns
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jonathan E. Dowell
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Leora Horn
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | | | - Sally J. York
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Keith D. Eaton
- Fred Hutchinson Cancer Center, University of Washington, Seattle, Washington, USA
| | - Kelly Kyle
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Farjana Fattah
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jialing Liu
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hong Mu-Mosley
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Arjun Gupta
- Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Urooba Nadeem
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ang Gao
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Song Zhang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David E. Gerber
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
11
|
Leite da Silva LF, Saldanha EF, de Menezes JSA, Halamy Pereira L, de Bragança dos Santos JAR, Buonopane IR, de Souza EM, de Menezes CUG, Lopes G. Plasma ctDNA kinetics as a predictor of systemic therapy response for advanced non-small cell lung cancer: a systematic review and meta-analysis. Oncologist 2025; 30:oyae344. [PMID: 39998904 PMCID: PMC11853598 DOI: 10.1093/oncolo/oyae344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/07/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Predicting early treatment response in advanced non-small cell lung cancer (NSCLC) is challenging. Longitudinal monitoring of circulating tumor DNA (ctDNA) can track tumor response to treatments like immune checkpoint blockade (ICB) and correlate with outcomes. This meta-analysis evaluated whether ctDNA clearance or decrease is associated with improved survival across various settings in NSCLC. METHODS A systematic review of MEDLINE, EMBASE, and Cochrane databases (up to April 2024) identified studies evaluating the impact of ctDNA kinetics on survival outcomes in non-curative NSCLC settings. Pooled hazard ratios (HR) for progression-free survival (PFS) and overall survival (OS) were calculated using a random effects model. RESULTS We included 32 studies with 3047 NSCLC patients receiving systemic therapies such as targeted therapy (TT), ICB, and chemotherapy. Meta-analysis of 31 studies showed that ctDNA decrease/clearance was linked to improved PFS (HR: 0.32 [0.26, 0.40], I² = 63%, P < .01). Subgroup analysis indicated strong PFS benefits from ctDNA clearance (HR: 0.27 [0.20, 0.36]). Similar improvements were seen across patients undergoing targeted therapy (HR: 0.34) and ICB (HR: 0.33). Analysis of 25 studies revealed a significant association between ctDNA reduction and better OS (HR: 0.31 [0.23, 0.42], I² = 47%, P < .01). Subgroup findings were consistent for both TT (HR: 0.41) and ICB (HR: 0.32). Sensitivity analysis demonstrated that ctDNA clearance/decrease was consistently associated with improved PFS across study designs and ctDNA analysis methods. There was no significant variation in hazard ratios for PFS based on NSCLC subtypes, smoking status, or sex. CONCLUSION Plasma ctDNA kinetics was associated with improved survival outcomes in patients diagnosed with advanced NSCLC undergoing treatment with TT and ICB.
Collapse
Affiliation(s)
- Luís F Leite da Silva
- Departmento de Ciências Médicas, Universidade Federal Fluminense, Niterói, RJ 24033-900, Brazil
| | - Erick F Saldanha
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, ON M5G 2M9, Canada
| | | | - Leonardo Halamy Pereira
- Departmento de Ciências Médicas, Universidade Federal Fluminense, Niterói, RJ 24033-900, Brazil
| | | | | | - Erito M de Souza
- Departmento de Ciências Médicas, Universidade Federal Fluminense, Niterói, RJ 24033-900, Brazil
| | | | - Gilberto Lopes
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, United States
| |
Collapse
|
12
|
Zhao H, Huang S, Wu J, Lu Y, Zou Y, Zeng H, Li C, Wang J, Zhang X, Duan S, Liang W. Efficacy and safety of first-line PD-1/PD-L1 inhibitor in combination with CTLA-4 inhibitor in the treatment of patients with advanced non-small cell lung cancer: a systemic review and meta-analysis. Front Immunol 2025; 16:1515027. [PMID: 39981238 PMCID: PMC11839650 DOI: 10.3389/fimmu.2025.1515027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction The combination of PD-1/PD-L1 inhibitor with CTLA-4 inhibitor for advanced non-small cell lung cancer(NSCLC) is presently a significant area of research, however its clinical application remains contentious. This meta-analysis aimed to assess the efficacy and safety of first-line PD-1/PD-L1 inhibitor in combination with CTLA-4 inhibitor (CP) in the treatment of patients with advanced NSCLC. Methods A systemic search was conducted in four databases (PubMed, Cochrane library, Embase, and Web of Science) from their establishment until January 17, 2024, for randomized controlled trials that investigated the use of the first-line PD-1/PD-L1 inhibitor plus CTLA-4 inhibitor in patients with advanced NSCLC. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), and adverse events (AEs) were subjected to meta-analyses. Results Totally 7 eligible randomized controlled trials including 4682 people were included. Two comparative analyses were performed: CP versus chemotherapy, CP versus PD-1/PD-L1 inhibitor (P). Compared with the chemotherapy group, CP improved OS (HR: 0.84, 95% CI: 0.75-0.94, p<0.05) but not PFS (HR: 0.94, 95%CI: 0.73-1.20, p = 0.63) or ORR (OR: 1.16, 95% CI: 0.79-1.71, p = 0.45). In terms of toxicity, CP had slightly fewer any AEs compared to chemotherapy (RR: 0.94, 95% CI: 0.91-0.97; p<0.05). Compared to the P group, there was no significant difference in OS (MD: -0,25, 95% CI: -2.47-1.98, p = 0.83), PFS (MD: -0.91, 95% CI: -3.19-1.36, p = 0.43), and ORR (OR:1.05, 95% CI. 0.80-1.36, p = 0.73). Subgroup analysis revealed that CP provided superior OS compared with P in patients with PD-L1 expression < 1%. Conclusion CP was a feasible and safe first-line therapy for patients with advanced NSCLC. Specifically, CP may function as a therapeutic alternative for individuals with low or negative PD-L1 expression, resulting in enhanced long-term outcomes compared to chemotherapy or P. Further randomized controlled trials with prolonged follow-up periods are necessary to validate these results, particularly focusing on efficacy in patients with differing PD-L1 expression levels, to improve the stratified implementation of immunotherapy. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024621116, identifier CRD42024621116.
Collapse
Affiliation(s)
- Huimin Zhao
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Shanshan Huang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Jianyu Wu
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Yanlan Lu
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Yue Zou
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Haijian Zeng
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Chunlan Li
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Jin Wang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Xiaochen Zhang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
- Medicine College, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Siliang Duan
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
- Medicine College, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Weiming Liang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| |
Collapse
|
13
|
Yang Y, Yang H, Gao Y, Yang Q, Zhu X, Miao Q, Xu X, Li Z, Zuo D. EML4-ALK G1202R and EML4-ALK L1196M mutations induce crizotinib resistance in non-small cell lung cancer cells through activating epithelial-mesenchymal transition mediated by MDM2/MEK/ERK signal axis. Cell Biol Int 2025; 49:55-67. [PMID: 39318039 DOI: 10.1002/cbin.12249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 08/04/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Crizotinib, as the first-generation of anaplastic lymphoma kinase (ALK) inhibitor, effectively improves the survival time of ALK-positive non-small cell lung cancer (NSCLC) patients. However, its efficacy is severely limited by drug resistance caused by secondary mutations. G1202R and L1196M are classical mutation sites located in ALK kinase domain. They may hinder the binding of ALK inhibitors to the target kinase domain, resulting in drug resistance in patients. However, the exact mechanism of drug resistance mediated by these mutations remains unclear. In this study, we aimed to evaluate how G1202R and L1196M mutations mediate crizotinib resistance. To explore the resistance mechanism, we constructed EML4-ALK G1202R and L1196M mutant cell lines with A549 cells. The results showed that the mutant cells exhibited significant epithelial-mesenchymal transition (EMT) and metastasis compared to control (A549-vector) or wild type (A549-EML4-ALK) cells. Subsequently, it was found that the occurrence of EMT was correlated to the high expression of murine double minute 2 (MDM2) protein and the activation of mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway in mutant cells. Down-regulation of MDM2 inhibited the activation of MEK/ERK pathway, thus reversed the EMT process and markedly increased the inhibitory effect of crizotinib on the growth of mutant cells. Collectively, resistance of ALK-positive NSCLC cells to crizotinib is induced by G1202R and L1196M mutations through activation of the MDM2/MEK/ERK signalling axis, promoting EMT process and metastasis. These findings suggest that the combination of MDM2 inhibitors and crizotinib could be a potential therapeutic strategy.
Collapse
MESH Headings
- Crizotinib/pharmacology
- Humans
- Epithelial-Mesenchymal Transition/drug effects
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Proto-Oncogene Proteins c-mdm2/metabolism
- Proto-Oncogene Proteins c-mdm2/genetics
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/genetics
- MAP Kinase Signaling System/drug effects
- Mutation
- Animals
- A549 Cells
- Cell Line, Tumor
- Mice, Nude
- Mice
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Yuying Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Huan Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yunhui Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Qian Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinya Zhu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Qianying Miao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaobo Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
14
|
Geng H, Xue Y, Yan B, Lu Z, Yang H, Li P, Zhou J. Network Pharmacology and Molecular Docking Study on the Mechanism of the Therapeutic Effect of Strychni Semen in NSCLC. Biol Proced Online 2024; 26:33. [PMID: 39736533 DOI: 10.1186/s12575-024-00259-w] [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: 08/23/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
Strychni Semen, characterized by its bitter taste and warm properties, has been confirmed to possess anti-tumor properties. However, the molecular mechanism of Strychni Semen in treating non-small cell lung cancer (NSCLC) needs further study. This study aimed to explore the molecular mechanism of Strychni Semen in treating NSCLC based on network pharmacology and molecular docking. The active components and targets of Strychni Semen were retrieved from the TCMSP, supplemented by the HERB database and the related literature. NSCLC-related targets were retrieved from the GeneCards, OMIM and DisGenet databases. The intersection targets of Strychni Semen in treating NSCLC were obtained via an online platform. The Protein-Protein Interaction (PPI) network was subsequently constructed to deeply analyse the interrelationship of the intersection targets via the String database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were carried out via the Metascape database. The interactive networks between Strychni Semen and NSCLC were constructed via Cytoscape 3.9.1. Molecular docking detected interactions between the key components and the core targets. The core targets were validated via GEO datasets. 21 active components and 67 targets were identified, with 47 associated with NSCLC. The key active components were Stigmasterol, IcarideA, 2-Hydroxymethylanthraquinone, (+)-catechin, (2R)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one, (S)-Stylopine, Brucine and Isobrucine. The core targets were PTGS2, NR3C1, ESR1, CASP3 and PRKACA. Molecular docking revealed that these compounds undergo strong binding affinity with the core genes. GEO database indicated that PTGS2 was the most promising core target. In addition, Strychni Semen's effects on NSCLC involved mainly the Calcium pathway, the Estrogen pathway, and the cGMP-PKG and cAMP pathways. This study visually demonstrated the mechanism of the therapeutic effect of Strychni Semen in NSCLC through multiple components, targets and pathways which provides a basis for clinical treatment and further experimental research.
Collapse
Affiliation(s)
- He Geng
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Yujie Xue
- Department of Pathology, Affiliated Huaian NO. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Binghua Yan
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Zhaoxue Lu
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Hengjin Yang
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Peng Li
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China.
| | - Jundong Zhou
- Department of Radiation Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, China.
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, China.
| |
Collapse
|
15
|
Shultes PV, Tadele DS, Durmaz A, Weaver DT, Barker-Clarke R, Dinh MN, Liu S, Alemu EA, Rayner S, Scott JG. Cell-Cell Fusion in NSCLC Confers a Fitness Benefit Under Drug Selection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626399. [PMID: 39677818 PMCID: PMC11642803 DOI: 10.1101/2024.12.02.626399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Cell-cell fusion has been implicated in various physiological and pathological processes, including cancer progression. This study investigated the role of cell-cell fusion in non-small cell lung cancer (NSCLC), focusing on its contribution to chemoresistance and tumor evolution. By co-culturing drug-sensitive and drug-resistant NSCLC cell lines, we observed spontaneous cell-cell fusion events, particularly under gefitinib selection. These fused cells exhibited enhanced fitness and a higher degree of chemoresistance compared to parental lines across a panel of 12 chemotherapeutic agents. Further analysis, including fluorescence imaging and cell cycle analysis, confirmed nuclear fusion and increased DNA content in the fused cells. Bulk RNA sequencing revealed genomic heterogeneity in fused cells, including enrichment of gene sets associated with cell cycle progression and epithelial-mesenchymal transition, hallmarks of cancer malignancy. These findings demonstrate that cell-cell fusion contributes significantly to therapeutic resistance and the promotion of aggressive phenotypes in NSCLC, highlighting its potential as a therapeutic target.
Collapse
|
16
|
Ibrahim S, Shenoy S, Kateel R, Hegde S, Parida A, Samantaray L. Navigating the complexity of BRAF mutations in non-small cell lung cancer: current insights and future prospects. Multidiscip Respir Med 2024; 19:992. [PMID: 39545749 PMCID: PMC11614001 DOI: 10.5826/mrm.2024.992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 11/17/2024] Open
Abstract
There are many challenges that are faced in the treatment of Non-Small Cell Lung Cancer (NSCLC) due to the complexities associated with the tumor. Association of different types of mutations are one of the major complexities. Among these mutations, BRAF mutations are significantly gathering more attention due to their impact on disease progression and therapeutic response. This review provides an analysis of the current understanding of BRAF mutations in NSCLC, focusing on the molecular intricacies, clinical implications, and therapeutic advancements. The article explores the diverse spectrum of BRAF mutations, highlighting the prevalence of specific mutations such as V600E and non-V600E alterations. The review also highlights the intricate signalling pathways influenced by BRAF mutations, shedding light on their role in tumorigenesis and metastasis. Therapeutically, we critically evaluate the existing targeted therapies tailored for BRAF-mutant NSCLC, addressing their efficacy, limitations, and emerging resistance mechanisms. Furthermore, we outline ongoing clinical trials and promising investigational agents that hold potential for reshaping the treatment of NSCLC. This review provides comprehensive current information about the role of BRAF mutations in NSCLC. Understanding the molecular diversity, clinical implications, and therapeutic strategies associated with BRAF-mutant NSCLC is crucial for optimizing patient outcomes and steering the direction of future research in this evolving field.
Collapse
Affiliation(s)
- Sufyan Ibrahim
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma Center, Oklahoma City, OK, USA
| | - Smita Shenoy
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ramya Kateel
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shreya Hegde
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Amrita Parida
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | |
Collapse
|
17
|
Lu Z, Hou G. Characterization of the function and clinical value of ERCC family genes in lung adenocarcinoma. Front Oncol 2024; 14:1476100. [PMID: 39582530 PMCID: PMC11581973 DOI: 10.3389/fonc.2024.1476100] [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: 08/05/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction ERCC genes, responsible for encoding enzymes involved in base excision repair, have been implicated in various cancers, contributing to chemoresistance. However, a comprehensive analysis of the prognostic and therapeutic significance of this gene family in lung adenocarcinoma (LUAD) is lacking. Methods This study conducted a multidimensional assessment of ERCC family genes in LUAD using bioinformatic approaches, including mRNA expression level, gene methylation, and copy number variation (CNV), as well as their correlations with clinical outcome, gene set variations, and tumor-infiltrating lymphocytes (TILs). In addition, We evaluated the anti-tumor effects of ERCC8 in cell lines, demonstrating its clinical potential on an experimental level. Results Overall, the expression of ERCC genes exhibited a negative correlation with good prognosis, with ERCC6L and ERCC8 demonstrating the most reliable predictive performance. Gene methylation level and CNV increases of ERCC genes generally displayed negative and positive associations with their expression levels, respectively. Additionally, GSVA analysis suggested that ERCC expression was positively correlated with cell cycle and apoptosis pathways but negatively correlated to the TSC/mTOR pathway. Furthermore, the expression of ERCC genes exhibited a complex relationship with TILs and the response to anti-tumor drugs. The results of in vitro cellular experiments show that inhibiting ERCC8 can alleviate the malignant phenotype of LUAD cells. Discussion Our study revealed the multifaceted biological and clinical significance of ERCC family members in LUAD. These findings provide new insights into the function of ERCC family genes in LUAD and their potential clinical applications.
Collapse
Affiliation(s)
- Zhimin Lu
- Department of Outpatient, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| | - Guoxin Hou
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| |
Collapse
|
18
|
Chen Q, Zheng X, Cheng W, Li J. Landscape of targeted therapies for lung squamous cell carcinoma. Front Oncol 2024; 14:1467898. [PMID: 39544292 PMCID: PMC11560903 DOI: 10.3389/fonc.2024.1467898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Lung cancer, a common type of malignant neoplasm, has seen significant advancements in the treatment of lung adenocarcinoma (LUAD). However, the management of lung squamous cell carcinoma (LSCC) continues to pose challenges. Traditional treatment methods for LSCC encompass surgical resection, chemotherapy, and radiotherapy. The introduction of targeted therapy and immunotherapy has greatly benefited LSCC patients, but issues such as limited immune response rates and adverse reactions persist. Therefore, gaining a deeper comprehension of the underlying mechanisms holds immense importance. This review provides an in-depth overview of classical signaling pathways and therapeutic targets, including the PI3K signaling pathway, CDK4/6 pathway, FGFR1 pathway and EGFR pathway. Additionally, we delve into alternative signaling pathways and potential targets that could offer new therapeutic avenues for LSCC. Lastly, we summarize the latest advancements in targeted therapy combined with immune checkpoint blockade (ICB) therapy for LSCC and discuss the prospects and challenges in this field.
Collapse
Affiliation(s)
- Qiuxuan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoshuo Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiting Cheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jian Li
- Institude of Experimental Immunology, University Clinic of Rheinische Friedrich-Wihelms-University, Bonn, Germany
| |
Collapse
|
19
|
Al-Shemri MI, Aliannezhadi M, Ghaleb RA, Al-Awady MJ. Au-H 2Ti 3O 7 nanotubes for non-invasive anticancer treatment by simultaneous photothermal and photodynamic therapy. Sci Rep 2024; 14:25998. [PMID: 39472598 PMCID: PMC11522396 DOI: 10.1038/s41598-024-75862-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
Treating lung and prostate cancer cells is a major health problem that may be solved through the interactions of laser beams with nanoparticles. In the paper, Au-H2Ti3O7 nanotubes (NTs) are proposed as a treatment agent and the interactions of different laser beams with the nanostructure are considered to solve the mentioned health problem. Also, the NTs are employed to treat the cancers in dark conditions. The results are motivating because Au-H2Ti3O7 NPs do not affect healthy cells, while they strongly affect cancer cells, and the viability percentage of LNCap cells reaches 16% for incubation times of 48 h. Furthermore, treating LNCap cells using the irradiated Au-H2Ti3O7 NTs by NIR beam at 808 nm has no cytotoxicity, while cytotoxicity of 92% is obtained using an irradiation laser beam at 532 nm. Also, applying the laser beam at 635 nm to the NTs leads to a cytotoxicity of ∼53% in lung cancer (A549 cells). In total, the Au-H2Ti3O7 NTs have a selective effect on cancer cells and greatly reduce the viability in the given dark and irradiation conditions, leading to the introduction of them as a promising agent for the non-invasive treatment of prostate cancer and a moderate candidate for lung cancer therapy.
Collapse
Affiliation(s)
- Maher I Al-Shemri
- Faculty of Physics, Semnan University, PO Box: 35195-363, Semnan, Iran
| | | | - Rana A Ghaleb
- Department of Anatomy and Histology, College of Medicine, University of Babylon, Babylon, Iraq
| | - Mohammed J Al-Awady
- Department of Medical Biotechnology, College of Biotechnology, Al Qasim Green University, Babylon, 51013, Iraq
| |
Collapse
|
20
|
Ghosh DD, McDonald H, Dutta R, Krishnan K, Thilakan J, Paul MK, Arya N, Rao M, Rangnekar VM. Prognostic Indicators for Precision Treatment of Non-Small Cell Lung Carcinoma. Cells 2024; 13:1785. [PMID: 39513892 PMCID: PMC11545304 DOI: 10.3390/cells13211785] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) has established predictive biomarkers that enable decisions on treatment regimens for many patients. However, resistance to therapy is widespread. It is therefore essential to have a panel of molecular biomarkers that may help overcome therapy resistance and prevent adverse effects of treatment. We performed in silico analysis of NSCLC prognostic indicators, separately for adenocarcinomas and squamous carcinomas, by using The Cancer Genome Atlas (TCGA) and non-TCGA data sources in cBioPortal as well as UALCAN. This review describes lung cancer biology, elaborating on the key genetic alterations and specific genes responsible for resistance to conventional treatments. Importantly, we examined the mechanisms associated with resistance to immune checkpoint inhibitors. Our analysis indicated that a robust prognostic biomarker was lacking for NSCLC, especially for squamous cell carcinomas. In this work, our screening uncovered previously unidentified prognostic gene expression indicators, namely, MYO1E, FAM83 homologs, and DKK1 for adenocarcinoma, and FGA and TRIB1 for squamous cell carcinoma. It was further observed that overexpression of these genes was associated with poor prognosis. Additionally, FAM83 homolog and TRIB1 unexpectedly harbored copy number amplifications. In conclusion, this study elucidated novel prognostic indicators for NSCLC that may serve as targets to overcome therapy resistance toward improved patient outcomes.
Collapse
Affiliation(s)
- Damayanti Das Ghosh
- Basic and Translational Research Division, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata 700063, West Bengal, India; (D.D.G.); (R.D.)
- School of Health Sciences and Translational Research, Sister Nivedita University, Newtown, Kolkata 700156, West Bengal, India
| | - Hannah McDonald
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA;
| | - Rajeswari Dutta
- Basic and Translational Research Division, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata 700063, West Bengal, India; (D.D.G.); (R.D.)
| | - Keerthana Krishnan
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Jaya Thilakan
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India;
- Department of Genetics, UTD, Barkatullah University Bhopal, Bhopal 462026, Madhya Pradesh, India
| | - Manash K. Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Neha Arya
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India;
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Vivek M. Rangnekar
- Markey Cancer Center and Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
21
|
Tomassi S, Natale B, Roggia M, Amato L, De Rosa C, Della Corte CM, Baglini E, Amendola G, Messere A, Di Maro S, Barresi E, Da Settimo F, Trincavelli ML, Ciardiello F, Taliani S, Morgillo F, Cosconati S. Discovery of N-substituted-2-oxoindolin benzoylhydrazines as c-MET/SMO modulators in EGFRi-resistant non-small cell lung cancer. RSC Med Chem 2024; 16:d4md00553h. [PMID: 39512947 PMCID: PMC11539002 DOI: 10.1039/d4md00553h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024] Open
Abstract
Non-small cell lung cancer (NSCLC), the leading cause of cancer-related mortality worldwide, poses a formidable challenge due to its heterogeneity and the emergence of resistance to targeted therapies. While initially effective, first- and third-generation EGFR-tyrosine kinase inhibitors (TKIs) often fail to control disease progression, leaving patients with limited treatment options. To address this unmet medical need, we explored the therapeutic potential of multitargeting agents that simultaneously inhibit two key signalling pathways, the mesenchymal-epithelial transition factor (c-MET) and the G protein-coupled receptor Smoothened (SMO), frequently dysregulated in NSCLC. By employing a combination of in silico drug repurposing and structure-based structure-activity relationship (SAR) studies, we identified and developed novel c-MET/SMO-targeting agents with antiproliferative activity against first- as well as third-generation EGFR-TKI-resistant NSCLC cells suggesting a synergistic effect arising from the simultaneous inhibition of c-MET and SMO.
Collapse
Affiliation(s)
- Stefano Tomassi
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
- Department of Life Science, Health, and Health Professions, LINK Campus University Via del Casale di San Pio V, 44 00165 Rome Italy
| | - Benito Natale
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Michele Roggia
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Luisa Amato
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Caterina De Rosa
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Carminia Maria Della Corte
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Emma Baglini
- CNR IFC, Institute of Clinical Physiology, National Research Council of Italy CNR Research Area, Via G. Moruzzi 1 Pisa 56124 Italy
| | - Giorgio Amendola
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Anna Messere
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Salvatore Di Maro
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | | | | | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Floriana Morgillo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Via Pansini, 5 80138 Naples Italy
| | - Sandro Cosconati
- DiSTABiF, University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| |
Collapse
|
22
|
Wei M, Shen X, Liu Y, Chen X, Su S, Lv X, Qian X, Yu L, Wang L. The antitumor effect of diisopropylamine dichloroacetate on non-small cell lung cancer and its influence on the tumor immune microenvironment. Front Oncol 2024; 14:1447828. [PMID: 39267851 PMCID: PMC11390397 DOI: 10.3389/fonc.2024.1447828] [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: 06/12/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Objective To evaluate the antitumor effects of diisopropylamine dichloroacetate (DADA) alone or in combination with chemotherapy/radiotherapy/immunotherapy in NSCLC and explore the underlying mechanisms involved. Methods MTT, UV spectrophotometry, flow cytometry, fluorescence microscopy, and clonogenic survival assays were used. In LLC mouse models, the antitumor effects of radiotherapy, DADA, and the anti-PD-1 antibody alone or in combination were evaluated, and the T cell numbers were evaluated in different groups. Results DADA significantly inhibited lactate production and promoted apoptosis in NSCLC in vitro. Compared with pemetrexed or DADA alone, the combination of DADA with pemetrexed significantly inhibited proliferation and promoted apoptosis (p<0.05). This may be related to the decrease in the mitochondrial membrane potential in the combined group. Moreover, compared with radiotherapy alone, the combination of DADA with radiotherapy induced remarkable DNA damage. In vivo, the combination of radiotherapy, an anti-PD-1 antibody and DADA resulted in superior tumor inhibition than the combination of radiotherapy and anti-PD-1 antibody did (p < 0.05). The underlying mechanism may be partially related to the increased number of CD3+ T cells in the triplet combination group (p < 0.05). Discussion Our results showed that DADA has strong antitumor effects on NSCLC, either alone or in combination with chemotherapy or radiotherapy. Interestingly, the combination of radiotherapy, anti-PD-1 antibody and DADA had a more pronounced tumor-suppressing effect, which may be related to DADA-induced T-cell generation by reducing local lactic acid production in the tumor microenvironment. This study lays the foundation for further exploration of DADA in lung cancer, especially in the era of immunotherapy, on the basis of its possible immunomodulatory effects.
Collapse
Affiliation(s)
- Min Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyan Shen
- Department of Microbiology, Immunology and Pathology, Changzhou Hygiene Vocational Technology College, Changzhou, China
| | - Ye Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaotong Chen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Shu Su
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xin Lv
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xiaoping Qian
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lifeng Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Zhu M, Wang B, Zhang X, Zhou K, Miao Z, Sun J. [Assessment of baseline CCL19 + dendritic cell infiltration for predicting responses to immunotherapy in lung adenocarcinoma patients]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1529-1536. [PMID: 39276048 PMCID: PMC11378044 DOI: 10.12122/j.issn.1673-4254.2024.08.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Indexed: 09/16/2024]
Abstract
OBJECTIVE To explore the correlation of baseline CCL19+ dendritic cell (CCL19+ DC) infiltration in lung adenocarcinoma microenvironment with immunotherapy efficacy and CD8+ T cell infiltration. METHODS We retrospectively analyzed the data of patients with lung adenocarcinoma hospitalized at First Affiliated Hospital of Henan University of Science and Technology from January, 2020 to December, 2023, and collected tissue samples from 96 patients undergoing immunotherapy for assessing CCL19+ DC and CD8+ T cell infiltration using immunofluorescence assay. We evaluated the predictive value of baseline CCL19+ DCs for patient responses to immunotherapy using receiver-operating characteristics (ROC) curves and analyzed the correlations of baseline CCL19+ DC expression with immunotherapy efficacy and CD8+ T cell and cytotoxic T lymphocyte (CTL) infiltrations. In co-culture systems of lung adenocarcinoma PC9 cells, CD8+ T cells and DCs (overexpressing CCL19 with or without anti PD-1 antibody treatment), the expressions of granzyme B, perforin, IFN-γ, and Ki-67 in T cells were analyzed using flow cytometry. RESULTS The patients with partial or complete remission following immunotherapy had a significantly higher baseline CCL19+ DC infiltration level in lung adenocarcinoma tissues than those with poor responses. CCL19+ DC infiltration had an area under ROC curve of 0.785, a sensitivity of 75.6%, and a specificity of 62.8% for predicting immunotherapy efficacy. The expression of CD8+ T cell surface molecules Granzyme B (P<0.01), Perforin (P<0.01), IFN-γ (P<0.01) and Ki-67 (P<0.001) in patients with high expression of CCL19+ DC were higher than those in patients with low expression of CCL19+ DC. The baseline CCL19+ DC infiltration level was positively correlated with immunotherapy efficacy (P=0.003), CTL infiltration of (r=0.6657, P<0.001) and CD8+ T cell infiltration (P=0.007). In the co-cultured cells, CCL19 overexpression combined with anti-PD1 treatment of the DCs more strongly enhanced cytotoxicity and proliferation of CD8+ T lymphocytes than either of the single treatments (P<0.01 or 0.001). CONCLUSION The baseline CCL19+ DC infiltration level in lung adenocarcinoma microenvironment is positively correlated with immunotherapy efficacy and CTL infiltration and can thus predict the response to immunotherapy.
Collapse
Affiliation(s)
- M Zhu
- Clinical Medical College, Henan University of Science and Technology,Luoyang 471003, China
| | - B Wang
- Clinical Medical College, Henan University of Science and Technology,Luoyang 471003, China
| | - X Zhang
- Clinical Medical College, Henan University of Science and Technology,Luoyang 471003, China
| | - K Zhou
- Clinical Medical College, Henan University of Science and Technology,Luoyang 471003, China
| | - Z Miao
- Clinical Medical College, Henan University of Science and Technology,Luoyang 471003, China
| | - J Sun
- Cancer Hospital of First Affiliated Hospital, Henan University of Science and Technology,Luoyang 471003, China
- Cancer Institute, Henan University of Science and Technology, Luoyang 471003, China
- Henan Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
| |
Collapse
|
24
|
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
|
25
|
Mendes C, Lemos I, Hipólito A, Abreu B, Freitas-Dias C, Martins F, Pires R, Barros H, Bonifácio V, Gonçalves L, Serpa J. Metabolic profiling and combined therapeutic strategies unveil the cytotoxic potential of selenium-chrysin (SeChry) in NSCLC cells. Biosci Rep 2024; 44:BSR20240752. [PMID: 38990147 PMCID: PMC11292474 DOI: 10.1042/bsr20240752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/12/2024] Open
Abstract
Lung cancer ranks as the predominant cause of cancer-related mortalities on a global scale. Despite progress in therapeutic interventions, encompassing surgical procedures, radiation, chemotherapy, targeted therapies and immunotherapy, the overall prognosis remains unfavorable. Imbalances in redox equilibrium and disrupted redox signaling, common traits in tumors, play crucial roles in malignant progression and treatment resistance. Cancer cells, often characterized by persistent high levels of reactive oxygen species (ROS) resulting from genetic, metabolic, and microenvironmental alterations, counterbalance this by enhancing their antioxidant capacity. Cysteine availability emerges as a critical factor in chemoresistance, shaping the survival dynamics of non-small cell lung cancer (NSCLC) cells. Selenium-chrysin (SeChry) was disclosed as a modulator of cysteine intracellular availability. This study comprehensively characterizes the metabolism of SeChry and investigates its cytotoxic effects in NSCLC. SeChry treatment induces notable metabolic shifts, particularly in selenocompound metabolism, impacting crucial pathways such as glycolysis, gluconeogenesis, the tricarboxylic acid (TCA) cycle, and amino acid metabolism. Additionally, SeChry affects the levels of key metabolites such as acetate, lactate, glucose, and amino acids, contributing to disruptions in redox homeostasis and cellular biosynthesis. The combination of SeChry with other treatments, such as glycolysis inhibition and chemotherapy, results in greater efficacy. Furthermore, by exploiting NSCLC's capacity to consume lactate, the use of lactic acid-conjugated dendrimer nanoparticles for SeChry delivery is investigated, showing specificity to cancer cells expressing monocarboxylate transporters.
Collapse
Affiliation(s)
- Cindy Mendes
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| | - Isabel Lemos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| | - Ana Hipólito
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| | - Bruna Abreu
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| | - Catarina Freitas-Dias
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| | - Filipa Martins
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| | - Rita F. Pires
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Hélio Barros
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Vasco D.B. Bonifácio
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Luís G. Gonçalves
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Jacinta Serpa
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto 1099-023, Lisboa, Portugal
| |
Collapse
|
26
|
Bauso LV, La Fauci V, Munaò S, Bonfiglio D, Armeli A, Maimone N, Longo C, Calabrese G. Biological Activity of Natural and Synthetic Peptides as Anticancer Agents. Int J Mol Sci 2024; 25:7264. [PMID: 39000371 PMCID: PMC11242495 DOI: 10.3390/ijms25137264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the leading causes of morbidity and death worldwide, making it a serious global health concern. Chemotherapy, radiotherapy, and surgical treatment are the most used conventional therapeutic approaches, although they show several side effects that limit their effectiveness. For these reasons, the discovery of new effective alternative therapies still represents an enormous challenge for the treatment of tumour diseases. Recently, anticancer peptides (ACPs) have gained attention for cancer diagnosis and treatment. ACPs are small bioactive molecules which selectively induce cancer cell death through a variety of mechanisms such as apoptosis, membrane disruption, DNA damage, immunomodulation, as well as inhibition of angiogenesis, cell survival, and proliferation pathways. ACPs can also be employed for the targeted delivery of drugs into cancer cells. With over 1000 clinical trials using ACPs, their potential for application in cancer therapy seems promising. Peptides can also be utilized in conjunction with imaging agents and molecular imaging methods, such as MRI, PET, CT, and NIR, improving the detection and the classification of cancer, and monitoring the treatment response. In this review we will provide an overview of the biological activity of some natural and synthetic peptides for the treatment of the most common and malignant tumours affecting people around the world.
Collapse
Affiliation(s)
- Luana Vittoria Bauso
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Valeria La Fauci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Serena Munaò
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Desirèe Bonfiglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Alessandra Armeli
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Noemi Maimone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Clelia Longo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98168 Messina, Italy
| |
Collapse
|
27
|
Kim M. Mitochondria of T Lymphocytes Promote Anti-Pulmonary Tumor Immune Response. World J Oncol 2024; 15:472-481. [PMID: 38751696 PMCID: PMC11092414 DOI: 10.14740/wjon1841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/30/2024] [Indexed: 05/18/2024] Open
Abstract
Background B-cell lymphoma 2 (Bcl-2), a protein involved in apoptosis, has been proven to have carcinogenic potential and is well documented. With the recent advancement in optical technology, it has become possible to observe subcellular organelles such as mitochondria in real-time without the need for staining. Consequently, we have examined the movement of mitochondria in cancer cells, correlating it with the regulation of Bcl-2. Methods Using a tomographic microscope, which can detect the internal structure of cells, we observed lung tumor cells. Cells were exposed to a laser beam (λ = 520 nm) inclined at 45°, and holographic images were recorded up to a depth of 30 µm of reconstruction. Results Intriguingly, lung tumor cells rapidly expelled mitochondria upon the attachment of Bcl-2 or B-cell lymphoma extra-large (Bcl-xL) inhibitors. On the other hand, we observed that tumor cells hijack mitochondria from T cells. The hijacked mitochondria were not immediately linked to tumor cell death, but they played a role in assisting granzyme B-induced tumor cell death. Due to lower levels of Bcl-2 and Bcl-xL on the mitochondria of T cells compared to lung tumor cells, immune cells depleted of Bcl-2 and Bcl-xL were co-cultured with the tumor cells. Conclusions As a result, a more effective tumor cell death induced by granzyme B was observed. Additionally, further enhanced anticancer immune response was observed in vivo. Together, we show that modified mitochondria of T cells can provide potential novel strategies towards tumor cell death.
Collapse
Affiliation(s)
- Minsuk Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07804, Korea.
| |
Collapse
|
28
|
Patil D, Raut S, Joshi M, Bhatt P, Bhatt LK. PAQR4 oncogene: a novel target for cancer therapy. Med Oncol 2024; 41:161. [PMID: 38767705 DOI: 10.1007/s12032-024-02382-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/06/2024] [Indexed: 05/22/2024]
Abstract
Despite decades of basic and clinical research and trials of promising new therapies, cancer remains a major cause of morbidity and mortality due to the emergence of drug resistance to anticancer drugs. These resistance events have a very well-understood underlying mechanism, and their therapeutic relevance has long been recognized. Thus, drug resistance continues to be a major obstacle to providing cancer patients with the intended "cure". PAQR4 (Progestin and AdipoQ Receptor Family Member 4) gene is a recently identified novel protein-coding gene associated with various human cancers and acts through different signaling pathways. PAQR4 has a significant influence on multiple proteins that may regulate various gene expressions and may develop chemoresistance. This review discusses the roles of PAQR4 in tumor immunity, carcinogenesis, and chemoresistance. This paper is the first review, discussing PAQR4 in the pathogenesis of cancer. The review further explores the PAQR4 as a potential target in various malignancies.
Collapse
Affiliation(s)
- Dipti Patil
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Swapnil Raut
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Mitesh Joshi
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India
| | - Purvi Bhatt
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Vile Parle (West), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
29
|
Horvat N, Chocarro S, Marques O, Bauer TA, Qiu R, Diaz-Jimenez A, Helm B, Chen Y, Sawall S, Sparla R, Su L, Klingmüller U, Barz M, Hentze MW, Sotillo R, Muckenthaler MU. Superparamagnetic Iron Oxide Nanoparticles Reprogram the Tumor Microenvironment and Reduce Lung Cancer Regrowth after Crizotinib Treatment. ACS NANO 2024; 18:11025-11041. [PMID: 38626916 PMCID: PMC11064219 DOI: 10.1021/acsnano.3c08335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 05/01/2024]
Abstract
ALK-positive NSCLC patients demonstrate initial responses to ALK tyrosine kinase inhibitor (TKI) treatments, but eventually develop resistance, causing rapid tumor relapse and poor survival rates. Growing evidence suggests that the combination of drug and immune therapies greatly improves patient survival; however, due to the low immunogenicity of the tumors, ALK-positive patients do not respond to currently available immunotherapies. Tumor-associated macrophages (TAMs) play a crucial role in facilitating lung cancer growth by suppressing tumoricidal immune activation and absorbing chemotherapeutics. However, they can also be programmed toward a pro-inflammatory tumor suppressive phenotype, which represents a highly active area of therapy development. Iron loading of TAMs can achieve such reprogramming correlating with an improved prognosis in lung cancer patients. We previously showed that superparamagnetic iron oxide nanoparticles containing core-cross-linked polymer micelles (SPION-CCPMs) target macrophages and stimulate pro-inflammatory activation. Here, we show that SPION-CCPMs stimulate TAMs to secrete reactive nitrogen species and cytokines that exert tumoricidal activity. We further show that SPION-CCPMs reshape the immunosuppressive Eml4-Alk lung tumor microenvironment (TME) toward a cytotoxic profile hallmarked by the recruitment of CD8+ T cells, suggesting a multifactorial benefit of SPION-CCPM application. When intratracheally instilled into lung cancer-bearing mice, SPION-CCPMs delay tumor growth and, after first line therapy with a TKI, halt the regrowth of relapsing tumors. These findings identify SPIONs-CCPMs as an adjuvant therapy, which remodels the TME, resulting in a delay in the appearance of resistant tumors.
Collapse
Affiliation(s)
- Natalie
K. Horvat
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Sara Chocarro
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Oriana Marques
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Tobias A. Bauer
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ruiyue Qiu
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Alberto Diaz-Jimenez
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Barbara Helm
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
| | - Yuanyuan Chen
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Stefan Sawall
- X-ray
Imaging and CT, German Cancer Research Center
(DKFZ), Im Neuenheimer
Feld 280, 69120, Heidelberg, Germany
| | - Richard Sparla
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Lu Su
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ursula Klingmüller
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Matthias Barz
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
- Department
of Dermatology, University Medical Center
of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias W. Hentze
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Meyerhofstr.1, 69117, Heidelberg, Germany
| | - Rocío Sotillo
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Martina U. Muckenthaler
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, 69120, Heidelberg/Mannheim, Germany
| |
Collapse
|
30
|
Lee JU, Kim SH, Lee SH, Ji MJ, Jin JA, So HJ, Song ML, Lee HK, Kang TW. Combinational Pulsing of TAAs Enforces Dendritic Cell-Based Immunotherapy through T-Cell Proliferation and Interferon-γ Secretion in LLC1 Mouse Model. Cancers (Basel) 2024; 16:409. [PMID: 38254898 PMCID: PMC10814594 DOI: 10.3390/cancers16020409] [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: 12/15/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
NSCLC, the most common type of lung cancer, is often diagnosed late due to minimal early symptoms. Its high risk of recurrence or metastasis post-chemotherapy makes DC-based immunotherapy a promising strategy, offering targeted cancer destruction, low side effects, memory formation, and overcoming the immune evasive ability of cancers. However, the limited response to DCs pulsed with single antigens remains a significant challenge. To overcome this, we enhanced DC antigen presentation by pulsing with TAAs. Our study focused on enhancing DC-mediated immune response specificity and intensity by combinatorial pulsing of TAAs, selected for their prevalence in NSCLC. We selected four types of TAAs expressed in NSCLC and pulsed DCs with the optimal combination. Next, we administered TAAs-pulsed DCs into the LLC1 mouse model to evaluate their anti-tumor efficacy. Our results showed that TAAs-pulsed DCs significantly reduced tumor size and promoted apoptosis in tumor tissue. Moreover, TAAs-pulsed DCs significantly increased total T cells in the spleen compared to the unpulsed DCs. Additionally, in vitro stimulation of splenocytes from the TAAs-pulsed DCs showed notable T-cell proliferation and increased IFN-γ secretion. Our findings demonstrate the potential of multiple TAA pulsing to enhance the antigen-presenting capacity of DCs, thereby strengthening the immune response against tumors.
Collapse
Affiliation(s)
- Jae-Ung Lee
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Sang-Heon Kim
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Sung-Hoon Lee
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Min-Jae Ji
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Jeong-Ah Jin
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | - Hyung-Joon So
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| | | | - Hong-Ki Lee
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
- EHLCell Clinic, Seoul 06029, Republic of Korea
| | - Tae-Wook Kang
- Institute of Cell Biology and Regenerative Medicine, EHLBio Co., Ltd., Uiwang-si 16006, Republic of Korea; (J.-U.L.); (S.-H.K.)
| |
Collapse
|
31
|
Abd Aziz NA, Awang N, Kamaludin NF, Anuar NNM, Hamid A, Chan KM, Arshad S. The Development of Organotin(IV) N-Ethyl-N-Benzyldithiocarbamate Complexes: A Study on Their Synthesis, Characterization, and Cytocidal Effects on A549 Cell Line. Anticancer Agents Med Chem 2024; 24:942-953. [PMID: 38629375 DOI: 10.2174/0118715206309421240402093335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Organotin(IV) complexes of dithiocarbamate are vital in medicinal chemistry, exhibiting potential in targeting cancer cells due to their unique properties that enhance targeted delivery. This study aimed to synthesize and characterize organotin(IV) N-ethyl-N-benzyldithiocarbamate complexes (ONBDCs) and evaluate their cytotoxicity against A549 cells, which are commonly used as a model for human lung cancer research. METHODS The two ONBDC derivatives - ONBDC 1 (dimethyltin(IV) N-ethyl-N-benzyldithiocarbamate) and ONBDC 2 (triphenyltin(IV) N-ethyl-N-benzyldithiocarbamate) - were synthesized via the reaction of tin(IV) chloride with N-ethylbenzylamine in the presence of carbon disulfide. A range of analytical techniques, including elemental analysis, IR spectroscopy, NMR spectroscopy, UV-Vis spectrometry, TGA/DTA analysis, and X-ray crystallography, was conducted to characterize these compounds comprehensively. The cytotoxic effects of ONBDCs against A549 cells were evaluated using MTT assay. RESULTS Both compounds were synthesized and characterized successfully via elemental and spectroscopies analysis. MTT assay revealed that ONBDC 2 demonstrated remarkable cytotoxicity towards A549 cells, with an IC50 value of 0.52 μM. Additionally, ONBDC 2 displayed significantly higher cytotoxic activity against the A549 cell line when compared to the commercially available chemotherapeutic agent cisplatin (IC50: 32 μM). CONCLUSION Thus, it was shown that ONBDC 2 could have important anticancer properties and should be further explored as a top contender for creating improved and specialized cancer treatments.
Collapse
Affiliation(s)
- Nurul Amalina Abd Aziz
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Normah Awang
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nurul Farahana Kamaludin
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Asmah Hamid
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, University Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Kok Meng Chan
- Product Stewardship and Toxicology, Petroliam Nasional Berhad, Level 13, Tower 1, PETRONAS Twin Towers, KLCC, 50088 Kuala Lumpur, Malaysia
| | - Suhana Arshad
- X-ray Crystallography Unit, School of Physics, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| |
Collapse
|
32
|
Eghtedari S, Behdani M, Kazemi-Lomedasht F. Neuropilin-1 Binding Peptide as Fusion to Diphtheria Toxin Induces Apoptosis in Non-small Cell Lung Cancer Cell Line. Curr Pharm Des 2024; 30:1317-1325. [PMID: 38584554 DOI: 10.2174/0113816128292382240325074032] [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: 11/11/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Targeted cancer therapy can be considered as a new strategy to overcome the side effects of current cancer treatments. Neuropilin-1 (NRP-1) is a transmembrane glycoprotein that is expressed in endothelial cells and tumor vessels to stimulate angiogenesis progression. Targeted diphtheria toxin (DT)- based therapeutics are promising tools for cancer treatment. This study aimed to construct a novel NRP-1 binding peptide (as three repeats) (CRGDK) as a fusion to truncated DT (DTA) (DTA-triCRGDK) for targeted delivery of DT into NRP-1 expressing cells. METHODS The concept of DTA-triCRGDK was designed, synthesized and cloned into the bacterial host. Expression of DTA-triCRGDK was induced by Isopropyl ß-D-1-thiogalactopyranoside (IPTG) and purification was performed using Ni-NTA chromatography. Biological activity of DTA-triCRGDK was evaluated using MTT, apoptosis, and wound healing assays. In addition, expression levels of apoptotic Bax, Bcl2, and Casp3 genes were determined by Real-time PCR. RESULTS Cytotoxicity analysis showed the IC50 values of DTA-triCRGDK for A549 and MRC5 were 0.43 nM and 4.12 nM after 24 h, respectively. Bcl2 expression levels decreased 0.4 and 0.72 fold in A549 and MRC5, respectively. However, Bax and Casp3 expression level increased by 6.75 and 8.19 in A549 and 2.51 and 3.6 in MRC5 cells. CONCLUSION Taken together, DTA-triCRGDK is a promising tool for targeted therapy of NRP-1 overexpressing cancer cells.
Collapse
Affiliation(s)
- Sara Eghtedari
- Department of Biology, Faculty of Basic Sciences, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
33
|
Wu L, Gao C. Comprehensive Overview the Role of Glycosylation of Extracellular Vesicles in Cancers. ACS OMEGA 2023; 8:47380-47392. [PMID: 38144130 PMCID: PMC10734006 DOI: 10.1021/acsomega.3c07441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 12/26/2023]
Abstract
Extracellular vesicles (EVs) are membranous structures secreted by various cells carrying diverse biomolecules. Recent advancements in EV glycosylation research have underscored their crucial role in cancer. This review provides a global overview of EV glycosylation research, covering aspects such as specialized techniques for isolating and characterizing EV glycosylation, advances on how glycosylation affects the biogenesis and uptake of EVs, and the involvement of EV glycosylation in intracellular protein expression, cellular metastasis, intercellular interactions, and potential applications in immunotherapy. Furthermore, through an extensive literature review, we explore recent advances in EV glycosylation research in the context of cancer, with a focus on lung, colorectal, liver, pancreatic, breast, ovarian, prostate, and melanoma cancers. The primary objective of this review is to provide a comprehensive update for researchers, whether they are seasoned experts in the field of EVs or newcomers, aiding them in exploring new avenues and gaining a deeper understanding of EV glycosylation mechanisms. This heightened comprehension not only enhances researchers' knowledge of the pathogenic mechanisms of EV glycosylation but also paves the way for innovative cancer diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Linlin Wu
- Department of Clinical
Laboratory
Medicine Center, Yueyang Hospital of Integrated Traditional Chinese
and Western Medicine, Shanghai University
of Traditional Chinese Medicine, Shanghai 200437, China
| | - Chunfang Gao
- Department of Clinical
Laboratory
Medicine Center, Yueyang Hospital of Integrated Traditional Chinese
and Western Medicine, Shanghai University
of Traditional Chinese Medicine, Shanghai 200437, China
| |
Collapse
|
34
|
Le MT, Nguyen HT, Nguyen XH, Do XH, Mai BT, Ngoc Nguyen HT, Trang Than UT, Nguyen TH. Regulation and therapeutic potentials of microRNAs to non-small cell lung cancer. Heliyon 2023; 9:e22080. [PMID: 38058618 PMCID: PMC10696070 DOI: 10.1016/j.heliyon.2023.e22080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for 80%-85% of total cases and leading to millions of deaths worldwide. Drug resistance is the primary cause of treatment failure in NSCLC, which urges scientists to develop advanced approaches for NSCLC treatment. Among novel approaches, the miRNA-based method has emerged as a potential approach as it allows researchers to modulate target gene expression. Subsequently, cell behaviors are altered, which leads to the death and the depletion of cancer cells. It has been reported that miRNAs possess the capacity to regulate multiple genes that are involved in various signaling pathways, including the phosphoinositide 3-kinase, receptor tyrosine kinase/rat sarcoma virus/mitogen-activated protein kinase, wingless/integrated, retinoblastoma, p53, transforming growth factor β, and nuclear factor-kappa B pathways. Dysregulation of these signaling pathways in NSCLC results in abnormal cell proliferation, tissue invasion, and drug resistance while inhibiting apoptosis. Thus, understanding the roles of miRNAs in regulating these signaling pathways may enable the development of novel NSCLC treatment therapies. However, a comprehensive review of potential miRNAs in NSCLC treatment has been lacking. Therefore, this review aims to fill the gap by summarizing the up-to-date information on miRNAs regarding their targets, impact on cancer-associated pathways, and prospective outcomes in treating NSCLC. We also discuss current technologies for delivering miRNAs to the target cells, including virus-based, non-viral, and emerging extracellular vesicle-based delivery systems. This knowledge will support future studies to develop an innovative miRNA-based therapy and select a suitable carrier to treat NSCLC effectively.
Collapse
Affiliation(s)
- Mai Thi Le
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, 100000, Viet Nam
| | - Huyen-Thu Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Xuan-Hung Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- College of Health Sciences, Vin University, Hanoi, 100000, Viet Nam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Xuan-Hai Do
- Department of Gastroenterology, 108 Military Central Hospital, Hanoi, Viet Nam
| | - Binh Thanh Mai
- Department of Practical and Experimental Surgery, Vietnam Military Medical University, 160 Phung Hung Street, Phuc La, Ha Dong, Hanoi, Viet Nam
| | - Ha Thi Ngoc Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Uyen Thi Trang Than
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| | - Thanh-Hong Nguyen
- Vinmec Hi-tech Center, Vinmec Healthcare System, Hanoi, 100000, Viet Nam
| |
Collapse
|
35
|
Abdel-Razeq H, Sharaf B, Al-Jaghbeer MJ, Abu-Fares H, Bater R, Shaer MA, Abu-Jaish H, Laban DA, Salamah O, Tamimi F, Ashouri K, Salameh T, Zughul R, Alhalaseh Y. COMPASS-CAT versus Khorana risk assessment model for predicting venous thromboembolic events in patients with non-small cell lung cancer on active treatment with chemotherapy and/or immunotherapy, the CK-RAM study. J Thromb Thrombolysis 2023; 56:447-453. [PMID: 37430158 DOI: 10.1007/s11239-023-02860-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/25/2023] [Indexed: 07/12/2023]
Abstract
Cancer patients are at higher risk for venous thromboembolism (VTE). Several risk assessment models (RAM), including the Khorana and COMPASS-CAT, were developed to help predict the occurrence of VTE in cancer patients on active anti-cancer therapy. We aim to study the prevalence and predictors of VTE among patients with non-small cell lung cancer (NSCLC) and compare both RAMs in predicting VTE in patients with NSCLC were retrospectively reviewed. Variables known to increase the risk of VTE were collected and risk of VTE was assessed using both Khorana and COMPASS-CAT RAM. A total of 508 patients (mean age ± SD, 58.4 ± 12.2 years) were enrolled. Most (n = 357, 70.3%) patients had adenocarcinoma, and 333 (65.6%) patients had metastatic disease. VTE were confirmed in 76 (15.0%) patients. Rates were higher among patients with metastatic disease (19.8%, p < 0.001), adenocarcinoma (17.4%, p = 0.01) and those treated with immunotherapy (23.5%, p = 0.014). VTE rates were 21.2%, 14.1% and 13.9% among those with high (n = 66), intermediate (n = 341) and low (n = 101) Khorana risk scores, respectively (p = 0.126). On the other hand, 190 (37.4%) were classified as high risk by the COMPASS-CAT RAM; 52 (27.4%) of them had VTE compared to 24 (7.5%) of the remaining 318 (62.6%) classified as Low/Intermediate risk level, p < 0.001. In conclusion, patients with NSCLC are at high risk for VTE, especially those with adenocarcinoma, metastatic disease and when treated with immunotherapy. Compared to Khorana RAM, COMPASS-CAT RAM was better in identifying more patients in high-risk group, with higher VTE rate.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan.
- School of Medicine, The University of Jordan, Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan.
| | - Baha' Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | | | - Hala Abu-Fares
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Rayan Bater
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Moataz Abu Shaer
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hala Abu-Jaish
- School of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Dima Abu Laban
- Department of Radiology, King Hussein Cancer Center, Amman, Jordan
| | - Osama Salamah
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Faris Tamimi
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Khalid Ashouri
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Tareq Salameh
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Raed Zughul
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Yazan Alhalaseh
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
36
|
Akash S, Bibi S, Biswas P, Mukerjee N, Khan DA, Hasan MN, Sultana NA, Hosen ME, Jardan YAB, Nafidi HA, Bourhia M. Revolutionizing anti-cancer drug discovery against breast cancer and lung cancer by modification of natural genistein: an advanced computational and drug design approach. Front Oncol 2023; 13:1228865. [PMID: 37817764 PMCID: PMC10561655 DOI: 10.3389/fonc.2023.1228865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/15/2023] [Indexed: 10/12/2023] Open
Abstract
Breast and lung cancer are two of the most lethal forms of cancer, responsible for a disproportionately high number of deaths worldwide. Both doctors and cancer patients express alarm about the rising incidence of the disease globally. Although targeted treatment has achieved enormous advancements, it is not without its drawbacks. Numerous medicines and chemotherapeutic drugs have been authorized by the FDA; nevertheless, they can be quite costly and often fall short of completely curing the condition. Therefore, this investigation has been conducted to identify a potential medication against breast and lung cancer through structural modification of genistein. Genistein is the active compound in Glycyrrhiza glabra (licorice), and it exhibits solid anticancer efficiency against various cancers, including breast cancer, lung cancer, and brain cancer. Hence, the design of its analogs with the interchange of five functional groups-COOH, NH2 and OCH3, Benzene, and NH-CH2-CH2-OH-have been employed to enhance affinities compared to primary genistein. Additionally, advanced computational studies such as PASS prediction, molecular docking, ADMET, and molecular dynamics simulation were conducted. Firstly, the PASS prediction spectrum was analyzed, revealing that the designed genistein analogs exhibit improved antineoplastic activity. In the prediction data, breast and lung cancer were selected as primary targets. Subsequently, other computational investigations were gradually conducted. The mentioned compounds have shown acceptable results for in silico ADME, AMES toxicity, and hepatotoxicity estimations, which are fundamental for their oral medication. It is noteworthy that the initial binding affinity was only -8.7 kcal/mol against the breast cancer targeted protein (PDB ID: 3HB5). However, after the modification of the functional group, when calculating the binding affinities, it becomes apparent that the binding affinities increase gradually, reaching a maximum of -11.0 and -10.0 kcal/mol. Similarly, the initial binding affinity was only -8.0 kcal/mol against lung cancer (PDB ID: 2P85), but after the addition of binding affinity, it reached -9.5 kcal/mol. Finally, a molecular dynamics simulation was conducted to study the molecular models over 100 ns and examine the stability of the docked complexes. The results indicate that the selected complexes remain highly stable throughout the 100-ns molecular dynamics simulation runs, displaying strong correlations with the binding of targeted ligands within the active site of the selected protein. It is important to further investigate and proceed to clinical or wet lab experiments to determine the practical value of the proposed compounds.
Collapse
Affiliation(s)
- Shopnil Akash
- Faculty of Allied Health Science, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Partha Biswas
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Nobendu Mukerjee
- Department of Microbiology, West Bengal State University, Kolkata, India
| | - Dhrubo Ahmed Khan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Nazmul Hasan
- Laboratory of Pharmaceutical Biotechnology and Bioinformatics, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Nazneen Ahmeda Sultana
- Faculty of Allied Health Science, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Md. Eram Hosen
- Professor Joarder DNA and Chromosome Research Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Yousef A. Bin Jardan
- Department of Food Science, Faculty of Agricultural and Food Sciences, Laval University, Quebec City, QC, Canada
| | - Hiba-Allah Nafidi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, Ibn Zohr University, Laayoune, Morocco
| |
Collapse
|
37
|
Kuo CW, Lin CY, Wei SH, Chou YT, Chen CW, Tsai JS, Su PL, Lin CC. Navigating the challenges of invasive pulmonary aspergillosis in lung cancer treatment: a propensity score study. Ther Adv Med Oncol 2023; 15:17588359231198454. [PMID: 37720497 PMCID: PMC10503299 DOI: 10.1177/17588359231198454] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Background Invasive pulmonary aspergillosis (IPA) can negatively impact cancer patients' survival. It remains uncertain whether IPA's impact on patient outcomes varies by treatment approach in advanced lung cancer. Objectives To explore the association between IPA and outcomes in patients with advanced lung cancer receiving different treatments. Design A retrospective cohort study. Methods We enrolled patients with advanced-stage lung cancer between 2013 and 2021 at a college hospital in Taiwan and used the 2021 European Organization for Research and Treatment of Cancer/Mycoses Study Group Education and Research Consortium consensus for IPA diagnosis. Multivariable logistic regression was used to identify the IPA risk factors. We compared overall survival (OS) and postgalactomannan (GM) test survival between the IPA and control groups using multivariable Cox proportional hazards regression and the Kaplan-Meier method with propensity score matching (PSM). Results Among 2543 patients with advanced-stage lung cancer, 290 underwent a GM test, of which 34 (11.7%) were diagnosed with IPA. Patients undergoing chemotherapy (HR = 4.02, p = 0.027) and immunotherapy [hazard ratio (HR) = 3.41, p = 0.076] tended to have IPA. Compared to the control group, the IPA group had shorter median OS (14.4 versus 9.9 months, p = 0.030) and post-GM test survival (4.5 versus 1.9 months, p = 0.003). IPA was associated with shorter OS (log-rank p = 0.014 and 0.018 before and after PSM, respectively) and shorter 1-year and 2-year survival post-GM test (HR = 1.65 and 1.66, respectively). Patients receiving chemotherapy or immunotherapy had a shorter post-GM test survival if they had IPA. Conclusions IPA tended to be diagnosed more frequently in patients receiving chemotherapy or immune checkpoint inhibitors. Patients diagnosed with IPA are associated with shorter survival. Larger cohort studies are needed to verify the observations.
Collapse
Affiliation(s)
- Chin-Wei Kuo
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Chien-Yu Lin
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Sheng-Huan Wei
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Yun-Tse Chou
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Chian-Wei Chen
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Jeng-Shiuan Tsai
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Po-Lan Su
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Chien-Chung Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan
- Division of Chest Medicine, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 704
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan
| |
Collapse
|
38
|
Cao B, Huang L, Liu M, Lin H, Ma T, Zhao Y, Geng Y, Yang Y, Guo H, Li J. Phase 1 study to evaluate the effects of rifampin or itraconazole on the pharmacokinetics of limertinib (ASK120067), a novel mutant-selective inhibitor of the epidermal growth factor receptor in healthy Chinese subjects. Expert Opin Drug Metab Toxicol 2023; 19:653-664. [PMID: 37811634 DOI: 10.1080/17425255.2023.2260738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Limertinib is a novel mutant-selective and irreversible inhibitor of the epidermal growth factor receptor under development. A phase 1 open, two-period, single-sequence, self-controlled, two-part study was initiated to characterize the effects of a strong CYP3A4 inducer (rifampin) or inhibitor (itraconazole) on the pharmacokinetics of limertinib. RESEARCH DESIGN AND METHODS Twenty-four healthy subjects in each part received a single dose of limertinib alone (160 mg, Part A; 80 mg, Part B) and with multiple doses of rifampin 600 mg once daily (Part A) or itraconazole 200 mg twice daily (Part B). RESULTS Coadministration of rifampin decreased exposure (area under the plasma concentration-time curve from time 0 to infinity, AUC0-inf) of limertinib and its active metabolite CCB4580030 by 87.86% (geometric least-squares mean [GLSM] ratio, 12.14%; 90% confidence interval [CI], 9.89-14.92) and 66.82% (GLSM ratio, 33.18%; 90% CI, 27.72-39.72), respectively. Coadministration of itraconazole increased the AUC0-inf of limertinib by 289.8% (GLSM ratio, 389.8%; 90% CI, 334.07-454.82), but decreased that of CCB4580030 by 35.96% (GLSM ratio, 64.04%; 90% CI, 50.78-80.77). CONCLUSIONS Our study demonstrates that the concomitant use of limertinib with strong CYP3A inducers or inhibitors is not recommended. A single dose of limertinib, administered with or without rifampin or itraconazole, is generally safe and well tolerated in healthy Chinese subjects. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov identifier is NCT05631678.
Collapse
Affiliation(s)
- Bei Cao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lei Huang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ming Liu
- Clinical Pharmacology Department, Jiangsu Aosaikang Pharmaceutical Co. Ltd, Nanjing, China
| | - Hui Lin
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tingting Ma
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yu Zhao
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Geng
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuanxun Yang
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haifang Guo
- Clinical Pharmacology Department, Jiangsu Aosaikang Pharmaceutical Co. Ltd, Nanjing, China
| | - Juan Li
- Phase I Clinical Trials Unit, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
39
|
Zhu Q, Ni R, Guan X. Cost-effectiveness analysis of anlotinib as a third-line or further treatment for advanced non-small cell lung cancer in China. Transl Lung Cancer Res 2023; 12:1782-1789. [PMID: 37691869 PMCID: PMC10483086 DOI: 10.21037/tlcr-23-456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
Background The health expenditure on treatment of advanced non-small cell lung cancer (NSCLC) is enormous, especially in third-line or further therapy. Cost-effectiveness analysis for the treatment of advanced NSCLC is particularly important. Anlotinib has been approved by the China Food and Drug Administration (CFDA) for the third-line or further treatment of advanced NSCLC. The price of anlotinib in China fell in 2022. Thus, this study evaluated the cost-effectiveness of anlotinib in the third-line or further treatment of patients with advanced NSCLC based on the newest price from the Chinese health-care system perspective. Methods A Markov model was developed to compare the lifetime costs and effectiveness of anlotinib and a placebo in the third-line or further treatment of patients with advanced NSCLC based on outcome data from the ALTER 0303 phase-3 randomized clinical trial, which included 437 patients with advanced NSCLC and investigated the efficacy of anlotinib. The lifetime costs and quality-adjusted life years (QALYs) were estimated. One-way and probabilistic sensitivity analyses were performed to evaluate the model uncertainty. Results Anlotinib provided an additional 0.1161 QALYs compared to the placebo. The corresponding incremental cost was ¥22,729. The incremental cost-effectiveness ratio (ICER) of anlotinib compared to the placebo was ¥195,768 per QALY. From the perspective of the Chinese health-care system, anlotinib was found to be cost-effective compared to the placebo in the third-line or further treatment of patients with advanced NSCLC at a willingness-to-pay (WTP) threshold of ¥242,928 per QALY. Moreover, 1-way sensitivity analysis found that the results were sensitive to the utility of progressive disease (PD). The lower this parameter was, the higher the probability of ICER for anlotinib not being cost-effective. The cost-effectiveness acceptability curves showed that the base-case analysis results were relatively stable. Conclusions Considering the clinical efficacy, safety, and cost-effectiveness of anlotinib, it may be a valuable third-line or further treatment for advanced NSCLC in China.
Collapse
Affiliation(s)
- Qiuyan Zhu
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, China
| | - Ronghua Ni
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Guan
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
40
|
O’Shaughnessy M, Sheils O, Baird AM. The Lung Microbiome in COPD and Lung Cancer: Exploring the Potential of Metal-Based Drugs. Int J Mol Sci 2023; 24:12296. [PMID: 37569672 PMCID: PMC10419288 DOI: 10.3390/ijms241512296] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer 17 are two of the most prevalent and debilitating respiratory diseases worldwide, both associated with high morbidity and mortality rates. As major global health concerns, they impose a substantial burden on patients, healthcare systems, and society at large. Despite their distinct aetiologies, lung cancer and COPD share common risk factors, clinical features, and pathological pathways, which have spurred increasing research interest in their co-occurrence. One area of particular interest is the role of the lung microbiome in the development and progression of these diseases, including the transition from COPD to lung cancer. Exploring novel therapeutic strategies, such as metal-based drugs, offers a potential avenue for targeting the microbiome in these diseases to improve patient outcomes. This review aims to provide an overview of the current understanding of the lung microbiome, with a particular emphasis on COPD and lung cancer, and to discuss the potential of metal-based drugs as a therapeutic strategy for these conditions, specifically concerning targeting the microbiome.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, St. James’s Hospital, D08 RX0X Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| |
Collapse
|
41
|
Restrepo JC, Dueñas D, Corredor Z, Liscano Y. Advances in Genomic Data and Biomarkers: Revolutionizing NSCLC Diagnosis and Treatment. Cancers (Basel) 2023; 15:3474. [PMID: 37444584 DOI: 10.3390/cancers15133474] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a significant public health concern with high mortality rates. Recent advancements in genomic data, bioinformatics tools, and the utilization of biomarkers have improved the possibilities for early diagnosis, effective treatment, and follow-up in NSCLC. Biomarkers play a crucial role in precision medicine by providing measurable indicators of disease characteristics, enabling tailored treatment strategies. The integration of big data and artificial intelligence (AI) further enhances the potential for personalized medicine through advanced biomarker analysis. However, challenges remain in the impact of new biomarkers on mortality and treatment efficacy due to limited evidence. Data analysis, interpretation, and the adoption of precision medicine approaches in clinical practice pose additional challenges and emphasize the integration of biomarkers with advanced technologies such as genomic data analysis and artificial intelligence (AI), which enhance the potential of precision medicine in NSCLC. Despite these obstacles, the integration of biomarkers into precision medicine has shown promising results in NSCLC, improving patient outcomes and enabling targeted therapies. Continued research and advancements in biomarker discovery, utilization, and evidence generation are necessary to overcome these challenges and further enhance the efficacy of precision medicine. Addressing these obstacles will contribute to the continued improvement of patient outcomes in non-small cell lung cancer.
Collapse
Affiliation(s)
- Juan Carlos Restrepo
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Diana Dueñas
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Zuray Corredor
- Grupo de Investigaciones en Odontología (GIOD), Facultad de Odontología, Universidad Cooperativa de Colombia, Pasto 520002, Colombia
- Facultad de Salud, Departamento de Ciencias Básicas, Universidad Libre, Cali 760026, Colombia
| | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| |
Collapse
|
42
|
Wang L, Yang Z, Guo F, Chen Y, Wei J, Dai X, Zhang X. Research progress of biomarkers in the prediction of anti-PD-1/PD-L1 immunotherapeutic efficiency in lung cancer. Front Immunol 2023; 14:1227797. [PMID: 37465684 PMCID: PMC10351040 DOI: 10.3389/fimmu.2023.1227797] [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: 05/23/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023] Open
Abstract
Currently, anti-PD-1/PD-L1 immunotherapy using immune checkpoint inhibitors is widely used in the treatment of multiple cancer types including lung cancer, which is a leading cause of cancer death in the world. However, only a limited proportion of lung cancer patients will benefit from anti-PD-1/PD-L1 therapy. Therefore, it is of importance to predict the response to immunotherapy for the precision treatment of patients. Although the expression of PD-L1 and tumor mutation burden (TMB) are commonly used to predict the clinical response of anti-PD-1/PD-L1 therapy, other factors such as tumor-specific genes, dMMR/MSI, and gut microbiome are also promising predictors for immunotherapy in lung cancer. Furthermore, invasive peripheral blood biomarkers including blood DNA-related biomarkers (e.g., ctDNA and bTMB), blood cell-related biomarkers (e.g., immune cells and TCR), and other blood-related biomarkers (e.g., soluble PD-L1 and cytokines) were utilized to predict the immunotherapeutic response. In this review, the current achievements of anti-PD-1/PD-L1 therapy and the potential biomarkers for the prediction of anti-PD-1/PD-L1 immunotherapy in lung cancer treatment were summarized and discussed.
Collapse
Affiliation(s)
- Luyao Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Zongxing Yang
- Department of Clinical Laboratory, First Hospital of Jilin University, Changchun, China
| | - Fucheng Guo
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Yurong Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Jiarui Wei
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Rajkovic J, Novakovic R, Grujic-Milanovic J, Ydyrys A, Ablaikhanova N, Calina D, Sharifi-Rad J, Al-Omari B. An updated pharmacological insight into calotropin as a potential therapeutic agent in cancer. Front Pharmacol 2023; 14:1160616. [PMID: 37138852 PMCID: PMC10149670 DOI: 10.3389/fphar.2023.1160616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/05/2023] [Indexed: 05/05/2023] Open
Abstract
Calotropin is a pharmacologically active compound isolated from milkweed plants like Calotropis procera, Calotropis gigantea, and Asclepias currasavica that belong to the Asclepiadaceae family. All of these plants are recognised as medical traditional plants used in Asian countries. Calotropin is identified as a highly potent cardenolide that has a similar chemical structure to cardiac glycosides (such as digoxin and digitoxin). During the last few years, cytotoxic and antitumor effects of cardenolides glycosides have been reported more frequently. Among cardenolides, calotropin is identified as the most promising agent. In this updated and comprehensive review, we aimed to analyze and discuss the specific mechanisms and molecular targets of calotropin in cancer treatment to open new perspectives for the adjuvant treatment of different types of cancer. The effects of calotropin on cancer have been extensively studied in preclinical pharmacological studies in vitro using cancer cell lines and in vivo in experimental animal models that have targeted antitumor mechanisms and anticancer signaling pathways. The analyzed information from the specialized literature was obtained from scientific databases until December 2022, mainly from PubMed/MedLine, Google Scholar, Scopus, Web of Science, and Science Direct databases using specific MeSH search terms. The results of our analysis demonstrate that calotropin can be a potential chemotherapeutic/chemopreventive adjunctive agent in cancer pharmacotherapeutic management.
Collapse
Affiliation(s)
- Jovana Rajkovic
- Institute for Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Radmila Novakovic
- Institute for Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelica Grujic-Milanovic
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Nurzhanat Ablaikhanova
- Department of Biophysics, Biomedicine and Neuroscience, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | | - Basem Al-Omari
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
44
|
Development of a 5-FU modified miR-129 mimic as a therapeutic for non-small cell lung cancer. Mol Ther Oncolytics 2023; 28:277-292. [PMID: 36911069 PMCID: PMC9995506 DOI: 10.1016/j.omto.2023.02.007] [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: 09/16/2022] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the United States with non-small cell lung cancer (NSCLC) accounting for most cases. Despite advances in cancer therapeutics, the 5-year survival rate has remained poor due to several contributing factors, including its resistance to therapeutics. Therefore, there is a pressing need to develop therapeutics that can overcome resistance. Non-coding RNAs, including microRNAs (miRNAs), have been found to contribute to cancer resistance and therapeutics by modulating the expression of several targets involving multiple key mechanisms. In this study, we investigated the therapeutic potential of miR-129 modified with 5-fluorouracil (5-FU) in NSCLC. Our results show that 5-FU modified miR-129 (5-FU-miR-129) inhibits proliferation, induces apoptosis, and retains function as an miRNA in NSCLC cell lines A549 and Calu-1. Notably, we observed that 5-FU-miR-129 was able to overcome resistance to tyrosine kinase inhibitors and chemotherapy in cell lines resistant to erlotinib or 5-FU. Furthermore, we observed that the inhibitory effect of 5-FU-miR-129 can also be achieved in NSCLC cells under vehicle-free conditions. Finally, 5-FU-miR-129 inhibited NSCLC tumor growth and extended survival in vivo without toxic side effects. Altogether, our results demonstrate the potential of 5-FU-miR-129 as a highly potent cancer therapeutic in NSCLC.
Collapse
|
45
|
Zhang X, Wang X, Hou L, Xu Z, Liu Y, Wang X. Nanoparticles overcome adaptive immune resistance and enhance immunotherapy via targeting tumor microenvironment in lung cancer. Front Pharmacol 2023; 14:1130937. [PMID: 37033636 PMCID: PMC10080031 DOI: 10.3389/fphar.2023.1130937] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/16/2023] [Indexed: 04/11/2023] Open
Abstract
Lung cancer is one of the common malignant cancers worldwide. Immune checkpoint inhibitor (ICI) therapy has improved survival of lung cancer patients. However, ICI therapy leads to adaptive immune resistance and displays resistance to PD-1/PD-L1 blockade in lung cancer, leading to less immune response of lung cancer patients. Tumor microenvironment (TME) is an integral tumor microenvironment, which is involved in immunotherapy resistance. Nanomedicine has been used to enhance the immunotherapy in lung cancer. In this review article, we described the association between TME and immunotherapy in lung cancer. We also highlighted the importance of TME in immunotherapy in lung cancer. Moreover, we discussed how nanoparticles are involved in regulation of TME to improve the efficacy of immunotherapy, including Nanomedicine SGT-53, AZD1080, Nanomodulator NRF2, Cisplatin nanoparticles, Au@PG, DPAICP@ME, SPIO NP@M-P, NBTXR3 nanoparticles, ARAC nanoparticles, Nano-DOX, MS NPs, Nab-paclitaxel, GNPs-hPD-L1 siRNA. Furthermore, we concluded that targeting TME by nanoparticles could be helpful to overcome resistance to PD-1/PD-L1 blockade in lung cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xuemei Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lijian Hou
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zheng Xu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- School of Medicine, Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, Tongji University, Shanghai, China
| | - Xueju Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
- *Correspondence: Xueju Wang,
| |
Collapse
|
46
|
Combined time-restricted feeding and cisplatin enhance the anti-tumor effects in cisplatin-resistant and -sensitive lung cancer cells. Med Oncol 2023; 40:63. [PMID: 36576605 PMCID: PMC9797463 DOI: 10.1007/s12032-022-01923-5] [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: 11/14/2022] [Accepted: 12/04/2022] [Indexed: 12/29/2022]
Abstract
Combination therapy as an important treatment option for lung cancer has been attracting attention due to the primary and acquired resistance of chemotherapeutic drugs in the clinical application. In the present study, as a new therapy strategy, concomitant treatment with time-restricted feeding (TRF) plus cisplatin (DDP) on lung cancer growth was investigated in DDP-resistant and DDP-sensitive lung cancer cells. We first found that TRF significantly enhanced the drug susceptibility of DDP in DDP-resistant A549 (A549/DDP) cell line, illustrated by reversing the inhibitory concentration 50 (IC50) values of A549/DDP cells to normal level of parental A549 cells. We also found that TRF markedly enhanced DDP inhibition on cell proliferation, migration, as well as promoted apoptosis compared to the DDP alone group in A549, H460 and A549/DDP cells lines. We further revealed that the synergistic anti-tumor effect of combined DDP and TRF was greater than that of combined DDP and simulated fasting condition (STS), a known anti-tumor cellular medium. Moreover, mRNA sequence analysis from A549/DDP cell line demonstrated the synergistic anti-tumor effect involved in upregulated pathways in p53 signaling pathway and apoptosis. Notably, compared with the DDP alone group, combination of TRF and DDP robustly upregulated the P53 protein expression without mRNA level change by regulating its stability via promoting protein synthesis and inhibiting degradation, revealed by cycloheximide and MG132 experiments. Collectively, our results suggested that TRF in combination with cisplatin might be an additional novel therapeutic strategy for patients with lung cancer.
Collapse
|
47
|
Zhao D, Zhu D, Cai F, Jiang M, Liu X, Li T, Zheng Z. Current Situation and Prospect of Adoptive Cellular Immunotherapy for Malignancies. Technol Cancer Res Treat 2023; 22:15330338231204198. [PMID: 38037341 PMCID: PMC10693217 DOI: 10.1177/15330338231204198] [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: 04/06/2023] [Revised: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 12/02/2023] Open
Abstract
Adoptive cell immunotherapy (ACT) is an innovative promising treatment for tumors. ACT is characterized by the infusion of active anti-tumor immune cells (specific and non-specific) into patients to kill tumor cells either directly or indirectly by stimulating the body's immune system. The patient's (autologous) or a donor's (allogeneic) immune cells are used to improve immune function. Chimeric antigen receptor (CAR) T cells (CAR-T) is a type of ACT that has gained attention. T cells from the peripheral blood are genetically engineered to express CARs that rapidly proliferate and specifically recognize target antigens to exert its anti-tumor effects. Clinical application of CAR-T therapy for hematological tumors has shown good results, but adverse reactions and recurrence limit its applicability. Tumor infiltrating lymphocyte (TIL) therapy is effective for solid tumors. TIL therapy exhibits T cell receptor (TCR) clonality, superior tumor homing ability, and low targeted toxicity, but its successful application is limited to a number of tumors. Regardless, TIL and CAR-T therapies are effective for treating cancer. Additionally, CAR-natural killer (NK), CAR-macrophages (M), and TCR-T therapies are currently being researched. In this review, we highlight the current developments and limitations of several types of ACT.
Collapse
Affiliation(s)
- Dong Zhao
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Dantong Zhu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Fei Cai
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Mingzhe Jiang
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Xuefei Liu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Tingting Li
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Zhendong Zheng
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, P. R. China
| |
Collapse
|
48
|
Wang M, Zhu L, Yang X, Li J, Liu Y, Tang Y. Targeting immune cell types of tumor microenvironment to overcome resistance to PD-1/PD-L1 blockade in lung cancer. Front Pharmacol 2023; 14:1132158. [PMID: 36874015 PMCID: PMC9974851 DOI: 10.3389/fphar.2023.1132158] [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: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Lung cancer is the common malignant tumor with the highest mortality rate. Lung cancer patients have achieved benefits from immunotherapy, including immune checkpoint inhibitors (ICIs) therapy. Unfortunately, cancer patients acquire adaptive immune resistance, leading to poor prognosis. Tumor microenvironment (TME) has been demonstrated to play a critical role in participating in acquired adaptive immune resistance. TME is associated with molecular heterogeneity of immunotherapy efficacy in lung cancer. In this article, we discuss how immune cell types of TME are correlated with immunotherapy in lung cancer. Moreover, we describe the efficacy of immunotherapy in driven gene mutations in lung cancer, including KRAS, TP53, EGFR, ALK, ROS1, KEAP1, ZFHX3, PTCH1, PAK7, UBE3A, TNF-α, NOTCH, LRP1B, FBXW7, and STK11. We also emphasize that modulation of immune cell types of TME could be a promising strategy for improving adaptive immune resistance in lung cancer.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lijie Zhu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoxu Yang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiahui Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Ying Tang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|