1
|
Srinivasarao DA, Shah S, Famta P, Vambhurkar G, Jain N, Pindiprolu SKSS, Sharma A, Kumar R, Padhy HP, Kumari M, Madan J, Srivastava S. Unravelling the role of tumor microenvironment responsive nanobiomaterials in spatiotemporal controlled drug delivery for lung cancer therapy. Drug Deliv Transl Res 2025; 15:407-435. [PMID: 39037533 DOI: 10.1007/s13346-024-01673-z] [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] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
Design and development of efficient drug delivery technologies that impart site-specificity is the need of the hour for the effective treatment of lung cancer. The emergence of materials science and nanotechnology partially helped drug delivery scientists to achieve this objective. Various stimuli-responsive materials that undergo degradation at the pathological tumor microenvironment (TME) have been developed and explored for drug delivery applications using nanotechnological approaches. Nanoparticles (NPs), owing to their small size and high surface area to volume ratio, demonstrated enhanced cellular internalization, permeation, and retention at the tumor site. Such passive accumulation of stimuli-responsive materials helped to achieve spatiotemporally controlled and targeted drug delivery within the tumors. In this review, we discussed various stimuli-physical (interstitial pressure, temperature, and stiffness), chemical (pH, hypoxia, oxidative stress, and redox state), and biological (receptor expression, efflux transporters, immune cells, and their receptors or ligands)-that are characteristic to the TME. We mentioned an array of biomaterials-based nanoparticulate delivery systems that respond to these stimuli and control drug release at the TME. Further, we discussed nanoparticle-based combinatorial drug delivery strategies. Finally, we presented our perspectives on challenges related to scale-up, clinical translation, and regulatory approvals.
Collapse
Affiliation(s)
- Dadi A Srinivasarao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Naitik Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Sai Kiran S S Pindiprolu
- Aditya Pharmacy College, Surampalem, 533 437, Andhra Pradesh, India
- Jawaharlal Nehru Technological University, Kakinada, 533 003, Andhra Pradesh, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Meenu Kumari
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
2
|
Li S, Feng S, Chen Y, Sun B, Zhang N, Zhao Y, Han J, Liu Z, He YQ, Wang Q. Ciclopirox platinum(IV) conjugates suppress tumors by promoting mitophagy and provoking immune responses. J Inorg Biochem 2024; 260:112696. [PMID: 39142055 DOI: 10.1016/j.jinorgbio.2024.112696] [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: 05/09/2024] [Revised: 07/11/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Mitophagy is an important target for antitumor drugs development. A series of ciclopirox (CPX) platinum(IV) hybrids targeting PTEN induced putative kinase 1 (PINK1)/Parkin mediated mitophagy were designed and prepared as antitumor agents. The dual CPX platinum(IV) complex with cisplatin core was screened out as a candidate, which displayed promising antitumor activities both in vitro and in vivo. Mechanistically, it caused serious DNA damage in tumor cells. Then, remarkable mitochondrial damage was induced accompanied by the mitochondrial membrane depolarization and reactive oxygen species generation, which further promoted apoptosis through the Bcl-2/Bax/Caspase3 pathway. Furthermore, mitophagy was ignited via the PINK1/Parkin/P62/LC3 axis, and exhibited positive influence on promoting the apoptosis of tumor cells. The antitumor immunity was boosted by the block of immune check point programmed cell death ligand-1 (PD-L1), which further increased the density of T cells in tumors. Subsequently, the metastasis of tumor cells was inhibited by inhibiting angiogenesis in tumors.
Collapse
Affiliation(s)
- Suying Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China; Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Bin Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China; Liaocheng High-Tech Biotechnology Co., Ltd, Liaocheng 252059, PR China
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Yan-Qin He
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, PR China.
| |
Collapse
|
3
|
Li L, Qu D, Wang B, Yuan S, Zhao Y, Liu N, Huo F, Zhang D, Zhang L. FTO blocks RNA translational activity via the loss of N6-methyladenosine methylation at 5' UTR regulated by RBM5 in cisplatin-resistant NSCLC. J Cell Physiol 2024; 239:e31296. [PMID: 38742685 DOI: 10.1002/jcp.31296] [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: 12/21/2023] [Revised: 04/09/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
N6-methyladenosine (m6A) methylation has been widely regarded in numerous biological functions including CR. Nonetheless, the molecular process of m6A methylation behind CR in non-small cell lung cancer (NSCLC) has no apparent significance. We identified in this study that the expression of FTO alpha-ketoglutarate dependent dioxygenase (FTO) was downregulated in CR NSCLC tissues and cells in vivo and in vitro. Additionally, RIP-seq indicated that loss of FTO contributed to the elevated m6A methylation at 5'-untranslated region of RNAs which were closely connected with tumor resistance and malignancy, and FTO exerted to exclude the recruitment of eIF3A to these target genes in CR NSCLC. Moreover, FTO-enriched transcripts displayed a reduced translational capability in CR NSCLC compared to the regular NSCLC cells. Finally, we also identified RNA binding motif protein 5 (RBM5) that could specially interact with FTO in regular NSCLC compared to CR NSCLC. Deficiency of RBM5 resulted in the abnormal recognition of transcripts by FTO, and led to the translation silencing of genes associated with CR such as ATP7A, ERCC1, CD99, CDKN3, XRCC5, and NOL3. Taken together, our data characterized FTO as a novel translation regulator and revealed the molecular mechanism on gene translation through the synergistic effects with RBM5 and m6A methylation in CR NSCLC cells.
Collapse
Affiliation(s)
- Liantao Li
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Debao Qu
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bo Wang
- Department of Radiotherapy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shiwang Yuan
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Zhao
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Nianli Liu
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fuchun Huo
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dan Zhang
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Longzhen Zhang
- The First School of Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
4
|
Li Y, Ye X, Huang H, Cao R, Huang F, Chen L. Construction of a prognostic model based on memory CD4+ T cell-associated genes for lung adenocarcinoma and its applications in immunotherapy. CPT Pharmacometrics Syst Pharmacol 2024; 13:837-852. [PMID: 38594917 PMCID: PMC11098152 DOI: 10.1002/psp4.13122] [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: 11/03/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
The association between memory CD4+ T cells and cancer prognosis is increasingly recognized, but their impact on lung adenocarcinoma (LUAD) prognosis remains unclear. In this study, using the cell-type identification by estimating relative subsets of RNA transcripts algorithm, we analyzed immune cell composition and patient survival in LUAD. Weighted gene coexpression network analysis helped identify memory CD4+ T cell-associated gene modules. Combined with module genes, a five-gene LUAD prognostic risk model (HOXB7, MELTF, ABCC2, GNPNAT1, and LDHA) was constructed by regression analysis. The model was validated using the GSE31210 data set. The validation results demonstrated excellent predictive performance of the risk scoring model. Correlation analysis was conducted between the clinical information and risk scores of LUAD samples, revealing that LUAD patients with disease progression exhibited higher risk scores. Furthermore, univariate and multivariate regression analyses demonstrated the model independent prognostic capability. The constructed nomogram results demonstrated that the predictive performance of the nomogram was superior to the prognostic model and outperformed individual clinical factors. Immune landscape assessment was performed to compare different risk score groups. The results revealed a better prognosis in the low-risk group with higher immune infiltration. The low-risk group also showed potential benefits from immunotherapy. Our study proposes a memory CD4+ T cell-associated gene risk model as a reliable prognostic biomarker for personalized treatment in LUAD patients.
Collapse
Affiliation(s)
- Yong Li
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Xiangli Ye
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Huiqin Huang
- Fujian Provincial Key Laboratory of Medical TestingFujian Academy of Medical SciencesFuzhouChina
| | - Rongxiang Cao
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Feijian Huang
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| | - Limin Chen
- Pulmonary and Critical Care MedicineFujian Medical University Union HospitalFuzhouChina
| |
Collapse
|
5
|
Li S, Mao L, Song L, Xia X, Wang Z, Cheng Y, Lai J, Tang X, Chen X. Extracellular Vesicles Derived from Glioma Stem Cells Affect Glycometabolic Reprogramming of Glioma Cells Through the miR-10b-5p/PTEN/PI3K/Akt Pathway. Stem Cell Rev Rep 2024; 20:779-796. [PMID: 38294721 DOI: 10.1007/s12015-024-10677-8] [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] [Accepted: 01/08/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVE Glioma is one of the most prevalently diagnosed types of primary malignant brain tumors. Glioma stem cells (GSCs) are crucial in glioma recurrence. This study aims to elucidate the mechanism by which extracellular vehicles (EVs) derived from GSCs modulate glycometabolic reprogramming in glioma. METHODS Xenograft mouse models and cell models of glioma were established and treated with GSC-EVs. Additionally, levels and activities of PFK1, LDHA, and FASN were assessed to evaluate the effect of GSC-EVs on glycometabolic reprogramming in glioma. Glioma cell proliferation, invasion, and migration were evaluated using MTT, EdU, Colony formation, and Transwell assays. miR-10b-5p expression was determined, with its target gene PTEN and downstream pathway PI3K/Akt evaluated. The involvement of miR-10b-5p and the PI3K/Akt pathway in the effect of GSC-EVs on glycometabolic reprogramming was tested through joint experiments. RESULTS GSC-EVs facilitated glycometabolic reprogramming in glioma mice, along with enhancing glucose uptake, lactate level, and adenosine monophosphate-to-adenosine triphosphate ratio. Moreover, GSC-EV treatment potentiated glioma cell proliferation, invasion, and migration, reinforced cell resistance to temozolomide, and raised levels and activities of PFK1, LDHA, and FASN. miR-10b-5p was highly-expressed in GSC-EV-treated glioma cells while being carried into glioma cells by GSC-EVs. miR-10b-5p targeted PTEN and activated the PI3K/Akt pathway, hence stimulating glycometabolic reprogramming. CONCLUSION GSC-EVs target PTEN and activate the PI3K/Akt pathway through carrying miR-10b-5p, subsequently accelerating glycometabolic reprogramming in glioma, which might provide new insights into glioma treatment.
Collapse
Affiliation(s)
- Shun Li
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Neurosurgical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Lifang Mao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Lvmeng Song
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Xiaochao Xia
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Zihao Wang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yinchuan Cheng
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jinqing Lai
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Xiaoping Tang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
- Neurosurgical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| | - Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
6
|
Nazari S, Mosaffa F, Poustforoosh A, Mortazavi M, Saso L, Firuzi O, Moosavi F. Foretinib, a c-MET receptor tyrosine kinase inhibitor, tackles multidrug resistance in cancer cells by inhibiting ABCB1 and ABCG2 transporters. Toxicol Appl Pharmacol 2024; 484:116866. [PMID: 38367674 DOI: 10.1016/j.taap.2024.116866] [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: 08/27/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND ABC transporter-mediated multidrug resistance (MDR) remains a major obstacle for cancer pharmacological treatment. Some tyrosine kinase inhibitors (TKIs) have been shown to reverse MDR. The present study was designed to evaluate for the first time whether foretinib, a multitargeted TKI, can circumvent ABCB1 and ABCG2-mediated MDR in treatment-resistant cancer models. METHODS Accumulation of fluorescent substrates of ABCB1 and ABCG2 in ABCB1-overexpressing MES-SA/DX5 and ABCG2-overexpressing MCF-7/MX and their parenteral cells was evaluated by flow cytometry. The growth inhibitory activity of single and combination therapy of foretinib and chemotherapeutic drugs on MDR cells was examined by MTT assay. Analysis of combined interaction effects was performed using CalcuSyn software. RESULTS It was firstly proved that foretinib increased the intracellular accumulation of rhodamine 123 and mitoxantrone in MES-SA/DX5 and MCF-7/MX cancer cells, with accumulation ratios of 12 and 2.2 at 25 μM concentration, respectively. However, it did not affect the accumulation of fluorescent substrates in the parental cells. Moreover, foretinib synergistically improved the cytotoxic effects of doxorubicin and mitoxantrone. The means of combination index (CI) values at fraction affected (Fa) values of 0.5, 0.75, and 0.9 were 0.64 ± 0.08 and 0.47 ± 0.09, in MES-SA/DX5 and MCF-7/MX cancer cells, respectively. In silico analysis also suggested that the drug-binding domain of ABCB1 and ABCG2 transporters could be considered as potential target for foretinib. CONCLUSION Overall, our results suggest that foretinib can target MDR-linked ABCB1 and ABCG2 transporters in clinical cancer therapy.
Collapse
Affiliation(s)
- Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Motahareh Mortazavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Sito H, Sharzehan MAK, Islam MA, Tan SC. Genetic Variants Associated With Response to Platinum-Based Chemotherapy in Non-Small Cell Lung Cancer Patients: A Field Synopsis and Meta-Analysis. Br J Biomed Sci 2024; 81:11835. [PMID: 38450253 PMCID: PMC10914946 DOI: 10.3389/bjbs.2024.11835] [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: 07/27/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
Background: Publications on the associations of genetic variants with the response to platinum-based chemotherapy (PBC) in NSCLC patients have surged over the years, but the results have been inconsistent. Here, a comprehensive meta-analysis was conducted to combine eligible studies for a more accurate assessment of the pharmacogenetics of PBC in NSCLC patients. Methods: Relevant publications were searched in PubMed, Scopus, and Web of Science databases through 15 May 2021. Inclusion criteria for eligible publications include studies that reported genotype and allele frequencies of NSCLC patients treated with PBC, delineated by their treatment response (sensitive vs. resistant). Publications on cell lines or animal models, duplicate reports, and non-primary research were excluded. Epidemiological credibility of cumulative evidence was assessed using the Newcastle-Ottawa Scale (NOS) and Venice criteria. Begg's and Egger's tests were used to assess publication bias. Cochran's Q-test and I2 test were used to calculate the odds ratio and heterogeneity value to proceed with the random effects or fixed-effects method. Venice criteria were used to assess the strength of evidence, replication methods and protection against bias in the studies. Results: A total of 121 publications comprising 29,478 subjects were included in this study, and meta-analyses were performed on 184 genetic variants. Twelve genetic variants from 10 candidate genes showed significant associations with PBC response in NSCLC patients with strong or moderate cumulative epidemiological evidence (increased risk: ERCC1 rs3212986, ERCC2 rs1799793, ERCC2 rs1052555, and CYP1A1 rs1048943; decreased risk: GSTM1 rs36631, XRCC1 rs1799782 and rs25487, XRCC3 rs861539, XPC rs77907221, ABCC2 rs717620, ABCG2 rs2231142, and CDA rs1048977). Bioinformatics analysis predicted possible damaging or deleterious effects for XRCC1 rs1799782 and possible low or medium functional impact for CYP1A1 rs1048943. Conclusion: Our results provide an up-to-date summary of the association between genetic variants and response to PBC in NSCLC patients.
Collapse
Affiliation(s)
- Hilary Sito
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Md Asiful Islam
- WHO Collaborating Centre for Global Women’s Health, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Sito H, Tan SC. Genetic polymorphisms as potential pharmacogenetic biomarkers for platinum-based chemotherapy in non-small cell lung cancer. Mol Biol Rep 2024; 51:102. [PMID: 38217759 DOI: 10.1007/s11033-023-08915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/08/2023] [Indexed: 01/15/2024]
Abstract
Platinum-based chemotherapy (PBC) is a widely used treatment for various solid tumors, including non-small cell lung cancer (NSCLC). However, its efficacy is often compromised by the emergence of drug resistance in patients. There is growing evidence that genetic variations may influence the susceptibility of NSCLC patients to develop resistance to PBC. Here, we provide a comprehensive overview of the mechanisms underlying platinum drug resistance and highlight the important role that genetic polymorphisms play in this process. This paper discussed the genetic variants that regulate DNA repair, cellular movement, drug transport, metabolic processing, and immune response, with a focus on their effects on response to PBC. The potential applications of these genetic polymorphisms as predictive indicators in clinical practice are explored, as are the challenges associated with their implementation.
Collapse
Affiliation(s)
- Hilary Sito
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| |
Collapse
|
9
|
Shi H, Tan Z, Duan B, Guo C, Li C, Luan T, Li N, Huang Y, Chen S, Gao J, Feng W, Xu H, Wang J, Fu S, Wang H. LASS2 enhances chemosensitivity to cisplatin by inhibiting PP2A-mediated β-catenin dephosphorylation in a subset of stem-like bladder cancer cells. BMC Med 2024; 22:19. [PMID: 38191448 PMCID: PMC10775422 DOI: 10.1186/s12916-023-03243-5] [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: 04/01/2023] [Accepted: 11/01/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND The benefits of first-line, cisplatin-based chemotherapy for muscle-invasive bladder cancer are limited due to intrinsic or acquired resistance to cisplatin. Increasing evidence has revealed the implication of cancer stem cells in the development of chemoresistance. However, the underlying molecular mechanisms remain to be elucidated. This study investigates the role of LASS2, a ceramide synthase, in regulating Wnt/β-catenin signaling in a subset of stem-like bladder cancer cells and explores strategies to sensitize bladder cancer to cisplatin treatment. METHODS Data from cohorts of our center and published datasets were used to evaluate the clinical characteristics of LASS2. Flow cytometry was used to sort and analyze bladder cancer stem cells (BCSCs). Tumor sphere formation, soft agar colony formation assay, EdU assay, apoptosis analysis, cell viability, and cisplatin sensitivity assay were used to investigate the functional roles of LASS2. Immunofluorescence, immunoblotting, coimmunoprecipitation, LC-MS, PCR array, luciferase reporter assays, pathway reporter array, chromatin immunoprecipitation, gain-of-function, and loss-of-function approaches were used to investigate the underlying mechanisms. Cell- and patient-derived xenograft models were used to investigate the effect of LASS2 overexpression and a combination of XAV939 on cisplatin sensitization and tumor growth. RESULTS Patients with low expression of LASS2 have a poorer response to cisplatin-based chemotherapy. Loss of LASS2 confers a stem-like phenotype and contributes to cisplatin resistance. Overexpression of LASS2 results in inhibition of self-renewal ability of BCSCs and increased their sensitivity to cisplatin. Mechanistically, LASS2 inhibits PP2A activity and dissociates PP2A from β-catenin, preventing the dephosphorylation of β-catenin and leading to the accumulation of cytosolic phospho-β-catenin, which decreases the transcription of the downstream genes ABCC2 and CD44 in BCSCs. Overexpression of LASS2 combined with a tankyrase inhibitor (XAV939) synergistically inhibits tumor growth and restores cisplatin sensitivity. CONCLUSIONS Targeting the LASS2 and β-catenin pathways may be an effective strategy to overcome cisplatin resistance and inhibit tumor growth in bladder cancer patients.
Collapse
Affiliation(s)
- Hongjin Shi
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
- Kunming Medical University, Kunming, China
| | - Zhiyong Tan
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
- Kunming Medical University, Kunming, China
| | - Bowen Duan
- Kunming Medical University, Kunming, China
| | - Chunming Guo
- School for Life Science, Yunnan University, Kunming, China
| | - Chong Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ting Luan
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Ning Li
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Yinglong Huang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Shi Chen
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
- Kunming Medical University, Kunming, China
| | - Jixian Gao
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
- Kunming Medical University, Kunming, China
| | - Wei Feng
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
- Kunming Medical University, Kunming, China
| | - Haole Xu
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
- Kunming Medical University, Kunming, China
| | - Jiansong Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China
| | - Shi Fu
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China.
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China.
| | - Haifeng Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, China.
- Yunnan Clinical Medical Center of Urological Disease, Kunming, China.
| |
Collapse
|
10
|
Mo G, Long X, Cao L, Tang Y, Yan Y, Guo T. A Six-gene Prognostic Model Based on Neutrophil Extracellular Traps (NETs)-related Gene Signature for Lung Adenocarcinoma. Comb Chem High Throughput Screen 2024; 27:1969-1983. [PMID: 38357943 DOI: 10.2174/0113862073282003240119064337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is one of the most common malignant cancers. Neutrophil extracellular traps (NETs) have been discovered to play a crucial role in the pathogenesis of LUAD. We aimed to establish an innovative prognostic model for LUAD based on the distinct expression patterns of NETs-related genes. METHODS The TCGA LUAD dataset was utilized as the training set, while GSE31210, GSE37745, and GSE50081 were undertaken as the verification sets. The patients were grouped into clusters based on the expression signature of NETs-related genes. Differentially expressed genes between clusters were identified through the utilization of the random forest and LASSO algorithms. The NETs score model for LUAD prognosis was developed by multiplying the expression levels of specific genes with their corresponding LASSO coefficients and then summing them. The validity of the model was confirmed by analysis of the survival curves and ROC curves. Additionally, immune infiltration, GSEA, mutation analysis, and drug analysis were conducted. Silencing ABCC2 in A549 cells was achieved to investigate its effect. RESULTS We identified six novel NETs-related genes, namely UPK1B, SFTA3, GGTLC1, SCGB3A1, ABCC2, and NTS, and developed a NETs score signature, which exhibited a significant correlation with the clinicopathological and immune traits of the LUAD patients. High-risk patients showed inhibition of immune-related processes. Mutation patterns exhibited variability among the different groups. AZD3759, lapatinib, and dasatinib have been identified as potential candidates for LUAD treatment. Moreover, the downregulation of ABCC2 resulted in the induction of apoptosis and suppression of migration and invasion in A549 cells. CONCLUSION Altogether, this study has identified a novel NET-score signature based on six novel NET-related genes to predict the prognosis of LUAD and ABCC2 and has also explored a new method for personalized chemo-/immuno-therapy of LUAD.
Collapse
Affiliation(s)
- Guiyan Mo
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410005, Hunan, China
| | - Xuan Long
- Department of Respiratory and Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Limin Cao
- Department of Respiratory and Critical Care Medicine, Lianyungang Second People's Hospital, Lianyungang, 222000, Jiangsu, China
| | - Yuling Tang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410005, Hunan, China
| | - Yusheng Yan
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410005, Hunan, China
| | - Ting Guo
- Department of Respiratory and Critical Care Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410005, Hunan, China
| |
Collapse
|
11
|
Tyavambiza C, Meyer M, Wusu AD, Madiehe A, Meyer S. The Cytotoxicity of Cotyledon orbiculata Aqueous Extract and the Biogenic Silver Nanoparticles Derived from the Extract. Curr Issues Mol Biol 2023; 45:10109-10120. [PMID: 38132477 PMCID: PMC10742177 DOI: 10.3390/cimb45120631] [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: 10/16/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Green synthesized silver nanoparticles (AgNPs) have become popular because of their promising biological activities. However, for most of these nanoparticles, the cytotoxic effects have not been determined and their safety is not guaranteed. In a previous study, we successfully synthesized AgNPs (Cotyledon-AgNPs) using an extract of Cotyledon orbiculata, a medicinal plant traditionally used in South Africa to treat skin conditions. Cotyledon-AgNPs were shown to have significant antimicrobial and wound-healing activities. Fibroblast cells treated with extracts of C. orbiculata and Cotyledon-AgNPs demonstrated an enhanced growth rate, which is essential in wound healing. These nanoparticles therefore have promising wound-healing activities. However, the cytotoxicity of these nanoparticles is not known. In this study, the toxic effects of C. orbiculata extract and Cotyledon-AgNPs on the non-cancerous skin fibroblast (KMST-6) were determined using in vitro assays to assess oxidative stress and cell death. Both the C. orbiculata extract and the Cotyledon-AgNPs did not show any significant cytotoxic effects in these assays. Gene expression analysis was also used to assess the cytotoxic effects of Cotyledon-AgNPs at a molecular level. Of the eighty-four molecular toxicity genes analysed, only eight (FASN, SREBF1, CPT2, ASB1, HSPA1B, ABCC2, CASP9, and MKI67) were differentially expressed. These genes are mainly involved in fatty acid and mitochondrial energy metabolism. The results support the finding that Cotyledon-AgNPs have low cytotoxicity at the concentrations tested. The upregulation of genes such as FASN, SERBF1, and MKI-67 also support previous findings that Cotyledon-AgNPs can promote wound healing via cell growth and proliferation. It can therefore be concluded that Cotyledon-AgNPs are not toxic to skin fibroblast cells at the concentration that promotes wound healing. These nanoparticles could possibly be safely used for wound healing.
Collapse
Affiliation(s)
- Caroline Tyavambiza
- Department of Science and Innovation–Mintek Nanotechnology Innovation Centre, Department of Biotechnology, University of the Western Cape, Cape Town 7530, South Africa; (C.T.); (M.M.); (A.D.W.); (A.M.)
- Department of Biomedical Sciences, Cape Peninsula University of Technology, Cape Town 7535, South Africa
| | - Mervin Meyer
- Department of Science and Innovation–Mintek Nanotechnology Innovation Centre, Department of Biotechnology, University of the Western Cape, Cape Town 7530, South Africa; (C.T.); (M.M.); (A.D.W.); (A.M.)
| | - Adedoja Dorcas Wusu
- Department of Science and Innovation–Mintek Nanotechnology Innovation Centre, Department of Biotechnology, University of the Western Cape, Cape Town 7530, South Africa; (C.T.); (M.M.); (A.D.W.); (A.M.)
| | - Abram Madiehe
- Department of Science and Innovation–Mintek Nanotechnology Innovation Centre, Department of Biotechnology, University of the Western Cape, Cape Town 7530, South Africa; (C.T.); (M.M.); (A.D.W.); (A.M.)
- Nanobiotechnology Research Group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Cape Town 7530, South Africa
| | - Samantha Meyer
- Department of Biomedical Sciences, Cape Peninsula University of Technology, Cape Town 7535, South Africa
| |
Collapse
|
12
|
Strachowska M, Gronkowska K, Sobczak M, Grodzicka M, Michlewska S, Kołacz K, Sarkar T, Korszun J, Ionov M, Robaszkiewicz A. I-CBP112 declines overexpression of ATP-binding cassette transporters and sensitized drug-resistant MDA-MB-231 and A549 cell lines to chemotherapy drugs. Biomed Pharmacother 2023; 168:115798. [PMID: 37913733 DOI: 10.1016/j.biopha.2023.115798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Despite extensive efforts and ongoing progress in personalized anticancer approaches, chemotherapy remains the first line or the only treatment for some tumors that may develop resistance to chemotherapeutics in time due to inter alia overexpression of ATP-binding cassette transporters. Using clinically-relevant resistant models of triple negative breast cancer (MDA-MB-231; TNBC) as well as non-small cell lung cancer (A549; NSCLC), we tested the efficacy of I-CBP112 - CBP/EP300 bromodomain inhibitor to overcome drug resistance by declining ABC gene transcription. I-CBP112 significantly reduced ABCB1, ABCC1, ABCC2, ABCC3, ABCC5 and ABCG2 in all resistant lines, as well as ABCC10 in TNBC and ABCC4 in paclitaxel-resistant NSCLC, thereby increasing intracellular drug accumulation and cytotoxicity in 2D and 3D cultures. This was phenocopied only by the joint effect of ABC inhibitors such as tariquidar (ABCB1 - P-glycoprotein and ABCG2) and MK-571 (ABCC), whereas single inhibition of ABCB1/ABCG2 or ABCC proteins did not affect drug accumulation, thereby implying the need of simultaneous deficiency in activity of majority of drug pumps for enhanced drug retention. I-CBP112 failed to directly inhibit activity of ABCB1, ABCG2 and ABCC subfamily members at the same time. Importantly, I-CBP112 treated cancer cells polarized human macrophages into proinflammatory phenotypes. Moreover, I-CBP112 remained non-toxic to primary cell lines, nor did it enhance anticancer drug toxicity to blood-immune cells. In silico assay of ADMET properties confirmed the desired pharmacokinetic features of I-CBP112. The results suggest that the CBP/p300 inhibitor is a promising co-adjuvant to chemotherapy in drug-resistant cancer phenotypes, capable of decreasing ABC transporter expression.
Collapse
Affiliation(s)
- Magdalena Strachowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha St. 12/16, 90-237 Lodz, Poland.
| | - Karolina Gronkowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha St. 12/16, 90-237 Lodz, Poland
| | - Maciej Sobczak
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, Pomorska St. 251, 92-213 Lodz, Poland
| | - Marika Grodzicka
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha St. 12/16, 90-237 Lodz, Poland
| | - Sylwia Michlewska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Banacha St. 12/16, 90-237 Lodz, Poland
| | - Kinga Kołacz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha St. 12/16, 90-237 Lodz, Poland
| | - Tuhin Sarkar
- Department of Microbiology, University of Kalyani, West Bengal 741245, India
| | - Joanna Korszun
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha St. 12/16, 90-237 Lodz, Poland; Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Szaserow St. 128, 04-349 Warsaw, Poland
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland; Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, 2 Dabrowskiego Sq, 09-402, Plock, Poland
| | - Agnieszka Robaszkiewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska St.141/143, 90-236 Lodz, Poland.
| |
Collapse
|
13
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
14
|
Guan X, Pavani KC, Chunduru J, Broeckx BJG, Van Soom A, Peelman L. Hsa-miR-665 Is a Promising Biomarker in Cancer Prognosis. Cancers (Basel) 2023; 15:4915. [PMID: 37894282 PMCID: PMC10605552 DOI: 10.3390/cancers15204915] [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: 08/26/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Biomarkers are biomolecules used to identify or predict the presence of a specific disease or condition. They play an important role in early diagnosis and may be crucial for treatment. MicroRNAs (miRNAs), a group of small non-coding RNAs, are more and more regarded as promising biomarkers for several reasons. Dysregulation of miRNAs has been linked with development of several diseases, including many different types of cancer, and abnormal levels can be present in early stages of tumor development. Because miRNAs are stable molecules secreted and freely circulating in blood and urine, they can be sampled with little or no invasion. Here, we present an overview of the current literature, focusing on the types of cancers for which dysregulation of miR-665 has been associated with disease progression, recurrence, and/or prognosis. It needs to be emphasized that the role of miR-665 sometimes seems ambiguous, in the sense that it can be upregulated in one cancer type and downregulated in another and can even change during the progression of the same cancer. Caution is thus needed before using miR-665 as a biomarker, and extrapolation between different cancer types is not advisable. Moreover, more detailed understanding of the different roles of miR-665 will help in determining its potential as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Xuefeng Guan
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; (X.G.); (B.J.G.B.)
| | - Krishna Chaitanya Pavani
- Department of Internal Medicine, Reproduction and Population Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (K.C.P.); (A.V.S.)
- Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Gent, Belgium
| | - Jayendra Chunduru
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA;
| | - Bart J. G. Broeckx
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; (X.G.); (B.J.G.B.)
| | - Ann Van Soom
- Department of Internal Medicine, Reproduction and Population Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (K.C.P.); (A.V.S.)
| | - Luc Peelman
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; (X.G.); (B.J.G.B.)
| |
Collapse
|
15
|
Chen H, Liang W, Zheng W, Li F, Pan X, Lu Y. A novel telomere-related gene prognostic signature for survival and drug treatment efficiency prediction in lung adenocarcinoma. Aging (Albany NY) 2023; 15:7956-7973. [PMID: 37589509 PMCID: PMC10497012 DOI: 10.18632/aging.204877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/19/2023] [Indexed: 08/18/2023]
Abstract
OBJECTIVE Telomere-related genes (TRGs) play a critical role in various types of tumors. However, there is a lack of comprehensive exploration of their relevance in lung cancer. This research aimed to verify the relationship between TRGs gene expression and the prognosis of patients with lung adenocarcinoma (LUAD), as well as the prediction of drug treatment efficiency. METHODS A total of 2093 TRGs were acquired from TelNet. The clinical information including age, tumor stage, follow up and outcome (death/survival) and TRGs expression profile of LUAD were obtained from the patients in The Cancer Genome Atlas (TCGA) database and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. The two databases were used to construct and verify a prognostic model based on the expression of hubTRGs. The tumor mutation burden, immune infiltration and subtypes, as well as IC50 prediction of multiple targeted drugs were also evaluated in TRGs-divided risk groups. RESULTS A total of 335 TRGs were significantly differentially expressed in LUAD as compared with normal control. Among them, 9 TRGs (ABCC2, ABCC8, ALDH2, FOXP3, GNMT, JSRP1, MACF1, PLCD3, SULT4A1) were finally identified as hubGenes and used to construct a TRG risk score. The TRG risk score showed favorable performance in constructing a prognostic nomogram in predicting survival of LUAD, and the ROC curves at 1, 3 and 5 years were plotted and the AUROC values were 0.743, 0.754 and 0.735, respectively. Higher TRGs risk score correlated with worse immune subtypes and higher tumor mutation burden in LUAD tissues. In addition, the patients in TRG high risk group harbored a lower TIDE score which indicated potentially better response to immunotherapy. CONCLUSION This study proposed a broad molecular signature of telomere-related genes that can be used in further functional and therapeutic investigations, and also represents an integrated modality for characterizing critical molecules when exploring novel targets for lung cancer immunotherapy.
Collapse
Affiliation(s)
- Haiming Chen
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong Province 528200, China
| | - Weiquan Liang
- Department of Respiration, Foshan Second People's Hospital, Foshan, Guangdong Province 528000, China
| | - Weiqiang Zheng
- Department of Respiration, Foshan Second People's Hospital, Foshan, Guangdong Province 528000, China
| | - Feilong Li
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong Province 528200, China
| | - Xingxi Pan
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong Province 528200, China
| | - Yiyu Lu
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong Province 528200, China
| |
Collapse
|
16
|
Eisner JR, Mayhew GM, Davison JM, Beebe KD, Shibata Y, Guo Y, Farhangfar C, Farhangfar F, Uronis JM, Conroy JM, Milburn MV, Hayes DN, Mileham KF. Association of Antifolate Response Signature Status and Clinical Activity of Pemetrexed-Platinum Chemotherapy in Non-Small Cell Lung Cancer: The Piedmont Study. Clin Cancer Res 2023; 29:3203-3213. [PMID: 37233991 PMCID: PMC10425722 DOI: 10.1158/1078-0432.ccr-22-2558] [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: 08/19/2022] [Revised: 12/13/2022] [Accepted: 05/24/2023] [Indexed: 05/27/2023]
Abstract
PURPOSE The Piedmont study is a prospectively designed retrospective evaluation of a new 48-gene antifolate response signature (AF-PRS) in patients with locally advanced/metastatic nonsquamous (NS) non-small cell lung cancer (NSCLC) treated with pemetrexed-containing platinum doublet chemotherapy (PMX-PDC). The study tested the hypothesis that AF-PRS identifies patients with NS-NSCLC who have a higher likelihood of responding positively to PMX-PDC. The goal was to gather clinical evidence supporting AF-PRS as a potential diagnostic test. EXPERIMENTAL DESIGN Residual pretreatment FFPE tumor samples and clinical data were analyzed from 105 patients treated with first-line (1L) PMX-PDC. Ninety-five patients had sufficient RNA sequencing (RNA-seq) data quality and clinical annotation for inclusion in the analysis. Associations between AF-PRS status and associate genes and outcome measures including progression-free survival (PFS) and clinical response were evaluated. RESULTS Overall, 53% of patients were AF-PRS(+), which was associated with extended PFS, but not overall survival, versus AF-PRS(-) (16.6 months vs. 6.6 months; P = 0.025). In patients who were stage I to III patients at the time of treatment, PFS was further extended in AF-PRS(+) versus AF-PRS(-) (36.2 months vs. 9.3 months; P = 0.03). Complete response (CR) to therapy was noted in 14 of 95 patients. AF-PRS(+) preferentially selected a majority (79%) of CRs, which were evenly split between patients stage I to III (six of seven) and stage IV (five of seven) at the time of treatment. CONCLUSIONS AF-PRS identified a significant population of patients with extended PFS and/or clinical response following PMX-PDC treatment. AF-PRS may be a useful diagnostic test for patients indicated for systemic chemotherapy, especially when determining the optimal PDC regimen for locally advanced disease.
Collapse
Affiliation(s)
| | | | | | - Kirk D. Beebe
- GeneCentric Therapeutics, Inc., Durham, North Carolina
| | | | - Yuelong Guo
- GeneCentric Therapeutics, Inc., Durham, North Carolina
| | - Carol Farhangfar
- Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | | | | | | | | | - David Neil Hayes
- University of Tennessee Health Science Center, Center for Cancer Research, Memphis, Tennessee
| | | |
Collapse
|
17
|
YÜCE M, GÜMÜŞKAPTAN Ç, ÇON AH, YAZICI F. Conjugated Linoleic Acid strengthens the apoptotic effect of low-dose cisplatin in A549 cells by inducing Bcl-2 downregulation. Prostaglandins Other Lipid Mediat 2023; 166:106731. [PMID: 37001725 DOI: 10.1016/j.prostaglandins.2023.106731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
One of the chemotherapeutic agents widely used in the treatment of non-small cell lung cancer (NSCLC) is cisplatin. However, the resistance of cancer cells to cisplatin and additionally serious side effects from cisplatin limit its use. Conjugated linoleic acid (CLA) has been shown to suppress the development of carcinogenesis in vitro and in vivo studies and has antitumoral activity in many cancers. The study aimed to investigate the potential effect of using cisplatin, the first-line treatment for NSCLC, in combination with CLA to increase its efficacy in low-dose use. MTT cytotoxicity assay was performed to determine the effects of CLA in combination with cisplatin on cell viability of NSCLC cell lines. The apoptotic effect of this combination on NSCLC cell lines and cell cycle distribution was determined by flow cytometry. At the same time, apoptosis and cell cycle-related gene expression levels were determined by Real-Time PCR. Combination treatment of low-dose cisplatin with CLA resulted in a significant decrease in cell viability compared to cisplatin alone, and an increase in the rate of apoptotic cells was observed. While cisplatin caused G1 phase arrest in cancer cells, there was an increase in cell percentages in S and G2 phases after combined application with CLA. In high-dose cisplatin administration, it was observed that the efficiency of the decrease in anti-apoptotic BCL2 expression related to resistance to chemotherapeutic agents was less than that of low-dose cisplatin administration. Combined administration of high-dose cisplatin with CLA significantly recovered BCL2 downregulation.
Collapse
|
18
|
Selim MS, Kassem AB, El-Bassiouny NA, Salahuddin A, Abu El-Ela RY, Hamza MS. Polymorphic renal transporters and cisplatin's toxicity in urinary bladder cancer patients: current perspectives and future directions. Med Oncol 2023; 40:80. [PMID: 36650399 PMCID: PMC9845168 DOI: 10.1007/s12032-022-01928-0] [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: 10/28/2022] [Accepted: 12/10/2022] [Indexed: 01/19/2023]
Abstract
Urinary bladder cancer (UBC) holds a potentially profound social burden and affects over 573,278 new cases annually. The disease's primary risk factors include occupational tobacco smoke exposure and inherited genetic susceptibility. Over the past 30 years, a number of treatment modalities have emerged, including cisplatin, a platinum molecule that has demonstrated effectiveness against UBC. Nevertheless, it has severe dose-limiting side effects, such as nephrotoxicity, among others. Since intracellular accumulation of platinum anticancer drugs is necessary for cytotoxicity, decreased uptake or enhanced efflux are the root causes of platinum resistance and response failure. Evidence suggests that genetic variations in any transporter involved in the entry or efflux of platinum drugs alter their kinetics and, to a significant extent, determine patients' responses to them. This review aims to consolidate and describe the major transporters and their polymorphic variants in relation to cisplatin-induced toxicities and resistance in UBC patients. We concluded that the efflux transporters ABCB1, ABCC2, SLC25A21, ATP7A, and the uptake transporter OCT2, as well as the organic anion uptake transporters OAT1 and OAT2, are linked to cisplatin accumulation, toxicity, and resistance in urinary bladder cancer patients. While suppressing the CTR1 gene's expression reduced cisplatin-induced nephrotoxicity and ototoxicity, inhibiting the expression of the MATE1 and MATE2-K genes has been shown to increase cisplatin's nephrotoxicity and resistance. The roles of ABCC5, ABCA8, ABCC10, ABCB10, ABCG1, ATP7B, ABCG2, and mitochondrial SLC25A10 in platinum-receiving urinary bladder cancer patients should be the subject of further investigation.
Collapse
Affiliation(s)
- Mohamed S Selim
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| | - Amira B Kassem
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Noha A El-Bassiouny
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Ahmad Salahuddin
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
- Biochemistry Department, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Raghda Y Abu El-Ela
- Medical Oncology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Marwa Samir Hamza
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
19
|
Chandra F, Tania TF, Nurcahyanti ADR. Bixin and Fuxoxanthin Alone and in Combination with Cisplatin Regulate ABCC1 and ABCC2 Transcription in A549 Lung Cancer Cells. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2023; 15:15-20. [PMID: 37313537 PMCID: PMC10259734 DOI: 10.4103/jpbs.jpbs_50_23] [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: 01/20/2023] [Revised: 02/04/2023] [Accepted: 02/16/2023] [Indexed: 06/15/2023] Open
Abstract
Background The ATP-Binding Cassette (ABC) transporter has long been studied to confer drug resistance in human tumors and play important role in metabolic processes and cellular signaling. The overexpression of ABCB1, ABCC1, ABCC2, ABCC3, and ABCG2 leads to decreased sensitivity of lung cancer to cisplatin. At the transcription level, the expression of ABC transporters is highly regulated and requires the complex interplay of factors involved in differentiation and development, cell survival and apoptosis upon intrinsic and environmental stress. The p53 regulation of drug-resistance genes is also complex yet not well understood. Previously, we demonstrated the synergistic interaction between bixin or fucoxanthin with cisplatin in A549 lung cancer cells. Objectives Current study aims to identify whether carotenoids enhancing therapeutic effect of Cisplatin due to the ability to reverse drug resistance associated proteins, such as ABC transporter and regulating the tumor suppressor corresponding gene, p53. Methods Real-Time Quantitative-Polymerase Chain Reaction (RT-qPCR) was performed to estimate the expression level of ABCC1 and ABCC2, and p53 of A549 cell lines in response to carotenoids alone and in combination with cisplatin. Results and Conclusion The administration of bixin or fucoxanthin decreases the expression of ABCC1 and ABCC2. Both carotenoids, either alone or in combination with cisplatin, upregulated p53 gene expression indicating the mechanism of proliferation inhibition and apoptosis occurs via the p53 caspase-independent pathway.
Collapse
Affiliation(s)
- Ferdy Chandra
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Teresa F. Tania
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Agustina D. R. Nurcahyanti
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| |
Collapse
|
20
|
Ding P, Gao Y, Wang J, Xiang H, Zhang C, Wang L, Ji G, Wu T. Progress and challenges of multidrug resistance proteins in diseases. Am J Cancer Res 2022; 12:4483-4501. [PMID: 36381332 PMCID: PMC9641395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/09/2022] [Indexed: 06/16/2023] Open
Abstract
Chemotherapy remains the first choice for patients with advanced cancers when other treatments are ineffective. Multidrug resistance (MDR) is an unavoidable factor that negatively affects the effectiveness of cancer chemotherapy drugs. Researchers are trying to reduce MDR, improve the effectiveness of chemotherapeutic drugs, and alleviate patient suffering to positively contribute to disease treatment. MDR also occurs in inflammation and genetic disorders, which increases the difficulty of clinically beneficial treatments. The ATP-binding cassette (ABC) is an active transporter that plays an important role in the barrier and secretory functions of many normal cells. As the C subfamily in the ABC family, multidrug resistance proteins (MRPs/ABCCs) export a variety of antitumour drugs and are expressed in a variety of cancers. The present review summarises the role of MRPs in cancer and other diseases and recent research progress of MRP inhibitors to better examine the mechanism and function of MRPs, and establish a good relationship with clinical treatment.
Collapse
Affiliation(s)
- Peilun Ding
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Ying Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Caiyun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| |
Collapse
|
21
|
Ma C, Li F, He Z, Zhao S. A more novel and powerful prognostic gene signature of lung adenocarcinoma determined from the immune cell infiltration landscape. Front Surg 2022; 9:1015263. [PMID: 36311939 PMCID: PMC9606711 DOI: 10.3389/fsurg.2022.1015263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the leading histological subtype of lung cancer worldwide, causing high mortality each year. The tumor immune cell infiltration (ICI) is closely associated with clinical outcome with LUAD patients. The present study was designed to construct a gene signature based on the ICI of LUAD to predict prognosis. Methods Downloaded the raw data of three cohorts of the TCGA-LUAD, GSE72094, and GSE68465 and treat them as training cohort, validation cohort one, and validation cohort two for this research. Unsupervised clustering detailed grouped LUAD cases of the training cohort based on the ICI profile. The univariate Cox regression and Kaplan-Meier was adopted to identify potential prognostic genes from the differentially expressed genes recognized from the ICI clusters. A risk score-based prognostic signature was subsequently developed using LASSO-penalized Cox regression analysis. The Kaplan-Meier analysis, Cox analysis, ROC, IAUC, and IBS were constructed to assess the ability to predict the prognosis and effects of clinical variables in another two independent validation cohorts. More innovatively, we searched similar papers in the most recent year and made comprehensive comparisons with ours. GSEA was used to discover the related signaling pathway. The immune relevant signature correlation identification and immune infiltrating analysis were used to evaluate the potential role of the signature for immunotherapy and recognize the critical immune cell that can influence the signature's prognosis capability. Results A signature composed of thirteen gene including ABCC2, CCR2, CERS4, CMAHP, DENND1C, ECT2, FKBP4, GJB3, GNG7, KRT6A, PCDH7, PLK1, and VEGFC, was identified as significantly associated with the prognosis in LUAD patients. The thirteen-gene signature exhibited independence in evaluating the prognosis of LUAD patients in our training and validation cohorts. Compared to our predecessors, our model has an advantage in predictive power. Nine well know immunotherapy targets, including TBX2, TNF, CTLA4, HAVCR2, GZMB, CD8A, PRF1, GZMA, and PDCD1 were recognized correlating with our signature. The mast cells were found to play vital parts in backing on the thirteen-gene signature's outcome predictive capacity. Conclusions Collectively, the current study indicated a robust thirteen-gene signature that can accurately predict LUAD prognosis, which is superior to our predecessors in predictive ability. The immune relevant signatures, TBX2, TNF, CTLA4, HAVCR2, GZMB, CD8A, PRF1, GZMA, PDCD1, and mast cells infiltrating were found closely correlate with the thirteen-gene signature's power.
Collapse
|
22
|
Correlation of Tryptophan Metabolic Pathway with Immune Activation and Chemosensitivity in Patients with Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:2158525. [PMID: 36185621 PMCID: PMC9520315 DOI: 10.1155/2022/2158525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
Lung adenocarcinoma (LUAD) is the most common type of lung cancer with high malignancy and easy metastasis in the early stage. In this study, we aimed to figure out the role of tryptophan metabolic pathway in LUAD prognosis and treatment. Different molecular subtypes were constructed based on tryptophan metabolism-related genes. Significant prognostic genes and clinical prognostic characteristics, immune infiltration level, and pathway activity in different subtypes were determined by algorithms, such as the least absolute shrinkage and selection operator (Lasso), CIBERSORT, Tumor Immune Dysfunction and Exclusion (TIDE), and gene set enrichment analysis (GSEA). The effect of different gene mutation types on the prognosis of patients with LUAD was explored. The clinical prognosis model was constructed and its reliability was verified. Of the 40 genes in the tryptophan metabolism pathway, 13 had significant prognostic significance. Based on these 13 genes, three molecular subtypes (C1, C2, and C3) were established. Among them, C1 had the worst prognosis and the lowest enrichment score of tryptophan metabolism. At the same time, C1 had the most genetic variation, the highest level of immune infiltration, and significantly activated pathways related to tumor development. The high-risk and low-risk groups had significant differences in prognosis, immune infiltration and pathway enrichment, which was consistent with the results of subtype analysis. Mutation in tryptophan metabolism-related genes leads to abnormal tryptophan metabolism, immune deficiency, and activation of cancer-promoting pathways. This results in high malignancy, poor prognosis, and failure of traditional clinical treatments. Through the establishment of risk score (RS) clinical prognosis model, we determined that RS could reliably predict the prognosis of patients with LUAD.
Collapse
|
23
|
He J, Wang Z, Zou T, Wang Y, Li XP, Chen J. The Association Between Genetic Polymorphisms of Transporter Genes and Prognosis of Platinum-Based Chemotherapy in Lung Cancer Patients. Pharmgenomics Pers Med 2022; 15:817-825. [PMID: 36131844 PMCID: PMC9484078 DOI: 10.2147/pgpm.s375284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Objective Platinum-based chemotherapy is the first-line treatment of lung cancer. However, different individual and genetic variation effect therapy for lung cancer. The purpose of this study was to evaluate the association between transport genes genetic polymorphisms and the prognosis of platinum-based chemotherapy in lung cancer patients. Methods A series of 593 patients with treatment of platinum-based chemotherapy were recruited for this study. A total of 21 single-nucleotide polymorphisms in nine transporter genes were selected to investigate their associations with platinum-based chemotherapy prognosis. Results Patients with ABCG2 rs1448784 CC genotype had a significantly shorter PFS than CT or TT genotypes (Additive model: HR = 1.54, 95% CI = 1.02–2.35, P = 0.040). In stratification analysis, SLC22A2 rs316003, SLC2A1 rs4658 were related to PFS and AQP9 rs1867380, SLC2A1 rs3820589, SLC22A2 rs316003 indicated were related to OS of platinum-based chemotherapy prognosis. Conclusion Genetic polymorphisms of rs1448784 in ABCG2 might be potential clinical marker for predicting the prognosis of lung cancer patients treated with platinum-based chemotherapy.
Collapse
Affiliation(s)
- Jia He
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Zhan Wang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, People’s Republic of China
| | - Ting Zou
- National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Ying Wang
- Hunan clinical Research Center in Gynecologic Cancer, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, 410013, People’s Republic of China
| | - Xiang-Ping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Juan Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Correspondence: Juan Chen, Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China, Tel +86-731-89753491, Email
| |
Collapse
|
24
|
Zhou R, Ma HC, Liu YH, Chen X, Chang XS, Chen YD, Liu LR, Li Y, Zhu YJ, Zhang HB. Dissecting the ferroptosis-related prognostic biomarker and immune microenvironment of driver gene-negative lung cancer. Exp Biol Med (Maywood) 2022; 247:1447-1465. [PMID: 35762414 PMCID: PMC9493762 DOI: 10.1177/15353702221102872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Despite significant advances in targeted and immune therapy for non-small cell lung cancer (NSCLC), effective therapies for wild-type epidermal growth factor receptor/anaplastic lymphoma kinase (EGFR/ALKWT) with low expression of programmed death ligand-1 (PD-L1) NSCLC remain elusive. Numerous studies have shown that ferroptosis plays an essential role in antitumor activity. To identify the molecular regulation patterns associated with ferroptosis, 351 EGFR/ALKWT NSCLC samples with low-level PD-L1 were extracted from The Cancer Genome Atlas (TCGA) and clustered using the k-means clustering technique. The two clusters associated with ferroptosis showed significantly different prognoses. In total, 169 differential expression genes (DEGs) were identified. Cluster differential analysis revealed that Cluster 1 had a significantly poorer overall survival (OS) and was associated with more negative immune regulation. In addition, TCGA samples were randomly assigned in a 7:3 ratio to a training group or testing group. A signature of eight genes associated with ferroptosis was established in the training cohort using DEGs and validated in the test cohort and three independent cohorts (GSE72049, GSE41271, and GSE50081). The 5-year area under the curve (AUC) was 0.713, which was significantly higher than that of other predictors, including TNM stage and age. Furthermore, the risk score was associated with immune function, immune infiltration, and immunotherapy response, with high-risk patients having a worse prognosis, an immune-suppressing phenotype, and a poor response to immune checkpoint inhibitors. This study aims to contribute to our understanding of the biological role of ferroptosis in EGFR/ALKWT NSCLC with low-level PD-L1, laying the groundwork for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Rui Zhou
- The Second Clinical Medical School of
Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hao-chuan Ma
- The Second Clinical Medical School of
Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yi-hong Liu
- Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Xian Chen
- The Second Clinical Medical School of
Guangzhou University of Chinese Medicine, Guangzhou 510405, China,Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Xue-song Chang
- Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Ya-dong Chen
- Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Li-rong Liu
- Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Yong Li
- Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Yan-juan Zhu
- The Second Clinical Medical School of
Guangzhou University of Chinese Medicine, Guangzhou 510405, China,Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China,Guangdong-Hong Kong-Macau Joint Lab on
Chinese Medicine and Immune Disease Research, Guangzhou 510120, China,Guangdong Provincial Key Laboratory of
Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120,
China,Yan-juan Zhu.
| | - Hai-bo Zhang
- Department of Oncology, The Second
Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong
Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China,Guangdong-Hong Kong-Macau Joint Lab on
Chinese Medicine and Immune Disease Research, Guangzhou 510120, China,Guangdong Provincial Key Laboratory of
Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120,
China,State Key Laboratory of Dampness
Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| |
Collapse
|
25
|
Zhang Z, Xiahou Z, Wu W, Song Y. Nitrogen Metabolism Disorder Accelerates Occurrence and Development of Lung Adenocarcinoma: A Bioinformatic Analysis and In Vitro Experiments. Front Oncol 2022; 12:916777. [PMID: 35903696 PMCID: PMC9315097 DOI: 10.3389/fonc.2022.916777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022] Open
Abstract
Background Nitrogen metabolism (NM) plays a pivotal role in immune regulation and the occurrence and development of cancers. The aim of this study was to construct a prognostic model and nomogram using NM-related genes for the evaluation of patients with lung adenocarcinoma (LUAD). Methods The differentially expressed genes (DEGs) related to NM were acquired from The Cancer Genome Atlas (TCGA) database. Consistent clustering analysis was used to divide them into different modules, and differentially expressed genes and survival analysis were performed. The survival information of patients was combined with the expressing levels of NM-related genes that extracted from TCGA and Gene Expression Omnibus (GEO) databases. Subsequently, univariate Cox analysis and the least absolute shrinkage and selection operator (LASSO) regression were used to build a prognostic model. GO and KEGG analysis were elaborated in relation with the mechanisms of NM disorder (NMD). Meanwhile, immune cells and immune functions related to NMD were discussed. A nomogram was built according to the univariate and multivariate Cox analysis to identify independent risk factors. Finally, real-time fluorescent quantitative PCR (RT-PCR) and Western bolt (WB) were used to verify the expression level of hub genes. Results There were 138 differential NM-related genes that were divided into two gene modules. Sixteen NM-related genes were used to build a prognostic model and the receiver operating characteristic curve (ROC) showed that the efficiency was reliable. GO and KEGG analysis suggested that NMD accelerated development of LUAD through the Wnt signaling pathway. The level of activated dendritic cells (aDCs) and type II interferon response in the low-risk group was higher than that of the high-risk group. A nomogram was constructed based on ABCC2, HMGA2, and TN stages, which was identified as four independent risk factors. Finally, RT-PCR and WB showed that CDH17, IGF2BP1, IGFBP1, ABCC2, and HMGA2 were differently expressed between human lung fibroblast (HLF) cells and cancer cells. Conclusions High NM levels were revealed as a poor prognosis of LUAD. NMD regulates immune system through affecting aDCs and type II interferon response. The prognostic model with NM-related genes could be used to effectively evaluate the outcomes of patients.
Collapse
Affiliation(s)
- Zexin Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Wenfeng Wu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafeng Song
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- *Correspondence: Yafeng Song,
| |
Collapse
|
26
|
Jiang M, Zhang Z, Li W, Man X, Sun H, Liang H, Yang F. Developing a Copper(II) Agent Based on His-146 and His-242 Residues of Human Serum Albumin Nanoparticles: Integration To Overcome Cisplatin Resistance and Inhibit the Metastasis of Nonsmall Cell Lung Cancer. J Med Chem 2022; 65:9447-9458. [PMID: 35786921 DOI: 10.1021/acs.jmedchem.2c00698] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To overcome the resistance of nonsmall cell lung cancer (NSCLC) cells to cisplatin and inhibit their metastasis, we proposed to develop a Cu(II) agent based on the specific residue(s) of HSA nanoparticles (NPs) for multitargeting the tumor microenvironment (TME). To this end, we not only synthesized four Cu(II) 2-hydroxy-3-methoxybenzaldehyde thiosemicarbazone compounds (C1-C4), obtaining a Cu compound (C4) with significant cytotoxicity by studying their structure-activity relationships, but also revealed the binding mechanism of C4 to HSA through X-ray crystallography and confirmed the successful construction of a new HSA-C4 NPs delivery system. C4 and HSA-C4 NPs inhibited the A549cisR tumor growth and metastasis, and HSA NPs optimized the anticancer behavior of C4. We further confirmed the anticancer mechanism of the C4/HSA-C4 NP multitargeting TME to overcome cisplatin resistance: killing tumor cells by acting on the mtDNA and inducing apoptosis, polarizing M2-type macrophages to the M1-type, and inhibiting angiogenesis.
Collapse
Affiliation(s)
- Ming Jiang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China.,School of Food and Biochemical Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Zhenlei Zhang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Wenjuan Li
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Xueyu Man
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hong Liang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
27
|
Huang W, Luo J, Wen J, Jiang M. The Relationship Between Systemic Immune Inflammatory Index and Prognosis of Patients With Non-Small Cell Lung Cancer: A Meta-Analysis and Systematic Review. Front Surg 2022; 9:898304. [PMID: 35846963 PMCID: PMC9280894 DOI: 10.3389/fsurg.2022.898304] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/03/2022] [Indexed: 12/25/2022] Open
Abstract
Background The relationship between systemic immune inflammation index (SII) and the prognosis of cancer has always been a subject of intense interest. However, the prognostic value of SII in non-small cell lung cancer (NSCLC) patients remains a controversial topic. Objective To evaluate the effect of SII index on prognosis of NSCLC. Methods We conducted a comprehensive search of PubMed, EMBASE, and the Cochrane Library databases to determine correlation between SII index, clinicopathological features, overall survival (OS), and progression-free survival (PFS). Odds ratio (ORs) and 95% confidence interval (CIs) were used to assess the connection between SII and clinicopathological parameters, and HRs and 95% CIs were used to assess the connection between SII and survival. Results Seventeen studies with 8,877 cases were included in the analysis. Compared with NSCLC patients with low SII level, patients with NSCLC with high SII level had a poor OS (HR = 1.75, 95% CI, 1.50–2.00; P < 0.001) and had a poor PFS (HR = 1.61, 95% CI, 1.25–1.96; P < 0.001). In addition, patients with higher pathological stage (II–III) had higher SII levels (OR = 2.32, 95% CI, 2.06–2.62; P < 0.001). Conclusions The SII index is a promising prognostic biomarker for NSCLC and may help clinicians choose appropriate NSCLC treatments.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cardiothoracic Surgery, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
- Correspondence: Wei Huang
| | - Jiayu Luo
- Department of Oncology, No.906 Hospital of People’s Liberation Army, Ningbo, China
| | - Jianbo Wen
- Department of Cardiothoracic Surgery, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| | - Mingjun Jiang
- Department of Cardiothoracic Surgery, The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
28
|
Gamal-Eldeen AM, Alrehaili AA, Alharthi A, Raafat BM. Perftoran® Inhibits Hypoxia-Associated Resistance in Lung Cancer Cells to Carboplatin. Front Pharmacol 2022; 13:860898. [PMID: 35401227 PMCID: PMC8987772 DOI: 10.3389/fphar.2022.860898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/24/2022] [Indexed: 11/29/2022] Open
Abstract
Perftoran® (perfluorodecalin) is an oxygen carrier, and carboplatin is a common chemotherapy drug used worldwide for lung cancer treatment. Hypoxia is one of the factors that induce resistance of lung cancer cells to carboplatin. This study explored the role of Perftoran®, as an oxygen carrier, in lowering the resistance of lung cancer cells to carboplatin through suppression of hypoxia pathway mediators. The effect of Perftoran® on the resistance of human lung cancer A549 cells to carboplatin was investigated through the evaluation of cytotoxicity by MTT, cell death mode by dual DNA staining, DNA damage by comet assay, DNA platination (DNA/carboplatin adducts) by atomic absorption spectroscopy, hypoxia degree by pimonidazole, HIF-1α/HIF-2α concentrations by ELISA, expression of miRNAs (hypoxamiRs miR-210, miR-21, and miR-181a) by qRT-PCR, and the content of drug resistance transporter MRP-2 by immunocytochemical staining. Results indicated that compared to carboplatin, Perftoran®/carboplatin decreased cell resistance to carboplatin by potentiating its cytotoxicity using only 45% of carboplatin IC50 and inducing apoptosis. Perftoran® induced DNA platination and DNA damage index in cells compared to carboplatin alone. Moreover, compared to treatment with carboplatin alone, co-treatment of cells with Perftoran® and carboplatin inhibited cellular pimonidazole hypoxia adducts, diminished HIF-1α/HIF-2α concentrations, suppressed hypoxamiR expression, and decreased MRP-2. In conclusion, Perftoran® inhibited resistance of lung cancer cells to carboplatin through the inhibition of both hypoxia pathway mediators and the drug resistance transporter MRP-2 and through the induction of DNA/carboplatin adduct formation.
Collapse
Affiliation(s)
- Amira M. Gamal-Eldeen
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- *Correspondence: Amira M. Gamal-Eldeen,
| | - Amani A. Alrehaili
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Afaf Alharthi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Bassem M. Raafat
- Radiological Sciences Department, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| |
Collapse
|
29
|
Nurcahyanti ADR, Kusmita L, Wink M. Bixin and fucoxanthin sensitize human lung cancer and cervical cancer cell to cisplatin in vitro. BMC Res Notes 2021; 14:454. [PMID: 34922615 PMCID: PMC8684137 DOI: 10.1186/s13104-021-05866-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
Objective Cisplatin is a conventional anticancer drug that generates reactive oxygen species and causes apoptosis. However, many cancer cells develop alterations in the ATP binding cassette transporter responsible for the uptake and efflux process, which leads to resistance. Many natural products have shown potential to compete with ATP binding cassette transporter and may sensitize resistant cells to cisplatin. Studies have shown pro-oxidant effect of carotenoids that promote apoptosis of cancer cells. Bixin and fucoxanthin are well-known carotenoids with known antioxidant properties, however their bioactivity in lung cancer cells, clinically known to develop resistance due to ATP binding cassette transporter, has been minimally studied. This study is the first to investigate the potential of bixin and fucoxanthin to sensitize human lung cancer cell line, A549 and cervical cancer cell line, HeLa, to cisplatin. Drug combination method developed by Chou and Talalay theorem was employed. Result Employing the best combination ratio, this study shows selective sensitization of cancer cells to cisplatin after bixin and fucoxanthin treatment. Further study on the mechanism of action in specific types of cancer cells is warranted. It may improve cisplatin sensitivity in tumors and rational use of cancer drugs. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13104-021-05866-4.
Collapse
Affiliation(s)
- Agustina Dwi Retno Nurcahyanti
- Department of Pharmacy, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Pluit Raya 2, Jakarta, 14440, Indonesia.
| | - Lia Kusmita
- Department of Pharmacy, STIFAR Yayasan Pharmasi Semarang, Letjend Sarwo Edhie Wibowo KM 1, Plamongansari Pucanggading, Semarang, Indonesia
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| |
Collapse
|
30
|
Targeting Drug Chemo-Resistance in Cancer Using Natural Products. Biomedicines 2021; 9:biomedicines9101353. [PMID: 34680470 PMCID: PMC8533186 DOI: 10.3390/biomedicines9101353] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is one of the leading causes of death globally. The development of drug resistance is the main contributor to cancer-related mortality. Cancer cells exploit multiple mechanisms to reduce the therapeutic effects of anticancer drugs, thereby causing chemotherapy failure. Natural products are accessible, inexpensive, and less toxic sources of chemotherapeutic agents. Additionally, they have multiple mechanisms of action to inhibit various targets involved in the development of drug resistance. In this review, we have summarized the basic research and clinical applications of natural products as possible inhibitors for drug resistance in cancer. The molecular targets and the mechanisms of action of each natural product are also explained. Diverse drug resistance biomarkers were sensitive to natural products. P-glycoprotein and breast cancer resistance protein can be targeted by a large number of natural products. On the other hand, protein kinase C and topoisomerases were less sensitive to most of the studied natural products. The studies discussed in this review will provide a solid ground for scientists to explore the possible use of natural products in combination anticancer therapies to overcome drug resistance by targeting multiple drug resistance mechanisms.
Collapse
|
31
|
Pasquariello KZ, Dey JM, Sprowl JA. Current Understanding of Membrane Transporters as Regulators or Targets for Cisplatin-Induced Hearing Loss. Mol Pharmacol 2021; 100:348-355. [PMID: 34330821 DOI: 10.1124/molpharm.121.000274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/22/2021] [Indexed: 11/22/2022] Open
Abstract
Cisplatin is a platinum-based drug which remains among the most efficacious anticancer treatment options. Unfortunately, use of cisplatin is hindered by dose-limiting toxicities, including irreversible hearing loss, which can grossly affect patient quality of life. Cisplatin-induced ototoxicity is the result of cochlear hair cell damage through a mechanism that is poorly understood. However, cisplatin cytotoxicity is reliant on intracellular accumulation, a process that is largely dependent on the presence of particular membrane transporters. This review will provide an update on our current understanding of the various transporters known to be involved in the disposition and cytotoxicity of platinum drugs or their metabolites, as well as their role in mediating cisplatin-induced hearing loss. We also provide a summary of the successes and opportunities in therapeutically targeting membrane transporters to alleviate platinum-induced hearing loss. Moreover, we describe how this approach could be used to reduce the severity or onset of other adverse events associated with exposure to various forms of platinum drugs, without diminishing anti-tumor efficacy. Significance Statement Cisplatin-induced hearing loss is a dose limiting and irreversible adverse event with no current preventative or curative treatment measures. Pharmacological targeting of membrane transporters that regulate platinum uptake into cochlear hair cells, if conducted appropriately, may alleviate this devastating side effect and could be applied to alleviate other platinum-induced toxicities.
Collapse
Key Words
- Uptake transporters (OATP, OAT, OCT, PEPT, MCT, NTCP, ASBT, etc.)
- cancer chemotherapy
- efflux transporters (P-gp, BCRP, MRP, MATE, BSEP, etc)
- ototoxicity
Collapse
Affiliation(s)
| | | | - Jason A Sprowl
- School of Pharmacy, University of Buffalo, United States
| |
Collapse
|
32
|
Chen Y, Zhou H, Yang S, Su D. Increased ABCC2 expression predicts cisplatin resistance in non-small cell lung cancer. Cell Biochem Funct 2020; 39:277-286. [PMID: 32815556 PMCID: PMC7983913 DOI: 10.1002/cbf.3577] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/26/2020] [Accepted: 07/05/2020] [Indexed: 12/23/2022]
Abstract
Long-term use of platinum-based drugs can cause non-small cell lung cancer (NSCLC) to develop extremely strong drug resistance. Increasing the drug dosage does not have better treatment effects and could lead to serious complications. High levels of drug resistance are considered to be characteristic of human tumours and are usually mediated by genes related to multidrug resistance. Multidrug resistance-associated protein 2 (ABCC2), an ATP-binding cassette multidrug resistance transporter, was found to be overexpressed in various human cancers. In this study, we found that ABCC2 was also upregulated in cisplatin (DDP)-resistant A549 cells (A549/DDP). Functional studies demonstrated that ABCC2 knockdown reversed DDP resistance and promoted G1 phase arrest in A549/DDP cells, and PARP and caspase-3 were activated in A549/DDP cells following ABCC2 knockdown. In vivo, ABCC2 knockdown enhanced the cytotoxicity of DDP to subcutaneous A549 tumours. Together, these results suggest that ABCC2 may be a potential therapeutic strategy for overcoming DDP resistance in NSCLC patients. SIGNIFICANCE OF THE STUDY: In this study, we investigated the role of ABCC2 in cisplatin resistance of NSCLC cells. Our data show that ABCC2 expression was associated with resistance to cisplatin and that knockdown ABCC2 could reverse cisplatin resistance in NSCLC cells. Taken together, our study suggests that reducing the expression of ABCC2 could become an important strategy for enhancing the sensitivity of NSCLC cells to cisplatin.
Collapse
Affiliation(s)
- Yun Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Hongying Zhou
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Sifu Yang
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Dan Su
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|