1
|
Li J, Shen X, Sun C, Hou Y, Hu Y, Ma S, Huang L, Ma L, Zhang Y, Dai X. Isogarcinol inhibits nasopharyngeal carcinoma growth through mitochondria-mediated autophagic cell death. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155745. [PMID: 38833788 DOI: 10.1016/j.phymed.2024.155745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/27/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND AND AIMS Isogarcinol, a natural compound extracted from the fruits of Garcinia oblongifolia, has potential chemopreventive activity. This study aimed to elucidate the anti-tumor effects and mechanism of action of isogarcinol on nasopharyngeal carcinoma (NPC). METHODS Isogarcinol was isolated from Garcinia oblongifolia by using chromatographic separation. The anti-tumor effects of isogarcinol in NPC cells were tested by MTT assay, flow cytometry, wound healing assay, western blotting, transwell assay, colony formation assay, immunofluorescence, and transmission electron microscopy (TEM). The anti-tumor efficacy in vivo was evaluated in NPC cells xenograft models. RESULTS Functional studies revealed that isogarcinol inhibited the proliferation, colony formation, migration and invasion abilities of NPC cells in vitro. Isogarcinol caused mitochondrial damage to overproduce reactive oxygen species through reducing the mitochondrial membrane potential and ΔΨm. Isogarcinol also substantially inhibited NPC cells growth in a xenograft tumor model without any obvious toxicity when compared with paclitaxel (PTX). Mechanistic studies have illustrated that isogarcinol increased the Bax/Bcl-2 ratio, cleaved caspase-3, and cytoplasmic cytochrome C levels to induce mitochondrial apoptosis. The ROS overproduction by isogarcinol could suppress EMT pathway via decreasing the levels of p-Akt and Snail. Furthermore, isogarcinol promoted the conversion of LC3-Ⅰ to LC3-Ⅱ, but increased p62 level to block autophagic flux, resulting in the accumulation of damaged mitochondria to promote autophagic cell death in NPC cells. CONCLUSION This study provides a new theoretical foundation for the anti-tumor application of Garcinia oblongifolia and confirms that isogarcinol could be developed as a candidate drug for NPC treatment with low toxicity.
Collapse
Affiliation(s)
- Jing Li
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, University Town, Nanshan, Shenzhen 518055, China
| | - Xi Shen
- Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, No. 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Chunhui Sun
- Research Center, Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen 518107, China
| | - Yibo Hou
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, University Town, Nanshan, Shenzhen 518055, China
| | - Ya Hu
- Department of Stomatology, Shenzhen People's Hospital, the First Affiliated Hospital, Southern University of Science and Technology, No. 1017 East Gate North Road, Luohu District, Shenzhen 518020, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, University Town, Nanshan, Shenzhen 518055, China
| | - Laiqiang Huang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, University Town, Nanshan, Shenzhen 518055, China
| | - Lan Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, University Town, Nanshan, Shenzhen 518055, China.
| | - Yubo Zhang
- Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, No. 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China.
| | - Xiaoyong Dai
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, University Town, Nanshan, Shenzhen 518055, China; Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, No. 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China.
| |
Collapse
|
2
|
Mo S, Liu T, Zhou H, Huang J, Zhao L, Lu F, Kuang Y. ATP6V1B1 regulates ovarian cancer progression and cisplatin sensitivity through the mTOR/autophagy pathway. Mol Cell Biochem 2024:10.1007/s11010-024-05025-w. [PMID: 38735913 DOI: 10.1007/s11010-024-05025-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
Early detection and effective chemotherapy for ovarian cancer, a serious gynecological malignancy, require further progress. This study aimed to investigate the molecular mechanism of ATPase H+-Transporting V1 Subunit B1 (ATP6V1B1) in ovarian cancer development and chemoresistance. Our data show that ATP6V1B1 is upregulated in ovarian cancer and correlated with decreased progression-free survival. Gain- and loss-of-function experiments demonstrated that ATP6V1B1 promotes the proliferation, migration, and invasion of ovarian cancer cells in vitro, while ATP6V1B1 knockout inhibits tumor growth in vivo. In addition, knocking down ATP6V1B1 increases the sensitivity of ovarian cancer cells to cisplatin. Mechanistic studies showed that ATP6V1B1 regulates the activation of the mTOR/autophagy pathway. Overall, our study confirmed the oncogenic role of ATP6V1B1 in ovarian cancer and revealed that ATP6V1B1 promotes ovarian cancer progression via the mTOR/autophagy axis.
Collapse
Affiliation(s)
- Shien Mo
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Gaungxi Medical University, Ministry of Education, Nanning, Guangxi, China
- Guangxi Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Nanning, Guangxi, China
| | - Tingji Liu
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Haiqin Zhou
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Gaungxi Medical University, Ministry of Education, Nanning, Guangxi, China
- Guangxi Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Nanning, Guangxi, China
| | - Junning Huang
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ling Zhao
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fangfang Lu
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Kuang
- Department of Gynecology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Gaungxi Medical University, Ministry of Education, Nanning, Guangxi, China.
- Guangxi Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
3
|
Zhou Y, Shan R, Xie W, Zhou Q, Yin Q, Su Y, Xiao J, Luo P, Yao X, Fang J, Wen F, Shen E, Weng J. Role of autophagy-related genes in liver cancer prognosis. Genomics 2024; 116:110852. [PMID: 38703969 DOI: 10.1016/j.ygeno.2024.110852] [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: 11/08/2023] [Revised: 03/01/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Autophagy, a highly conserved process of protein and organelle degradation, has emerged as a critical regulator in various diseases, including cancer progression. In the context of liver cancer, the predictive value of autophagy-related genes remains ambiguous. Leveraging chip datasets from the TCGA and GTEx databases, we identified 23 differentially expressed autophagy-related genes in liver cancer. Notably, five key autophagy genes, PRKAA2, BIRC5, MAPT, IGF1, and SPNS1, were highlighted as potential prognostic markers, with MAPT showing significant overexpression in clinical samples. In vitro cellular assays further demonstrated that MAPT promotes liver cancer cell proliferation, migration, and invasion by inhibiting autophagy and suppressing apoptosis. Subsequent in vivo studies further corroborated the pro-tumorigenic role of MAPT by suppressing autophagy. Collectively, our model based on the five key genes provides a promising tool for predicting liver cancer prognosis, with MAPT emerging as a pivotal factor in tumor progression through autophagy modulation.
Collapse
Affiliation(s)
- Yuling Zhou
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Rong Shan
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Wangti Xie
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Qiang Zhou
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Qinghua Yin
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Yuqi Su
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Jia Xiao
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Pan Luo
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Xiang Yao
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Jianlong Fang
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Fang Wen
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China
| | - Erdong Shen
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China.
| | - Jie Weng
- Department of Oncology, Yueyang Central Hospital, Yueyang 414000, PR China.
| |
Collapse
|
4
|
Chan SM, Raglow Z, Pal A, Gitlin SD, Legendre M, Thomas D, Mehta RK, Tan M, Nyati MK, Rehemtulla A, Markovitz DM. A molecularly engineered lectin destroys EGFR and inhibits the growth of non-small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585535. [PMID: 38562773 PMCID: PMC10983887 DOI: 10.1101/2024.03.18.585535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Survival rates for non-small cell lung cancer (NSCLC) remain low despite the advent of novel therapeutics. Tyrosine kinase inhibitors (TKIs) targeting mutant epidermal growth factor receptor (EGFR) in NSCLC have significantly improved mortality but are plagued with challenges--they can only be used in the small fraction of patients who have susceptible driver mutations, and resistance inevitably develops. Aberrant glycosylation on the surface of cancer cells is an attractive therapeutic target as these abnormal glycosylation patterns are typically specific to cancer cells and are not present on healthy cells. H84T BanLec (H84T), a lectin previously engineered by our group to separate its antiviral activity from its mitogenicity, exhibits precision binding of high mannose, an abnormal glycan present on the surface of many cancer cells, including NSCLC. Here, we show that H84T binds to and inhibits the growth of diverse NSCLC cell lines by inducing lysosomal degradation of EGFR and leading to cancer cell death through autophagy. This is a mechanism distinct from EGFR TKIs and is independent of EGFR mutation status; H84T inhibited proliferation of both cell lines expressing wild type EGFR and those expressing mutant EGFR that is resistant to all TKIs. Further, H84T binds strongly to multiple and diverse clinical samples of both pulmonary adenocarcinoma and squamous cell carcinoma. H84T is thus a promising potential therapeutic in NSCLC, with the ability to circumvent the challenges currently faced by EGFR TKIs.
Collapse
|
5
|
Samare-Najaf M, Samareh A, Savardashtaki A, Khajehyar N, Tajbakhsh A, Vakili S, Moghadam D, Rastegar S, Mohsenizadeh M, Jahromi BN, Vafadar A, Zarei R. Non-apoptotic cell death programs in cervical cancer with an emphasis on ferroptosis. Crit Rev Oncol Hematol 2024; 194:104249. [PMID: 38145831 DOI: 10.1016/j.critrevonc.2023.104249] [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: 08/30/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Cervical cancer, a pernicious gynecological malignancy, causes the mortality of hundreds of thousands of females worldwide. Despite a considerable decline in mortality, the surging incidence rate among younger women has raised serious concerns. Immortality is the most important characteristic of tumor cells, hence the carcinogenesis of cervical cancer cells pivotally requires compromising with cell death mechanisms. METHODS The current study comprehensively reviewed the mechanisms of non-apoptotic cell death programs to provide possible disease management strategies. RESULTS Comprehensive evidence has stated that focusing on necroptosis, pyroptosis, and autophagy for disease management is associated with significant limitations such as insufficient understanding, contradictory functions, dependence on disease stage, and complexity of intracellular pathways. However, ferroptosis represents a predictable role in cervix carcinogenesis, and ferroptosis-related genes demonstrate a remarkable correlation with patient survival and clinical outcomes. CONCLUSION Ferroptosis may be an appropriate option for disease management strategies from predicting prognosis to treatment.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran.
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nastaran Khajehyar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Delaram Moghadam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Rastegar
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Mohsenizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | | | - Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Zarei
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Tang Q, Xu M, Long S, Yu Y, Ma C, Wang R, Li J, Wang X, Fang F, Han L, Wu W, Wang S. FZKA reverses gefitinib resistance by regulating EZH2/Snail/EGFR signaling pathway in lung adenocarcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116646. [PMID: 37269912 DOI: 10.1016/j.jep.2023.116646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 04/08/2023] [Accepted: 05/15/2023] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzheng Kang-Ai (FZKA) decoction is mainly composed of 12 components with different types of herbs. In the last decade, FZKA has been used as an adjuvant treatment for lung cancer in clinical practice. Our previous studies have confirmed that FZKA shows a strong anti-cancer activity, significantly increases the clinical efficacy of gefitinib and reverses gefitinib resistance in non-small cell lung cancer (NSCLC). However, the molecular mechanism still needs to be further elucidated. AIM OF THE STUDY The aim of this study was to investigate the role and mechanism by which FZKA inhibited the cell growth, proliferation and invasion of lung adenocarcinoma(LUAD) and reversed the acquired resistance of gefitinib for the therapy in LUAD. MATERIALS AND METHODS Cell viability assay and EDU assay were used for detecting of cell viability and cell proliferation. Transwell assay was performed to measure cell invasion. Western Blot and qRT-PCR were used for protein and gene expression test. The gene promoter activity was determined by dul-luciferase reporter assay. The in situ expression of protein was measured by cell immunofluorescence. Stabilized cell lines were established for stable overexpression of EZH2. Transient transfection assay was used for gene silence and overexpression. Xenograft tumors and bioluminescent imaging were used for in vivo experiments. RESULTS FZKA significantly inhibited the cell viability, proliferation and cell invasion of LUAD, the combination of FZKA and gefitinib had a great synergy on the above processes. Moreover, FZKA significantly decreased EZH2 mRNA and protein expression, FZKA reversed the resistance of gefitinib by down-regulation of EZH2 protein. ERK1/2 kinase mediated the down-regulation of EZH2 reduced by FZKA. In addition, FZKA decreased the expression of Snail and EGFR by decreasing EZH2. Overexpression of Snail and EGFR significantly reversed the effect of FZKA-inhibited cell invasion and cell proliferation. More important, the combination of FZKA and gefitinib enhanced the inhibitory effect on EZH2, Snail and EGFR proteins. Furthermore, the growth inhibition and reversal of gefitinib resistance induced by FZKA were further validated in vivo. Finally, the expression and clinical correlation of EZH2,EGFR and Snail in cancer patients were further validated using bioinformatics analysis. CONCLUSIONS FZKA significantly suppressed tumor progression and reversed gefitinib resistance by regulating the p-ERK1/2-EZH2-Snail/EGFR signaling pathway in LUAD.
Collapse
Affiliation(s)
- Qing Tang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, 510120, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China.
| | - Mengfei Xu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Shunqin Long
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, 510120, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Yaya Yu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Changju Ma
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Rui Wang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Jing Li
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Xi Wang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China
| | - Fang Fang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, 530000, PR China
| | - Ling Han
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, 510120, PR China.
| | - Wanyin Wu
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, 510120, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China.
| | - Sumei Wang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, 510120, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, PR China.
| |
Collapse
|
7
|
Thomas BJ, Guldenpfennig C, Guan Y, Winkler C, Beecher M, Beedy M, Berendzen AF, Ma L, Daniels MA, Burke DH, Porciani D. Targeting lung cancer with clinically relevant EGFR mutations using anti-EGFR RNA aptamer. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102046. [PMID: 37869258 PMCID: PMC10589377 DOI: 10.1016/j.omtn.2023.102046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
A significant fraction of non-small cell lung cancer (NSCLC) cases are due to oncogenic mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR). Anti-EGFR antibodies have shown limited clinical benefit for NSCLC, whereas tyrosine kinase inhibitors (TKIs) are effective, but resistance ultimately occurs. The current landscape suggests that alternative ligands that target wild-type and mutant EGFRs are desirable for targeted therapy or drug delivery development. Here we evaluate NSCLC targeting using an anti-EGFR aptamer (MinE07). We demonstrate that interaction sites of MinE07 overlap with clinically relevant antibodies targeting extracellular domain III and that MinE07 retains binding to EGFR harboring the most common oncogenic and resistance mutations. When MinE07 was linked to an anti-c-Met aptamer, the EGFR/c-Met bispecific aptamer (bsApt) showed superior labeling of NSCLC cells in vitro relative to monospecific aptamers. However, dual targeting in vivo did not improve the recognition of NSCLC xenografts compared to MinE07. Interestingly, biodistribution of Cy7-labeled bsApt differed significantly from Alexa Fluor 750-labeled bsApt. Overall, our findings demonstrate that aptamer formulations containing MinE07 can target ectopic lung cancer without additional stabilization or PEGylation and highlights the potential of MinE07 as a targeting reagent for the recognition of NSCLC harboring clinically relevant EGFRs.
Collapse
Affiliation(s)
- Brian J. Thomas
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | - Caitlyn Guldenpfennig
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | - Yue Guan
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | - Calvin Winkler
- Department of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Margaret Beecher
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Michaela Beedy
- Department of Biochemistry, Westminster College, Fulton, MO 65251, USA
| | - Ashley F. Berendzen
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Lixin Ma
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Department of Radiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Mark A. Daniels
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | - Donald H. Burke
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri School of Medicine, Columbia, MO 65211, USA
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - David Porciani
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri School of Medicine, Columbia, MO 65211, USA
| |
Collapse
|
8
|
Crintea A, Constantin AM, Motofelea AC, Crivii CB, Velescu MA, Coșeriu RL, Ilyés T, Crăciun AM, Silaghi CN. Targeted EGFR Nanotherapy in Non-Small Cell Lung Cancer. J Funct Biomater 2023; 14:466. [PMID: 37754880 PMCID: PMC10532491 DOI: 10.3390/jfb14090466] [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: 08/03/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Despite advances in treatment, the prognosis remains poor, highlighting the need for novel therapeutic strategies. The present review explores the potential of targeted epidermal growth factor receptor (EGFR) nanotherapy as an alternative treatment for NSCLC, showing that EGFR-targeted nanoparticles are efficiently taken up by NSCLC cells, leading to a significant reduction in tumor growth in mouse models. Consequently, we suggest that targeted EGFR nanotherapy could be an innovative treatment strategy for NSCLC; however, further studies are needed to optimize the nanoparticles and evaluate their safety and efficacy in clinical settings and human trials.
Collapse
Affiliation(s)
- Andreea Crintea
- Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania; (A.C.); (T.I.); (C.N.S.)
| | - Anne-Marie Constantin
- Department of Morphological Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania; (A.-M.C.); (C.-B.C.)
| | - Alexandru C. Motofelea
- Department of Internal Medicine, University of Medicine and Pharmacy “Victor Babeș”, 300041 Timișoara, Romania;
| | - Carmen-Bianca Crivii
- Department of Morphological Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania; (A.-M.C.); (C.-B.C.)
| | - Maria A. Velescu
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania;
| | - Răzvan L. Coșeriu
- Department of Microbiology, University of Medicine, Pharmacy, Science and Technology “George Emil Palade”, 540142 Târgu-Mureș, Romania;
| | - Tamás Ilyés
- Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania; (A.C.); (T.I.); (C.N.S.)
| | - Alexandra M. Crăciun
- Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania; (A.C.); (T.I.); (C.N.S.)
| | - Ciprian N. Silaghi
- Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400349 Cluj-Napoca, Romania; (A.C.); (T.I.); (C.N.S.)
| |
Collapse
|
9
|
Jin C, Wang T, Yang Y, Zhou P, Li J, Wu W, Lv X, Ma G, Wang A. Rational targeting of autophagy in colorectal cancer therapy: From molecular interactions to pharmacological compounds. ENVIRONMENTAL RESEARCH 2023; 227:115721. [PMID: 36965788 DOI: 10.1016/j.envres.2023.115721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 05/08/2023]
Abstract
The abnormal progression of tumors has been a problem for treatment of cancer and therapeutic should be directed towards targeting main mechanisms involved in tumorigenesis in tumors. The genomic mutations can result in changes in biological mechanisms in human cancers. Colorectal cancer is one of the most malignant tumors of gastrointestinal tract and its treatment has been faced some difficulties due to development of resistance in tumor cells and also, their malignant behavior. Hence, new therapeutic modalities for colorectal cancer are being investigated. Autophagy is a "self-digestion" mechanism that is responsible for homeostasis preserving in cells and its aberrant activation/inhibition can lead to tumorigenesis. The current review focuses on the role of autophagy mechanism in colorectal cancer. Autophagy may be associated with increase/decrease in progression of colorectal cancer due to mutual function of this molecular mechanism. Pro-survival autophagy inhibits apoptosis to increase proliferation and survival rate of colorectal tumor cells and it is also involved in cancer metastasis maybe due to EMT induction. In contrast, pro-death autophagy decreases growth and invasion of colorectal tumor cells. The status of autophagy (upregulation and down-regulation) is a determining factor for therapy response in colorectal tumor cells. Therefore, targeting autophagy can increase sensitivity of colorectal tumor cells to chemotherapy and radiotherapy. Interestingly, nanoparticles can be employed for targeting autophagy in cancer therapy and they can both induce/suppress autophagy in tumor cells. Furthermore, autophagy modulators can be embedded in nanostructures in improving tumor suppression and providing cancer immunotherapy.
Collapse
Affiliation(s)
- Canhui Jin
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Tianbao Wang
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Yanhui Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pin Zhou
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Juncheng Li
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Wenhao Wu
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Xin Lv
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Guoqing Ma
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China
| | - Aihong Wang
- Department of Gastrointestinal Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, PR China.
| |
Collapse
|
10
|
Sanati M, Afshari AR, Ahmadi SS, Kesharwani P, Sahebkar A. Aptamers against cancer drug resistance: Small fighters switching tactics in the face of defeat. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166720. [PMID: 37062453 DOI: 10.1016/j.bbadis.2023.166720] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 04/18/2023]
Abstract
Discovering novel cancer therapies has attracted extreme interest in the last decade. In this regard, multidrug resistance (MDR) to chemotherapies is the primary challenge in cancer treatment. Cancerous cells are growingly become resistant to existing chemotherapeutics by employing diverse mechanisms, highlighting the significance of discovering approaches to overcome MDR. One promising strategy is utilizing aptamers as unique tools to target elements or signalings incorporated in resistance mechanisms or develop active targeted drug delivery systems or chimeras enabling the precise delivery of novel agents to inhibit the conventionally undruggable resistance elements. Further, due to their advantages over their proteinaceous counterparts, particularly antibodies, including improved targeting action, enhanced thermal stability, easier production, and superior tumor penetration, aptamers are emerging and have frequently been considered for developing cancer therapeutics. Here, we highlighted significant chemoresistance pathways and thoroughly discussed using aptamers as prospective tools to surmount cancer MDR.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Pan Q, Lu Y, Xie L, Wu D, Liu R, Gao W, Luo K, He B, Pu Y. Recent Advances in Boosting EGFR Tyrosine Kinase Inhibitors-Based Cancer Therapy. Mol Pharm 2023; 20:829-852. [PMID: 36588471 DOI: 10.1021/acs.molpharmaceut.2c00792] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epidermal growth factor receptor (EGFR) plays a key role in signal transduction pathways associated with cell proliferation, growth, and survival. Its overexpression and aberrant activation in malignancy correlate with poor prognosis and short survival. Targeting inhibition of EGFR by small-molecular tyrosine kinase inhibitors (TKIs) is emerging as an important treatment model besides of chemotherapy, greatly reshaping the landscape of cancer therapy. However, they are still challenged by the off-targeted toxicity, relatively limited cancer types, and drug resistance after long-term therapy. In this review, we summarize the recent progress of oral, pulmonary, and injectable drug delivery systems for enhanced and targeting TKI delivery to tumors and reduced side effects. Importantly, EGFR-TKI-based combination therapies not only greatly broaden the applicable cancer types of EGFR-TKI but also significantly improve the anticancer effect. The mechanisms of TKI resistance are summarized, and current strategies to overcome TKI resistance as well as the application of TKI in reversing chemotherapy resistance are discussed. Finally, we provide a perspective on the future research of EGFR-TKI-based cancer therapy.
Collapse
Affiliation(s)
- Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Yao Lu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Li Xie
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Di Wu
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rong Liu
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|