Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Research Article

Mechanism Research of PZD Inhibiting Lung Cancer Cell Proliferation, Invasion, and Migration based on Network Pharmacology

Author(s): Fan Feng*, Ping Hu, Lei Peng, Jun Chen and Xingkui Tao

Volume 30, Issue 16, 2024

Published on: 03 April, 2024

Page: [1279 - 1293] Pages: 15

DOI: 10.2174/0113816128296328240329032332

open access plus

Abstract

Background: A classic Chinese medicine decoction, Pinellia ternata (Thunb.) Breit.-Zingiber officinale Roscoe (Ban-Xia and Sheng-Jiang in Chinese) decoction (PZD), has shown significant therapeutic effects on lung cancer.

Objective: This study aimed to explore and elucidate the mechanism of action of PZD on lung cancer using network pharmacology methods.

Methods: Active compounds were selected according to the ADME parameters recorded in the TCMSP database. Potential pathways related to genes were identified through GO and KEGG analysis. The compoundtarget network was constructed by using Cytoscape 3.7.1 software, and the core common targets were obtained by protein-protein interaction (PPI) network analysis. Batch molecular docking of small molecule compounds and target proteins was carried out by using the AutoDock Vina program. Different concentrations of PZD water extracts (10, 20, 40, 80, and 160 μg/mL) were used on lung cancer cells. Moreover, MTT and Transwell experiments were conducted to validate the prominent therapeutic effects of PZD on lung cancer cell H1299.

Results: A total of 381 components in PZD were screened, of which 16 were selected as bioactive compounds. The compound-target network consisting of 16 compounds and 79 common core targets was constructed. MTT experiment showed that the PZD extract could inhibit the cell proliferation of NCI-H1299 cells, and the IC50 was calculated as 97.34 ± 6.14 μg/mL. Transwell and wound-healing experiments showed that the PZD could significantly decrease cell migration and invasion at concentrations of 80 and 160 μg/mL, respectively. The in vitro experiments confirmed that PZD had significant therapeutic effects on lung cancer cells, mainly through the PI3K/AKT signaling pathway.

Conclusion: PZD could inhibit the cell proliferation, migration, and invasion of NCI-H1299 cells partially through the PI3K/AKT signaling pathway. These findings suggested that PZD might be a potential treatment strategy for lung cancer patients.

Keywords: Network pharmacology, Pinellia ternata, lung cancer, traditional Chinese medicine, PI3K/AKT signaling pathway, KEGG analysis.

« Previous
[1]
Moris D, Stathopoulos NI, Tsilimigras DI, et al. Insights into novel prognostic and possible predictive biomarkers of lung neuroendocrine tumors. Can Geno Prot 2018; 15(2): 153-63.
[PMID: 29496694]
[2]
He J, Li N, Chen WQ, et al. China guideline for the screening and early detection of lung cancer. Zhonghua Zhong Liu Za Zhi 2021; 43(3): 243-68.
[PMID: 33752304]
[3]
Li Z, Feiyue Z, Gaofeng L. Traditional Chinese medicine and lung cancer-From theory to practice. Biomed Pharmacother 2021; 137: 111381.
[http://dx.doi.org/10.1016/j.biopha.2021.111381] [PMID: 33601147]
[4]
Jiang Y, Liu LS, Shen LP, et al. Traditional Chinese medicine treatment as maintenance therapy in advanced non-small-cell lung cancer: A randomized controlled trial. Complement Ther Med 2016; 24: 55-62.
[http://dx.doi.org/10.1016/j.ctim.2015.12.006] [PMID: 26860802]
[5]
Ye L, Jia Y, Ji K, et al. Traditional Chinese medicine in the prevention and treatment of cancer and cancer metastasis. Oncol Lett 2015; 10(3): 1240-50.
[http://dx.doi.org/10.3892/ol.2015.3459] [PMID: 26622657]
[6]
McCubrey JA, Lertpiriyapong K, Steelman LS, et al. Effects of resveratrol, curcumin, berberine and other nutraceuticals on aging, cancer development, cancer stem cells and microRNAs. Aging 2017; 9(6): 1477-536.
[http://dx.doi.org/10.18632/aging.101250] [PMID: 28611316]
[7]
Izzo C, Annunziata M, Melara G, et al. The role of resveratrol in liver disease: A comprehensive review from in vitro to clinical trials. Nutrients 2021; 13(3): 933.
[http://dx.doi.org/10.3390/nu13030933] [PMID: 33805795]
[8]
Sarawi WS, Alhusaini AM, Fadda LM, et al. Curcumin and nano-curcumin mitigate copper neurotoxicity by modulating oxidative stress, inflammation, and Akt/GSK-3β signaling. Molecules 2021; 26(18): 5591.
[http://dx.doi.org/10.3390/molecules26185591] [PMID: 34577062]
[9]
Xiang Y, Guo Z, Zhu P, Chen J, Huang Y. Traditional Chinese medicine as a cancer treatment: Modern perspectives of ancient but advanced science. Cancer Med 2019; 8(5): 1958-75.
[http://dx.doi.org/10.1002/cam4.2108] [PMID: 30945475]
[10]
Wang SF, Wu MY, Cai CZ, Li M, Lu JH. Autophagy modulators from traditional Chinese medicine: Mechanisms and therapeutic potentials for cancer and neurodegenerative diseases. J Ethnopharmacol 2016; 194: 861-76.
[http://dx.doi.org/10.1016/j.jep.2016.10.069] [PMID: 27793785]
[11]
Zhang Y, Lou Y, Wang J, Yu C, Shen W. Research status and molecular mechanism of the traditional Chinese medicine and antitumor therapy combined strategy based on tumor microenvironment. Front Immunol 2021; 11: 609705.
[http://dx.doi.org/10.3389/fimmu.2020.609705] [PMID: 33552068]
[12]
Tang H, Shu P, Liu S, Zhang X, Belmonte MM. Traditional Chinese medicine in oncotherapy: The research status. Nutr Cancer 2020; 72(6): 992-8.
[http://dx.doi.org/10.1080/01635581.2019.1664599] [PMID: 31526143]
[13]
Wang S, Long S, Wu W. Application of traditional Chinese medicines as personalized therapy in human cancers. Am J Chin Med 2018; 46(5): 953-70.
[http://dx.doi.org/10.1142/S0192415X18500507] [PMID: 29986595]
[14]
Ma Y, Zhang X, Su Z, et al. Insight into the molecular mechanism of a herbal injection by integrating network pharmacology and in vitro. J Ethnopharmacol 2015; 173: 91-9.
[http://dx.doi.org/10.1016/j.jep.2015.07.016] [PMID: 26192807]
[15]
Zhao L, Zhang H, Li N, et al. Network pharmacology, A promising approach to reveal the pharmacology mechanism of Chinese medicine formula. J Ethnopharmacol 2023; 309: 116306.
[http://dx.doi.org/10.1016/j.jep.2023.116306] [PMID: 36858276]
[16]
Yang HY, Liu ML, Luo P, Yao XS, Zhou H. Network pharmacology provides a systematic approach to understanding the treatment of ischemic heart diseases with traditional Chinese medicine. Phytomedicine 2022; 104: 154268.
[http://dx.doi.org/10.1016/j.phymed.2022.154268] [PMID: 35777118]
[17]
Guo W, Huang J, Wang N, et al. Integrating network pharmacology and pharmacological evaluation for deciphering the action mechanism of herbal formula Zuojin pill in suppressing hepatocellular carcinoma. Front Pharmacol 2019; 10: 1185.
[http://dx.doi.org/10.3389/fphar.2019.01185] [PMID: 31649545]
[18]
Yang J, Tian S, Zhao J, Zhang W. Exploring the mechanism of TCM formulae in the treatment of different types of coronary heart disease by network pharmacology and machining learning. Pharmacol Res 2020; 159: 105034.
[http://dx.doi.org/10.1016/j.phrs.2020.105034] [PMID: 32565312]
[19]
Zhang P, Zhang D, Zhou W, et al. Network pharmacology: Towards the artificial intelligence-based precision traditional Chinese medicine. Brief Bioinform 2023; 25(1): bbad518.
[http://dx.doi.org/10.1093/bib/bbad518] [PMID: 38197310]
[20]
Huang J, Cheung F, Tan HY, et al. Identification of the active compounds and significant pathways of Yinchenhao decoction based on network pharmacology. Mol Med Rep 2017; 16(4): 4583-92.
[http://dx.doi.org/10.3892/mmr.2017.7149] [PMID: 28791364]
[21]
Zhang J, Liu X, Wu J, et al. A bioinformatics investigation into the pharmacological mechanisms of the effect of the Yinchenhao decoction on hepatitis C based on network pharmacology. BMC Complement Med Therapies 2020; 20(1): 50.
[http://dx.doi.org/10.1186/s12906-020-2823-y] [PMID: 32050950]
[22]
Yan F, Feng M, Wang X, et al. Molecular targets of Yangyin Fuzheng Jiedu prescription in the treatment of hepatocellular carcinoma based on network pharmacology analysis. Cancer Cell Int 2020; 20(1): 540.
[http://dx.doi.org/10.1186/s12935-020-01596-y] [PMID: 33292207]
[23]
Denisov EV, Schegoleva AA, Gervas PA, et al. Premalignant lesions of squamous cell carcinoma of the lung: The molecular make-up and factors affecting their progression. Lung Cancer 2019; 135: 21-8.
[http://dx.doi.org/10.1016/j.lungcan.2019.07.001] [PMID: 31446997]
[24]
Su J, Tan S, Gong H, et al. The evaluation of prognostic value and immune characteristics of ferroptosis-related genes in lung squamous cell carcinoma. Global Med Genet 2023; 10(4): 285-300.
[http://dx.doi.org/10.1055/s-0043-1776386] [PMID: 37915460]
[25]
Panakkal N, Lekshmi A, Saraswathy VV, Sujathan K. Effective lung cancer control: An unaccomplished challenge in cancer research. Cytojournal 2023; 20: 16.
[http://dx.doi.org/10.25259/Cytojournal_36_2022] [PMID: 37681073]
[26]
Xin T, Zhang Y, Pu X, Gao R, Xu Z, Song J. Trends in herbgenomics. Sci China Life Sci 2019; 62(3): 288-308.
[http://dx.doi.org/10.1007/s11427-018-9352-7] [PMID: 30128965]
[27]
Li X, Tang Z, Wen L, Jiang C, Feng Q. Matrine: A review of its pharmacology, pharmacokinetics, toxicity, clinical application and preparation researches. J Ethnopharmacol 2021; 269: 113682.
[http://dx.doi.org/10.1016/j.jep.2020.113682] [PMID: 33307055]
[28]
Zou T, Wang J, Wu X, et al. A review of the research progress on Pinellia ternata (Thunb.) Breit.: Botany, traditional uses, phytochemistry, pharmacology, toxicity and quality control. Heliyon 2023; 9(11): e22153.
[http://dx.doi.org/10.1016/j.heliyon.2023.e22153] [PMID: 38058630]
[29]
Bi L, Xie C, Jiao L, et al. CPF impedes cell cycle re‐entry of quiescent lung cancer cells through transcriptional suppression of FACT and c‐MYC. J Cell Mol Med 2020; 24(3): 2229-39.
[http://dx.doi.org/10.1111/jcmm.14897] [PMID: 31960591]
[30]
de Lima RMT, dos Reis AC, de Menezes AAPM, et al. Protective and therapeutic potential of ginger (Zingiber officinale) extract and [6]‐gingerol in cancer: A comprehensive review. Phytother Res 2018; 32(10): 1885-907.
[http://dx.doi.org/10.1002/ptr.6134] [PMID: 30009484]
[31]
Liu CM, An L, Wu Z, et al. 6 Gingerol suppresses cell viability, migration and invasion via inhibiting EMT, and inducing autophagy and ferroptosis in LPS stimulated and LPS unstimulated prostate cancer cells. Oncol Lett 2022; 23(6): 187.
[http://dx.doi.org/10.3892/ol.2022.13307] [PMID: 35527779]
[32]
Ru J, Li P, Wang J, et al. TCMSP: A database of systems pharmacology for drug discovery from herbal medicines. J Cheminform 2014; 6(1): 13.
[http://dx.doi.org/10.1186/1758-2946-6-13] [PMID: 24735618]
[33]
Wang M, Zhao F, Li Z, Li X, Dong L. Tectoridin and PLK1 inhibitor synergistically promote the apoptosis of lung adenocarcinoma cells: Bioinformatic analysis of TCGA and TCMSP. Naunyn Schmiedebergs Arch Pharmacol 2023; 396(10): 2417-26.
[http://dx.doi.org/10.1007/s00210-023-02460-2] [PMID: 37014402]
[34]
Xu X, Zhang W, Huang C, et al. A novel chemometric method for the prediction of human oral bioavailability. Int J Mol Sci 2012; 13(6): 6964-82.
[http://dx.doi.org/10.3390/ijms13066964] [PMID: 22837674]
[35]
Tao W, Xu X, Wang X, et al. Network pharmacology-based prediction of the active ingredients and potential targets of Chinese herbal Radix curcumae formula for application to cardiovascular disease. J Ethnopharmacol 2013; 145(1): 1-10.
[http://dx.doi.org/10.1016/j.jep.2012.09.051] [PMID: 23142198]
[36]
Shang L, Wang Y, Li J. Mechanism of Sijunzi decoction in the treatment of colorectal cancer based on network pharmacology and experimental validation. J Ethnopharmacol 2023; 302(Pt A): 115876.
[37]
Lu S, Sun X, Zhou Z, et al. Mechanism of Bazhen decoction in the treatment of colorectal cancer based on network pharmacology, molecular docking, and experimental validation. Front Immunol 2023; 14: 1235575.
[http://dx.doi.org/10.3389/fimmu.2023.1235575] [PMID: 37799727]
[38]
Milano M, Zucco C, Settino M, Cannataro M. An extensive assessment of network embedding in PPI network alignment. Entropy 2022; 24(5): 730.
[http://dx.doi.org/10.3390/e24050730] [PMID: 35626613]
[39]
Chen L, Zhang YH, Wang S, Zhang Y, Huang T, Cai YD. Prediction and analysis of essential genes using the enrichments of gene ontology and KEGG pathways. PLoS One 2017; 12(9): e0184129.
[http://dx.doi.org/10.1371/journal.pone.0184129] [PMID: 28873455]
[40]
Li C, Xu H, Chen X, et al. Aqueous extract of clove inhibits tumor growth by inducing autophagy through AMPK/ULK pathway. Phytother Res 2019; 33(7): 1794-804.
[http://dx.doi.org/10.1002/ptr.6367] [PMID: 30993793]
[41]
Idriss H, Siddig B, Maldonado PG, et al. Phytochemical discrimination, biological activity and molecular docking of water-soluble inhibitors from Saussurea costus herb against main protease of SARS-CoV-2. Molecules 2022; 27(15): 4908.
[http://dx.doi.org/10.3390/molecules27154908] [PMID: 35956858]
[42]
Zhou H, Feng X, Yan Y, et al. Optimization of an ultrasonic-assisted aqueous two-phase extraction method for four flavonoids from Lysionotus pauciflorus. Prep Biochem Biotechnol 2022; 52(7): 770-82.
[http://dx.doi.org/10.1080/10826068.2021.1992783] [PMID: 34704892]
[43]
Feng F, Zhang J, Lian C, et al. Nitidine chloride triggers autophagy and apoptosis of ovarian cancer cells through Akt/mTOR signaling pathway. Curr Pharm Des 2023; 29(19): 1524-34.
[http://dx.doi.org/10.2174/1381612829666230614154847] [PMID: 37317923]
[44]
Zhang X, Tao X, Feng F. Downregulation of C12orf75 gene inhibits migration and invasion of liver cancer cell via suppressing the Wnt/β-catenin signaling pathway in vitro. Biochem Biophys Res Commun 2022; 614: 92-9.
[http://dx.doi.org/10.1016/j.bbrc.2022.05.018] [PMID: 35576683]
[45]
Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Proc Natl Acad Sci USA 1979; 76(9): 4350-4.
[http://dx.doi.org/10.1073/pnas.76.9.4350] [PMID: 388439]
[46]
Feng F, Zhu X, Wang C, et al. Downregulation of hypermethylated in cancer-1 by miR-4532 promotes adriamycin resistance in breast cancer cells. Cancer Cell Int 2018; 18(1): 127.
[http://dx.doi.org/10.1186/s12935-018-0616-x] [PMID: 30202238]
[47]
Song S, Huang W, Lu X, et al. A network pharmacology study based on the mechanism of Citri Reticulatae Pericarpium-Pinelliae Rhizoma in the treatment of gastric cancer. Evid Based Complement Alternat Med 2021; 2021: 1-17.
[http://dx.doi.org/10.1155/2021/6667560] [PMID: 33953786]
[48]
Hereñú CB, Crespo R. Phytochemicals as estrogen receptor modulators?-a commentary of a network pharmacology study of two commonly employed Chinese herbal medicines in non-small cell lung cancer treatment. Transl Cancer Res 2023; 12(12): 3249-54.
[http://dx.doi.org/10.21037/tcr-23-1440] [PMID: 38197069]
[49]
Li C, Lu H, Jiang X, Guo X, Zhong H, Li H. Network pharmacology study of Citrus reticulata and Pinellia ternata in the treatment of non-small cell lung cancer. Cell Mol Biol 2022; 67(4): 10-7.
[http://dx.doi.org/10.14715/cmb/2021.67.4.2] [PMID: 35809307]
[50]
Zhai Z, Tao X, Alami MM, Shu S, Wang X. Network pharmacology and molecular docking combined to analyze the molecular and pharmacological mechanism of Pinellia ternata in the treatment of hypertension. Curr Issues Mol Biol 2021; 43(1): 65-78.
[http://dx.doi.org/10.3390/cimb43010006] [PMID: 34062719]
[51]
Rajavel T, Packiyaraj P, Suryanarayanan V, Singh SK, Ruckmani K, Pandima Devi K. β-Sitosterol targets Trx/Trx1 reductase to induce apoptosis in A549 cells via ROS mediated mitochondrial dysregulation and p53 activation. Sci Rep 2018; 8(1): 2071.
[http://dx.doi.org/10.1038/s41598-018-20311-6] [PMID: 29391428]
[52]
Khan Z, Nath N, Rauf A, et al. Multifunctional roles and pharmacological potential of β-sitosterol: Emerging evidence toward clinical applications. Chem Biol Interact 2022; 365: 110117.
[http://dx.doi.org/10.1016/j.cbi.2022.110117] [PMID: 35995256]
[53]
Vo VG, Guest PC, Nguyen TT, Vo TK. Evaluation of anti-hepatocellular-cancer properties of β-sitosterol and β-sitosterol-glucoside from Indigofera zollingeriana Miq. Methods Mol Biol 2022; 2343: 229-40.
[http://dx.doi.org/10.1007/978-1-0716-1558-4_15] [PMID: 34473326]
[54]
Raj RK, Rajeshkumar S. β‐sitosterol‐assisted silver nanoparticles activates Nrf2 and triggers mitochondrial apoptosis via oxidative stress in human hepatocellular cancer cell line. J Biomed Mater Res A 2020; 108(9): 1899-908.
[http://dx.doi.org/10.1002/jbm.a.36953] [PMID: 32319188]
[55]
Wang Z, Zhan Y, Xu J, et al. Beta-sitosterol reverses multidrug resistance via BCRP suppression by inhibiting the p53-MDM2 interaction in colorectal cancer. J Agric Food Chem 2020; 68(12): 3850-8.
[http://dx.doi.org/10.1021/acs.jafc.0c00107] [PMID: 32167760]
[56]
Zhao H, Zhang X, Wang M, Lin Y, Zhou S. Stigmasterol simultaneously induces apoptosis and protective autophagy by inhibiting Akt/mTOR pathway in gastric cancer cells. Front Oncol 2021; 11: 629008.
[http://dx.doi.org/10.3389/fonc.2021.629008] [PMID: 33708631]
[57]
Zhang X, Wang J, Zhu L, et al. Advances in stigmasterol on its anti-tumor effect and mechanism of action. Front Oncol 2022; 12: 1101289.
[http://dx.doi.org/10.3389/fonc.2022.1101289] [PMID: 36578938]
[58]
Liao H, Zhu D, Bai M, et al. Stigmasterol sensitizes endometrial cancer cells to chemotherapy by repressing Nrf2 signal pathway. Cancer Cell Int 2020; 20(1): 480.
[http://dx.doi.org/10.1186/s12935-020-01470-x] [PMID: 33041661]
[59]
Cioccoloni G, Soteriou C, Websdale A, Wallis L, Zulyniak MA, Thorne JL. Phytosterols and phytostanols and the hallmarks of cancer in model organisms: A systematic review and meta-analysis. Crit Rev Food Sci Nutr 2020; 25: 1-21.
[PMID: 33238719]
[60]
Yu M, Qi B, Xiaoxiang W, Xu J, Liu X. Baicalein increases cisplatin sensitivity of A549 lung adenocarcinoma cells via PI3K/] Akt/NF-κB pathway. Biomed Pharmacother 2017; 90: 677-85.
[http://dx.doi.org/10.1016/j.biopha.2017.04.001] [PMID: 28415048]
[61]
Yan W, Ma X, Zhao X, Zhang S. Baicalein induces apoptosis and autophagy of breast cancer cells via inhibiting PI3K/AKT pathway in vivo and vitro. Drug Des Devel Ther 2018; 12: 3961-72.
[http://dx.doi.org/10.2147/DDDT.S181939] [PMID: 30510404]
[62]
Bie B, Sun J, Guo Y, et al. Baicalein: A review of its anti-cancer effects and mechanisms in Hepatocellular Carcinoma. Biomed Pharmacother 2017; 93: 1285-91.
[http://dx.doi.org/10.1016/j.biopha.2017.07.068] [PMID: 28747003]
[63]
Liu ZH, Yang CX, Zhang L, Yang CY, Xu XQ. Baicalein, as a prooxidant, triggers mitochondrial apoptosis in MCF-7 human breast cancer cells through mobilization of intracellular copper and reactive oxygen species generation. OncoTargets Ther 2019; 12(3): 10749-61.
[http://dx.doi.org/10.2147/OTT.S222819] [PMID: 31849483]
[64]
Yan JJ, Gao L, Qin XM. Baicalein attenuates neuroinflammation in LPS-activated BV-2 microglial cells through suppression of pro-inflammatory cytokines, COX2/NF-κB expressions and regulation of metabolic disorder. Zhongguo Yaolixue Yu Dulixue Zazhi 2019; 33(10): 854.
[65]
Zagórska A. Phosphodiesterase 10 (PDE10) inhibitors: An updated patent review (2014-present). Expert Opin Ther Pat 2020; 30(2): 147-57.
[http://dx.doi.org/10.1080/13543776.2020.1709444] [PMID: 31874060]
[66]
Yoshioka T, Itagaki Y, Abe Y, et al. NaCl dependent production of coniferin in Alluaudiopsis marnieriana suspension cultured cells. Plant Biotechnol 2021; 38(1): 183-6.
[http://dx.doi.org/10.5511/plantbiotechnology.21.0102a] [PMID: 34177341]
[67]
Gai F, Janiak MA, Sulewska K, Peiretti PG, Karamać M. Phenolic compound profile and antioxidant capacity of flax (Linum usitatissimum L.) harvested at different growth stages. Molecules 2023; 28(4): 1807.
[http://dx.doi.org/10.3390/molecules28041807] [PMID: 36838795]
[68]
Fan L, He Z, Head SA, et al. Clofoctol and sorafenib inhibit prostate cancer growth via synergistic induction of endoplasmic reticulum stress and UPR pathways. Cancer Manag Res 2018; 10: 4817-29.
[http://dx.doi.org/10.2147/CMAR.S175256] [PMID: 30425575]
[69]
Hu Y, Zhang M, Tian N, et al. The antibiotic clofoctol suppresses glioma stem cell proliferation by activating KLF13. J Clin Invest 2019; 129(8): 3072-85.
[http://dx.doi.org/10.1172/JCI124979] [PMID: 31112526]
[70]
Liu K, Pu J, Nie Z, et al. Ivacaftor inhibits glioblastoma stem cell maintenance and tumor progression. Front Cell Dev Biol 2021; 9: 678209.
[http://dx.doi.org/10.3389/fcell.2021.678209] [PMID: 34046412]
[71]
Kinoshita T, Goto T. Molecular mechanisms of pulmonary fibrogenesis and its progression to lung cancer: A review. Int J Mol Sci 2019; 20(6): 1461.
[http://dx.doi.org/10.3390/ijms20061461] [PMID: 30909462]
[72]
Tzouvelekis A, Gomatou G, Bouros E, Trigidou R, Tzilas V, Bouros D. Common pathogenic mechanisms between idiopathic pulmonary fibrosis and lung cancer. Chest 2019; 156(2): 383-91.
[http://dx.doi.org/10.1016/j.chest.2019.04.114] [PMID: 31125557]
[73]
Bordoloi D, Banik K, Padmavathi G, et al. TIPE2 induced the proliferation, survival, and migration of lung cancer cells through modulation of Akt/mTOR/NF-κB signaling cascade. Biomolecules 2019; 9(12): 836.
[http://dx.doi.org/10.3390/biom9120836] [PMID: 31817720]
[74]
Lee KY, Shueng PW, Chou CM, et al. Elevation of CD109 promotes metastasis and drug resistance in lung cancer via activation of EGFR‐AKT‐mTOR signaling. Cancer Sci 2020; 111(5): 1652-62.
[http://dx.doi.org/10.1111/cas.14373] [PMID: 32133706]
[75]
Tan AC. Targeting the PI3K/Akt/mTOR pathway in non‐small cell lung cancer (NSCLC). Thorac Cancer 2020; 11(3): 511-8.
[http://dx.doi.org/10.1111/1759-7714.13328] [PMID: 31989769]

© 2024 Bentham Science Publishers | Privacy Policy