Letter to the editor

Fisetin-loaded nanoemulsion ameliorates lung cancer pathogenesis via downregulating cathepsin-B, galectin-3 and enolase in an in vitro setting

Ayeh Bani Saeid1,2, Keshav Raj Paudel3, Gabriele De Rubis1,2, Samir Mehndiratta1, Sofia Kokkinis1,2, Sukriti Vishwas4, Stewart Yeung1,2, Gaurav Gupta5,6, Sachin Kumar Singh2[*],4, Kamal Dua1,2

1Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia

2Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia

3Center for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2007, Australia

4School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, 144411, India

5Center for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Punjab, India

6Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates

EXCLI J 2024;23:Doc1238

 

Lung cancer (LC) remains one of the leading causes of cancer-related deaths worldwide, with a high incidence and mortality rate (Barta et al., 2019[6]). According to the World Health Organization (WHO), in 2020 alone, around 2 million LC diagnoses and 1.8 million deaths worldwide were recorded, showing its great burden on public health (WHO, 2023[53]). LC was also the fifth most frequently diagnosed cancer in Australia in 2019, with 13,140 new cases reported that year (Cancer Australia, 2019[7]). Lung cancer often stems from a complex interplay of genetic predisposition, environmental exposures, and lifestyle choices (Travis, 2020[48]). Statistics reveal that smokers are 15 to 30 times more likely to develop lung cancer than non-smokers, with nearly 90 % of lung cancer deaths in men and 80 % in women linked to smoking (Alharbi et al., 2022[1]; Chang et al., 2021[9]). Moreover, exposure to second-hand smoke increases the risk by 20-30 % (Chang et al., 2021)[9]. Furthermore, genetic predispositions, including inherited mutations, play a role, although they are less common (Travis, 2020[48]). Hence, lung cancer's pathogenesis is multifaceted, driven by genetic mutations, epigenetic alterations, and dysregulated signaling pathways, including but not limited to EGFR, RAS/MAPK, PI3K/AKT/mTOR, JAK/STAT, and Wnt/β-catenin, promotes tumor growth and survival (Alharbi et al., 2022[2]; Koni et al., 2020[21]; Mehta et al., 2020[31]). In addition to these signaling pathways and the proteins involved in them, there are some other proteins whose functions, as indicated by some studies, can indirectly play important roles in the progression of lung cancer, including cathepsin B (CTSB), enolase, and galectin-3 (GAL3) (Gondi and Rao; 2013[11], Huang et al., 2017[18]; Pokhare et al., 2022[35]).

Understanding these molecular mechanisms provides insights into potential therapeutic targets and strategies for personalized medicine in lung cancer treatment (Alnuqaydan et al., 2022[3][4]; Singh et al., 2024[42]). Current lung cancer treatments, such as chemotherapy, radiation therapy, and targeted therapy, often face limitations like resistance to treatment, reducing their long-term effectiveness (Hirsch et al., 2017[16]; Saeid et al., 2023[39]). Additionally, severe side effects can significantly impact patients' quality of life (Hirsch et al., 2017[16]). One of the treatment options that has attracted researchers' attention in recent years is fisetin, a naturally occurring flavonoid found in various fruits and vegetables such as strawberries, apples, and onions (Ling et al., 2022[27]; Paudel et al., 2022[33]; Prasher et al., 2020[36]; Saeid et al., 2023[39]). Fisetin has garnered attention for its wide array of biological activities, including anti-inflammatory, antioxidant, and anticancer properties (Imran et al., 2021[20]; Ling et al., 2022[27]). Its anticancer activity is linked to its regulation of key signaling pathways like PI3K/AKT, mTOR, and Wnt/β-catenin, as well as its capacity to induce apoptosis and inhibit angiogenesis (Imran et al., 2021[20]). Despite its promising therapeutic potential, the clinical application of fisetin is often limited by its poor bioavailability and rapid metabolism due to its poor water solubility, which necessitates strategies to enhance and prolong its circulation time for optimal therapeutic efficacy (Szymczak and Cielecka-Piontek, 2023[45]).

Given these challenges, nano emulsions, classified as colloidal dispersions consisting of two immiscible liquids, offer a novel solution to enhance the delivery and efficacy of fisetin (Kumar et al., 2023[25]). They form transparent dispersions of oil in water or water in oil with particle sizes typically ranging from 20 to 200 nm, stabilized by surfactants (Ranjbar et al., 2023[38]; Szymczak and Cielecka-Piontek, 2023[45]). More recently, self-nanoemulsifying drug delivery systems (SNEDDS) have emerged as anhydrous nano emulsions composed of oil, a surfactant, a co-surfactant, and a drug molecule, which, upon oral administration, form nano emulsions in the stomach (Liu et al., 2021[28]; Sharma et al., 2019[41]; Vishwas et al., 2022[50]). This smaller particle size in the nano range increases the surface area for absorption, thus enhancing drug bioavailability (Vishwas et al., 2022[50]). These advantages have prompted researchers to explore the use of self-nanoemulsifying drug delivery systems, including fisetin, to improve its solubility, permeability, and bioefficacy (Kumar et al., 2022[24]; Vishwas et al., 2022[50]). Given the intricate role of CTSB, enolase, and GAL3 enzymes in lung cancer progression and the potential benefits of fisetin, this study aims to explore the effectiveness of a fisetin-loaded nano emulsion (FNE) in attenuating these pathological markers, in vitro, in lung cancer cells, specifically the A549 human lung adenocarcinoma cells. We show that the FNE significantly downregulates CTSB, enolase, and GAL3 enzymes compared to the control group.

The SNEDDS formulation for fisetin (FS-SNEDDS) was prepared by combining specific amounts of castor oil (0.1 mL), Lauroglycol FCC (0.1 mL), Tween 80 (0.4 mL), and Transcutol P (0.6 mL) in a clean glass vial. These components were thoroughly mixed before adding fisetin (5 mg). The formulation and evaluation processes were based on the methods previously detailed by Kumar et al (Kumar et al., 2019[23], 2022[24]). A549 lung cancer cells were cultured in a humidified environment with 5 % CO2 at 37 °C. The medium was refreshed every 48 hours. Cells were plated at a density of 250,000 cells per well in six-well plates and treated with 10 µg/mL fisetin nano emulsion (FNE) for 24 hours the following day. Notably, the non-cytotoxic concentration of 10 µg/mL was determined through an MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cell viability assay, which indicated that the minimum cytotoxic concentration of FNE in A549 cells was 20 µg/mL (data not shown). After treatment, the medium was removed, and cells were washed with ice-cold phosphate-buffered saline (PBS). Cells were lysed using a radioimmunoprecipitation assay (RIPA) buffer containing protease inhibitor tablets, and the mixture was incubated on ice for 15 minutes. The lysate was then centrifuged at 4 °C for 15 minutes at 15,000 g to remove debris. The supernatant was collected for protein concentration determination using the Pierce™ BCA Assay Kit. The influence of FNE on cancer-related protein expression was analyzed using the Proteome Profiler Human XL Oncology Array Kit. Each array membrane was loaded with 300 µg of protein, hybridized, processed, and imaged using a ChemiDoc™ MP imaging system. Pixel intensity for each protein was measured using ImageJ software, and statistical analysis was performed using PRISM software (Paudel et al., 2024[34]).

The results of this study demonstrate that treating A549 cells with 10 µg/mL FNE can significantly reduce the expression levels of three key proteins involved in the pathogenesis of lung cancer, specifically in cancer invasion and metastasis: CTSB, enolase, and GAL3. While some studies have already explored the link between fisetin and its effect on these markers (Hsieh et al., 2019[17]; Kumar et al., 2022[24]; Malathi et al., 2022[29]; Singh et al., 2018[43]; Zhang and Cui, 2021[56]), only a limited number of in vitro studies have been conducted on lung cancer in this context, making the present study unique. For instance, GAL3, a protein encoded by the LGALS3 gene, is known to play diverse roles in cancer initiation, progression, and drug resistance, as discussed in an earlier section (Capone et al., 2021[8]; Zhang et al., 2021[55]). Kusuhara et al. (2021[26]) demonstrated that the high GAL3 protein expression in tumor cells is associated with poor prognosis in non-small cell lung cancer (NSCLC) (Kusuhara et al., 2021[26]). Moreover, GAL3 has been identified as a predictive factor for recurrence-free survival (RFS) in NSCLC patients receiving platinum-based adjuvant chemotherapy (AC) (Kusuhara et al., 2021[26]). Our observation of decreased GAL3 expression following FNE treatment (Supplementary informationexcli2024-7583_supplementary_information.pdf, Figure 1c) aligns with the notion that targeting GAL3 may hold therapeutic promise in lung cancer management (Kusuhara et al., 2021[26]; Torres‐Martínez et al., 2024[47]). In addition, squamous cell carcinoma of the lung is recognized as an aggressive disease with a poor prognosis. While the majority of patients do not survive longer than five years, a minority show prolonged survival without disease progression (Pokhare et al., 2022[35]). According to Pokhare and colleagues (2022[35]), the identification of biomarkers, such as GAL3, using easily available immunohistochemical assays could improve risk stratification in lung cancer patients (Pokhare et al., 2022[35]), highlighting the importance of considering this biomarker even more in future studies.

Research has also shown that most malignant cells can increase the transcriptional activation of CTSB, an intracellular catabolic protein involved in the processing of antigens in the immune response. For instance, Sloane et al. (1981[44]) have indicated a strong connection between the activity of CTSB and the aggressive metastatic nature of pancreatic cancer and melanoma (Sloane et al., 1981[44]). In pancreatic cancer, CTSB is crucial in fostering tumor growth, angiogenesis, and invasion (Tzanakakis et al., 2003[49]). It is speculated that Matrix Metalloproteinases (MMPs), enzymes that break down proteins in the extracellular matrix (ECM), are activated or shed into the ECM via the action of CTSB and urokinase-type plasminogen activator (uPA). Essentially, CTSB is crucial in managing the protease cascade and guiding it to the invasive edges of metastatic cells (Gondi and Rao, 2013[11]). This pattern of elevated CTSB expression has also been reported for lung cancer, where it is associated with increased tumor aggressiveness and poorer patient prognosis, potentially hindering the prediction of patients' response to chemotherapy (Gormley et al., 2011[12]; Harbeck et al., 2001[14]; Werle et al., 1999[52]). Moreover, a molecular docking study analyzed the interaction of fisetin with different immune-related proteins and biomarkers, concluding that CTSB had one of the strongest predicted interactions with fisetin, exhibiting the lowest binding energy (Malathi et al., 2022[29]). The latter two points, (i) the relevance of CTSB in lung cancer, and (ii) the potential of fisetin in interacting with CTSB, highlight the significance of the results of the present study (Supplementary informationexcli2024-7583_supplementary_information.pdf, Figure 1a), suggesting the potential of fisetin as suitable treatment for lung cancer through targeting the CTSB expression level.

Furthermore, the role of enolase, an enzyme catalyzing an important glycolytic pathway reaction, has been extensively linked to lung cancer, as discussed in an earlier section (Schofield et al., 2020[40]; Tian et al., 2020[46]). However, this role is primarily in LC diagnosis as a preventive value (Xu et al., 2019[54]). According to many studies, evaluating the expression of enolase aids in lung cancer diagnosis, distinguishing between different subtypes, and assessing tumor characteristics (Fujita et al., 1987[10]; Huang et al., 2017[18]; Kostovski and Petrushevska, 2014[22]; Wang et al., 2016[51]). Serum enolase levels correlate with tumor size, stage, and metastasis in small-cell lung cancer (SCLC) (Huang et al., 2016[19]). Furthermore, enolase levels serve as a sensitive marker for monitoring therapy and predicting relapse, guiding treatment decisions (Hirose et al., 2011[15]; Nitta et al., 1995[32]). With substantial evidence highlighting the significance of enolase in lung cancer, therapy strategies are now considering targeting this biomarker to reduce its levels, especially in SCLC (Tian et al., 2020[46]). A recent study found that fisetin can trigger apoptosis in cancer cells through multiple pathways, many of which are supported by enolase activity. These pathways include promoting proteasomal degradation and reducing the half-life of specific proteins, deactivating receptors, decreasing enolase phosphorylation, and modifying PI3K/AKT signaling (Guo et al., 2019[13]). Inhibiting enolase activity could disrupt these metabolic pathways that support tumor growth and metastasis (Rahmani et al., 2022[37]), offering a potential strategy for lung cancer treatment. This comprehensive logic behind the role of enolase in lung cancer and the possible effect of fisetin in targeting it aligns with the results (shown in Supplementary informationexcli2024-7583_supplementary_information.pdf, Figure 1b), where the expression level of enolase in A549 cells significantly decreased after treatment with FNE.

Although the result of the present study shows that 10 µg/mL of fisetin nano emulsion can significantly reduce the expression levels of relevant markers such as CTSB, enolase, and GAL3 in lung cancer cells, we also have several limitations to consider. The study's reliance on an in vitro model restricts the extrapolation of these findings to in vivo conditions, where factors such as metabolism, immune response, and tissue-specific interactions may influence treatment outcomes differently (Antunes et al., 2022[5]; Malyla et al., 2020[30]). Furthermore, while the investigation targeted key markers associated with lung cancer progression, the complexity of lung cancer's molecular pathways suggests that other important biomarkers and pathways may not have been comprehensively examined. Therefore, further research employing diverse experimental models and clinical trials is essential to validate these results and ascertain fisetin nano emulsions broader therapeutic potential and safety profile in lung cancer treatment.

Notes

Ayeh Bani Saeid and Keshav Raj Paudel contributed equally as first author.

Sachin Kumar Singh and Kamal Dua (Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; E-mail: kamal.dua@uts.edu.au) contributed equally as corresponding author.

Declaration

Acknowledgments

The authors are thankful to the Graduate School of Health, University of Technology Sydney, Australia. KD, GDR, and KRP are supported by the Maridulu Budyari Gumal Sydney Partnership for Health, Education, Research and Enterprise (SPHERE) RSEOH CAG and Triple I Seed Grant 2023 and Extension Grant, Triple I Skills Advancement Grants 2023 and 2024, and the UTS Faculty of Health Industry Partnership Grant. GDR is also supported by the UTS Faculty of Health Category 1 Seeding grants for Early Career Researchers.

Declaration of competing interest

The authors declare that there is no conflict of interest regarding the publication of this letter.

 

References

1. Alharbi KS, Shaikh MAJ, Afzal O, Altamimi ASA, Almalki WH, Alzarea SI, et al. An overview of epithelial growth factor receptor (EGFR) inhibitors in cancer therapy. Chem Biol Interact. 2022;366:110108. doi: 10.1016/j.cbi.2022.110108
2. Alharbi KS, Shaikh MAJ, Almalki WH, Kazmi I, Al-Abbasi FA, Alzarea SI, et al. PI3K/Akt/mTOR pathways inhibitors with potential prospects in non-small-cell lung cancer. J Environ Pathol Toxicol Oncol. 2022;41(4):85-102. doi: 10.1615/JEnvironPatholToxicolOncol.2022042281
3. Alnuqaydan AM, Almutary AG, Azam M, Manandhar B, De Rubis G, Madheswaran T, et al. Phytantriol-based berberine-loaded liquid crystalline nanoparticles attenuate inflammation and oxidative stress in lipopolysaccharide-induced RAW264.7 macrophages. Nanomaterials (Basel). 2022;12(23):4312. doi: 10.3390/nano12234312
4. Alnuqaydan AM, Almutary AG, Azam M, Manandhar B, Yin GHS, Yen LL, et al. Evaluation of the cytotoxic activity and anti-migratory effect of berberine-phytantriol liquid crystalline nanoparticle formulation on non-small-cell lung cancer in vitro. Pharmaceutics. 2022;14(6):1119. doi: 10.3390/pharmaceutics14061119
5. Antunes N, Kundu B, Kundu SC, Reis RL, Correlo V. In vitro cancer models: a closer look at limitations on translation. Bioengineering. 2022;9(4):166. doi: 10.3390/bioengineering9040166
6. Barta JA, Powell CA, Wisnivesky JP. Global epidemiology of lung cancer. Ann Global Health. 2019;85(1):8. doi: 10.5334/aogh.2419
7. Cancer Australia. Lung cancer in Australia statistics 2019. Available from: https://www.canceraustralia.gov.au/cancer-types/lung-cancer/statistics (Access date 27/09/2024)
8. Capone E, Iacobelli S, Sala G. Role of galectin 3 binding protein in cancer progression: a potential novel therapeutic target. J Transl Med. 2021;19(1):405. doi: 10.1186/s12967-021-03085-w
9. Chang JT, Anic GM, Rostron BL, Tanwar M, Chang CM. Cigarette smoking reduction and health risks: a systematic review and meta-analysis. Nicotine Tob Res. 2021;23:635-42. doi: 10.1093/ntr/ntaa156
10. Fujita K, Haimoto H, Imaizumi M, Abe T, Kato K. Evaluation of γ-enolase as a tumor marker for lung cancer. Cancer. 1987;60:362-9. doi: 10.1002/1097-0142(19870801)60:3<362::aid-cncr2820600313>3.0.co;2-u
11. Gondi CS, Rao JS. Cathepsin B as a cancer target. Expert Opin Ther Targets. 2013;17:281-91. doi: 10.1517/14728222.2013.740461
12. Gormley JA, Hegarty SM, O'Grady A, Stevenson MR, Burden RE, Barrett HL, et al. The role of Cathepsin S as a marker of prognosis and predictor of chemotherapy benefit in adjuvant CRC: a pilot study. Br J Cancer. 2011;105:1487-94. doi: 10.1038/bjc.2011.408
13. Guo G, Zhang W, Dang M, Yan M, Chen Z. Fisetin induces apoptosis in breast cancer MDA-MB-453 cells through degradation of HER2/neu and via the PI3K/Akt pathway. J Biochem Mol Toxicol. 2019;33 (4):e22268. doi: 10.1002/jbt.22268
14. Harbeck N, Alt U, Berger U, Krüger A, Thomssen C, Jänicke F, et al. Prognostic impact of proteolytic factors (urokinase-type plasminogen activator, plasminogen activator inhibitor 1, and cathepsins B, D, and L) in primary breast cancer reflects effects of adjuvant systemic therapy. Clin Cancer Res. 2001;7:2757-64
15. Hirose T, Okuda K, Yamaoka T, Ishida K, Kusumoto S, Sugiyama T, et al. Are levels of pro-gastrin-releasing peptide or neuron-specific enolase at relapse prognostic factors after relapse in patients with small-cell lung cancer? Lung Cancer. 2011;71:224-8. doi: 10.1016/j.lungcan.2010.05.004
16. Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Wu Y-L, et al. Lung cancer: current therapies and new targeted treatments. The Lancet. 2017;389(10066):299-311. doi: 10.1016/s0140-6736(16)30958-8
17. Hsieh MH, Tsai JP, Yang SF, Chiou HL, Lin CL, Hsieh YH, et al. Fisetin suppresses the proliferation and metastasis of renal cell carcinoma through upregulation of MEK/ERK-targeting CTSS and ADAM9. Cells. 2019;8(9):948. doi: 10.3390/cells8090948
18. Huang L, Zhou JG, Yao WX, Tian X, Lv SP, Zhang TY, et al. Systematic review and meta-analysis of the efficacy of serum neuron-specific enolase for early small cell lung cancer screening. Oncotarget. 2017;8(38):64358. doi: 10.18632/oncotarget.17825
19. Huang Z, Xu D, Zhang F, Ying Y, Song L. Pro-gastrin-releasing peptide and neuron-specific enolase: useful predictors of response to chemotherapy and survival in patients with small cell lung cancer. Clin Transl Oncol. 2016;18:1019-25. doi: 10.1007/s12094-015-1479-4
20. Imran M, Saeed F, Gilani SA, Shariati MA, Imran A, Afzaal M, et al. Fisetin: An anticancer perspective. Food Sci Nutr. 2021;9(1):3-16. doi: 10.1002/fsn3.1872
21. Koni M, Pinnarò V, Brizzi MF. The Wnt signalling pathway: a tailored target in cancer. Int J Mol Sci. 2020;21(20):7697. doi: 10.3390/ijms21207697
22. Kostovski M, Petrushevska G. Antigenic phenotype of lung carcinomas: usual spectrum of distribution of thyroid transcription factor-1, cytokeratin 7, cytokeratin 20, and neuron specific enolase - basic immunohistochemical study of 21 cases. Pril (Makedon Akad Nauk Umet Odd Med Nauki). 2014;35(1):199-207
23. Kumar R, Khursheed R, Kumar R, Awasthi A, Sharma N, Khurana S, et al. Self-nanoemulsifying drug delivery system of fisetin: Formulation, optimization, characterization and cytotoxicity assessment. J Drug Deliv Sci Technol. 2019;54:101252. doi: 10.1016/j.jddst.2019.101252
24. Kumar R, Kumar R, Khurana N, Singh SK, Khurana S, Verma S, et al. Improved neuroprotective activity of Fisetin through SNEDDS in ameliorating the behavioral alterations produced in rotenone-induced Parkinson’s model. Environ Sci Pollut Res. 2022;29:50488-99. doi: 10.1016/j.jddst.2019.101252
25. Kumar RM, Kumar H, Bhatt T, Jain R, Panchal K, Chaurasiya A, et al. Fisetin in cancer: attributes, developmental aspects, and nanotherapeutics. Pharmaceuticals. 2023;16(2):196. doi: 10.3390/ph16020196
26. Kusuhara S, Igawa S, Ichinoe M, Nagashio R, Kuchitsu Y, Hiyoshi Y, et al. Prognostic significance of galectin-3 expression in patients with resected NSCLC treated with platinum‐based adjuvant chemotherapy. Thorac Cancer. 2021;12:1570-8. doi: 10.1111/1759-7714.13945
27. Ling J, Wang Y, Ma L, Chang A, Meng L, Zhang L. Exploration of potential targets and mechanisms of fisetin in the treatment of non-small-cell lung carcinoma via network pharmacology and in vitro validation. Evid Based Complement Alternat Med. 2022;2022:2383527. doi: 10.1155/2022/2383527
28. Liu WY, Lin CC, Hsieh YS, Wu YT. Nanoformulation development to improve the biopharmaceutical properties of fisetin using design of experiment approach. Molecules. 2021;26(10):3031. doi: 10.3390/molecules26103031
29. Malathi R, Dsouza V, Rithika R, Sneha P. Molecular docking of fisetin as a multi-target drug in the treatment of multiple sclerosis. J Stress Physiol Biochem. 2022;18(4):41-7
30. Malyla V, Paudel KR, Shukla SD, Donovan C, Wadhwa R, Pickles S, et al. Recent advances in experimental animal models of lung cancer. Fut Med Chem. 2020;12:567-70. doi: 10.4155/fmc-2019-0338
31. Mehta M, Dhanjal DS, Paudel KR, Singh B, Gupta G, Rajeshkumar S, et al. Cellular signalling pathways mediating the pathogenesis of chronic inflammatory respiratory diseases: an update. Inflammopharmacology. 2020;28:795-817. doi: 10.1007/s10787-020-00698-3-
32. Nitta T, Fukuoka M, Masuda N, Kusunoki Y, Matsui K, Kudoh S, et al. Significance of serum neuron-specific enolase as a predictor of relapse of small cell lung cancer. Jap J Clin Oncol. 1995;25:179-83
33. Paudel KR, De Rubis G, Panth N, Singh SK, Chellappan DK, Hansbro PM, et al. Nanomedicine and medicinal plants: Emerging symbiosis in managing lung diseases and associated infections. EXCLI J. 2022;21:1299-303. doi: 10.17179/excli2022-5376
34. Paudel KR, Mohamad MSB, De Rubis G, Reyes R-J, Panth N, Dureja H, et al. 18-β-Glycyrrhetinic acid encapsulated PLGA nanoparticles attenuate lung cancer proliferation and migration. J Drug Deliv Sci Technol. 2024;95:105523. doi: 10.1016/j.prp.2024.155295
35. Pokhare S, Sharma UC, Attwood K, Mansoor S. Clinical significance of galectin-3 expression in squamous cell carcinoma of lung. J Cancer Sci Clin Ther. 2022;6:32-7. doi: 10.26502/jcsct.5079169
36. Prasher P, Sharma M, Mehta M, Paudel KR, Satija S, Chellappan DK, et al. Plants derived therapeutic strategies targeting chronic respiratory diseases: Chemical and immunological perspective. Chem Biol Interact. 2020;325:109125. doi: 10.1016/j.cbi.2020.109125
37. Rahmani AH, Almatroudi A, Allemailem KS, Khan AA, Almatroodi SA. The potential role of fisetin, a flavonoid in cancer prevention and treatment. Molecules. 2022;27(24):9009. doi: 10.3390/molecules27249009
38. Ranjbar S, Emamjomeh A, Sharifi F, Zarepour A, Aghaabbasi K, Dehshahri A, et al. Lipid-based delivery systems for flavonoids and flavonolignans: liposomes, nanoemulsions, and solid lipid nanoparticles. Pharmaceutics. 2023;15(7):1944. doi: 10.3390/pharmaceutics15071944
39. Saeid AB, Patel VK, Mehndiratta S, Rajput R, Kundu RK, Singh SK, et al. Dissecting the in vitro fate of plant-derived bioactive encapsulated nanoparticles in lung diseases. Food Bioscience. 2023;56:103205. doi: 10.1016/j.fbio.2023.103205
40. Schofield L, Lincz LF, Skelding KA. Unlikely role of glycolytic enzyme α-enolase in cancer metastasis and its potential as a prognostic biomarker. J Cancer Metastasis Treat. 2020;6:10. doi: 10.20517/2394-4722.2019.43
41. Sharma P, Mehta M, Dhanjal DS, Kaur S, Gupta G, Singh H, et al. Emerging trends in the novel drug delivery approaches for the treatment of lung cancer. Chem Biol Interact. 2019;309:108720. doi: 10.1016/j.cbi.2019.06.033
42. Singh S, Saxena S, Sharma H, Paudel KR, Chakraborty A, MacLoughlin R, et al. Emerging role of tumor suppressing microRNAs as therapeutics in managing Non-small cell lung cancer. Pathol Res Pract. 2024;256:155222. doi: 10.1016/j.prp.2024.155222
43. Singh S, Singh AK, Garg G, Rizvi SI. Fisetin as a caloric restriction mimetic protects rat brain against aging induced oxidative stress, apoptosis and neurodegeneration. Life Sci. 2018;193:171-9. doi: 10.1016/j.lfs.2017.11.004
44. Sloane BF, Dunn JR, Honn KV. Lysosomal cathepsin B: correlation with metastatic potential. Science. 1981;212(4499):1151-3. doi: 10.1126/science.7233209
45. Szymczak J, Cielecka-Piontek J. Fisetin-in search of better bioavailability-from macro to nano modifications: a review. Int J Mol Sci. 2023;24(18):14158. doi: 10.3390/ijms241814158
46. Tian Z, Liang C, Zhang Z, Wen H, Feng H, Ma Q et al. Prognostic value of neuron-specific enolase for small cell lung cancer: a systematic review and meta-analysis. World J Surg Oncol. 2020;18(1):116. doi: 10.1186/s12957-020-01894-9
47. Torres-Martínez S, Calabuig-Fariñas S, Moreno-Manuel A, Bertolini G, Herreros-Pomares A, Escorihuela E, et al. Soluble galectin-3 as a microenvironment-relevant immunoregulator with prognostic and predictive value in lung adenocarcinoma. Mol Oncol. 2024;18(1):190-215. doi: 10.1002/1878-0261.13505
48. Travis WD. Lung cancer pathology: current concepts. Clin Chest Med. 2020;41:67-85. doi: 10.1016/j.ccm.2019
49. Tzanakakis GN, Margioris AN, Tsatsakis AM, Vezeridis MP. The metastatic potential of human pancreatic cell lines in the liver of nude mice correlates well with cathepsin B activity. Int J Gastrointest Cancer. 2003;34:27-38. doi: 10.1385/ijgc:34:1:27
50. Vishwas S, Singh SK, Gulati M, Awasthi A, Khursheed R, Corrie L, et al. Harnessing the therapeutic potential of fisetin and its nanoparticles: Journey so far and road ahead. Chem Biol Interact. 2022;356:109869. doi: 10.1016/j.cbi.2022.109869
51. Wang L, Wang D, Zheng G, Yang Y, Du L, Dong Z, et al. Clinical evaluation and therapeutic monitoring value of serum tumor markers in lung cancer. Int J Biol Mark. 2016;31(1):80-7. doi: 10.5301/jbm.5000177
52. Werle B, Lötterle H, Schanzenbächer U, Lah T, Kalman E, Kayser K, et al. Immunochemical analysis of cathepsin B in lung tumours: an independent prognostic factor for squamous cell carcinoma patients. Br J Cancer. 1999;81:510-9. doi: 10.1038/sj.bjc.6690723
53. WHO, World Health Orgainzation. Lung cancer 2023. Geneva: WHO, 2023. Available from: https://www.who.int/news-room/fact-sheets/detail/lung-cancer (Access date 27/092024)
54. Xu CM, Luo YL, Li S, Li ZX, Jiang L, Zhang G-X, et al. Multifunctional neuron-specific enolase: its role in lung diseases. Biosci Rep. 2019;39(11):BSR20192732. doi: 10.1042/BSR20192732
55. Zhang H, Liu P, Zhang Y, Han L, Hu Z, Cai Z, et al. Inhibition of galectin-3 augments the antitumor efficacy of PD-L1 blockade in non‐small‐cell lung cancer. FEBS Open Bio. 2021;11:911-20. doi: 10.1002/2211-5463.13088
56. Zhang P, Cui J. Neuroprotective effect of fisetin against the cerebral ischemia-reperfusion damage via suppression of oxidative stress and inflammatory parameters. Inflammation. 2021;44:1490-506. doi: 10.1007/s10753-021-01434-x
 
 

File-Attachments

  1. excli2024-7583_supplementary_information.pdf (254,23 KB)
    Supplementary information
 
 
 

[*] Corresponding Author:

Sachin Kumar Singh, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, 144411, India, eMail: sachin.16030@lpu.co.in, singhsachin23@gmail.com