PREPARATION, CHARACTERIZATION, AND IN VITRO ANTI CANCER ACTIVITY EVALUATION OF IRINOTECAN AND SUNITINIB-LOADED NANOBUBBLES

Authors

  • ANJANEYULU PATAMSETTI GITAM School of Pharmacy, GITAM Deemed to be University, Hyderabad-502329, Telangana, India https://orcid.org/0009-0004-2246-1390
  • KUMAR SHIVA GUBBIYAPPA GITAM School of Pharmacy, GITAM Deemed to be University, Hyderabad-502329, Telangana, India

DOI:

https://doi.org/10.22159/ijap.2025v17i3.53698

Keywords:

Nanobubbles, Irinotecan, Sunitinib, Toxicity, Cell lines, Cancer

Abstract

Objective: The objective of the present research is to develop, analyze, and evaluate the cytotoxic effects of nanobubbles loaded with irinotecan and sunitinib for colorectal cancer therapy.

Methods: Drug-loaded dextran sulfate nanobubbles were formulated using the emulsification technique and the prepared nanobubbles were evaluated for qualitative and quantitative parameters. Cell viability MTT assay was performed to evaluate the irinotecan and sunitinib-loaded nanobubbles for cytotoxicity or ability to inhibit cell proliferation spectrophotometrically as a function of mitochondrial activity in living CR4 and A-549(irinotecan), CR4 and A-498 (sunitinib) cell lines.

Results: The irinotecan and sunitinib-loaded nanobubbles were successfully prepared, with all qualitative and quantitative parameters within the acceptable range. The optimized nanobubbles demonstrated excellent physicochemical properties, with particle sizes of 177.8±5.2 nm for sunitinib and 89.8±9.5 nm for irinotecan. In vitro drug release studies showed significantly enhanced release profiles, with ultrasound-triggered drug release reaching 99% for both drugs, compared to only 39% for plain sunitinib and 35% for plain irinotecan. This highlights the ability of nanobubbles to enable controlled and targeted drug delivery, potentially improving therapeutic precision. The in vitro anticancer activity results revealed IC50 values of 70.41 and 73.26 µg/ml for irinotecan against CR4 and A-549 cell lines, respectively, and 84.34 and 60.08 µg/ml for sunitinib against CR4 and A-498 cell lines, respectively, demonstrating strong cytotoxic effects.

Conclusion: The nanobubble-based delivery system enhances drug bioavailability, cellular uptake, and tumor penetration, thereby improving cytotoxic efficacy compared to conventional drug formulations. These findings underscore the potential of ultrasound-responsive nanobubbles as a promising strategy for targeted colorectal cancer therapy, potentially leading to improved treatment outcomes with reduced systemic toxicity.

References

Dekker E, Tanis PJ, Vleugels JL, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019 Oct;394(10207):1467-80. doi: 10.1016/S0140-6736(19)32319-0, PMID 31631858.

Klimeck L, Heisser T, Hoffmeister M, Brenner H. Colorectal cancer: a health and economic problem. Best Pract Res Clin Gastroenterol. 2023 Oct;66:101839. doi: 10.1016/j.bpg.2023.101839, PMID 37852707.

Konopke R, Schubert J, Stoltzing O, Thomas T, Kersting S, Denz A. Palliative surgery in colorectal cancer which factors should influence the choice of the surgical procedure? [Palliative surgery in colorectal cancer: which factors should influence the choice of the surgical procedure?]. Zentralbl Chir. 2021 Feb;146(1):44-57. doi: 10.1055/a-1291-8293, PMID 33296936.

Kciuk M, Marciniak B, Kontek R. Irinotecan still an important player in cancer chemotherapy: a comprehensive overview. Int J Mol Sci. 2020 Jul;21(14):4919. doi: 10.3390/ijms21144919, PMID 32664667.

Jin J, Xie Y, Zhang JS, Wang JQ, Dai SJ, HE WF. Sunitinib resistance in renal cell carcinoma: from molecular mechanisms to predictive biomarkers. Drug Resist Updat. 2023 Mar;67:100929. doi: 10.1016/j.drup.2023.100929, PMID 36739809.

Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther. 2023 Aug 7;8(1):293. doi: 10.1038/s41392-023-01536-y, PMID 37544972.

Garg NK, Tandel N, Jadon RS, Tyagi RK, Katare OP. Lipid polymer hybrid nanocarrier mediated cancer therapeutics: current status and future directions. Drug Discov Today. 2018 Sep;23(9):1610-21. doi: 10.1016/j.drudis.2018.05.033, PMID 29857164.

Munir M, Zaman M, Waqar MA, Khan MA, Alvi MN. Solid lipid nanoparticles: a versatile approach for controlled release and targeted drug delivery. J Liposome Res. 2024 Jun;34(2):335-48. doi: 10.1080/08982104.2023.2268711, PMID 37840238.

Dadhaniya T, Chaudhary K, Mehta P. Development of LC-MS/MS method for determination of iloperidone in rabbit plasma: application to a pharmacokinetic study. Int J Pharm Pharm Sci. 2013 Apr;7(4):294-7.

Miura Y, Fuchigami Y, Hagimori M, Sato H, Ogawa K, Munakata C. Evaluation of the targeted delivery of 5-fluorouracil and ascorbic acid into the brain with ultrasound responsive nanobubbles. J Drug Target. 2018 Sep;26(8):684-91. doi: 10.1080/1061186X.2017.1419354, PMID 29251518.

Zhang Y, LI X, Wang X. Idarubicin loaded chitosan nanobubbles to improve survival and reduce side effects in leukemia treatment. Drug Deliv. 2025 Jan 15;32(1):123-34.

Kumar MK, Prakash DJ, Rao VV. Chitosan nanobubbles development and evaluation for the delivery of sunitinib an anticancer agent. Int J App Pharm. 2022 Jun;14(6):58-67. doi: 10.22159/ijap.2022v14i6.45821.

LI J, WU K, Zhang J, Gao H, XU X. Progress in the treatment of drug-loaded nanomaterials in renal cell carcinoma. Biomed Pharmacother. 2023 Nov;167:115444. doi: 10.1016/j.biopha.2023.115444, PMID 37716114.

Buranaamnuay K. The MTT assay application to measure the viability of spermatozoa: a variety of the assay protocols. Open Vet J. 2021 Apr-Jun;11(2):251-69. doi: 10.5455/OVJ.2021.v11.i2.9, PMID 34307082.

Kumar P, Nagarajan A, Uchil PD. Analysis of cell viability by the MTT assay. Cold Spring Harb Protoc. 2018 Jun;2018(6). doi: 10.1101/pdb.prot095505, PMID 29858338.

Hema AN, Gaayathri G, Gundeti S. Development of orodispersible tablets of loratadine containing an amorphous solid dispersion of the drug in soluplus® using design of experiments. Int J Pharm Pharm Sci. 2023 Aug;15(8):19-27.

Khorsandi L, Orazizadeh M, Niazvand F, Abbaspour MR, Mansouri E, Khodadadi A. Quercetin induces apoptosis and necroptosis in MCF-7 breast cancer cells. Bratisl Lek Listy. 2017 Feb;118(2):123-8. doi: 10.4149/BLL_2017_025, PMID 28814095.

Arafath AA, Jayakar B. Enhancement of oral bioavailability via solid lipid nanoparticles of anticancer drug dasatinib an in vitro cytotoxicity and pharmacokinetic study. Asian J Pharm Clin Res. 2019 Jun;12(6):143-5.

Cheng X, Tan S, Duan F, Yuan Q, LI Q, Deng G. Icariin induces apoptosis by suppressing autophagy in tamoxifen-resistant breast cancer cell line MCF-7/TAM. Breast Cancer. 2019 Nov;26(6):766-75. doi: 10.1007/s12282-019-00980-5, PMID 31172425.

XU B, Peng YJ, Zhu WJ. Curcumin inhibits viability of clear cell renal cell carcinoma by down-regulating ADAMTS18 gene methylation though NF-κ B and AKT signaling pathway. Chin J Integr Med. 2022 May;28(5):419-24. doi: 10.1007/s11655-021-3445-z, PMID 33997938.

Tang X, Zhao Q, Liu J, Wang S, Zhang N, Yang Y. The compound AST-003 could effectively promote apoptosis of renal cell carcinoma cells in vitro. Transl Cancer Res. 2021 May;10(5):2120-33. doi: 10.21037/tcr-20-3330, PMID 35116532.

Anil L, Mohandas S. In vitro antioxidant and anticancer activity of macrangapeltata leaf extracts on lung cancer cell lines. Int J Curr Pharm Res. 2023 Apr;15(4):26-32.

Published

07-05-2025

How to Cite

PATAMSETTI, A., & GUBBIYAPPA, K. S. (2025). PREPARATION, CHARACTERIZATION, AND IN VITRO ANTI CANCER ACTIVITY EVALUATION OF IRINOTECAN AND SUNITINIB-LOADED NANOBUBBLES. International Journal of Applied Pharmaceutics, 17(3), 383–389. https://doi.org/10.22159/ijap.2025v17i3.53698

Issue

Section

Original Article(s)

Similar Articles

1 2 3 4 5 > >> 

You may also start an advanced similarity search for this article.