Skip to main content

Advertisement

Log in

Stimuli-responsive magnetic silica-poly-lactic-co-glycolic acid hybrid nanoparticles for targeted cancer chemo-immunotherapy

  • Original Article
  • Published:
Drug Delivery and Translational Research Aims and scope Submit manuscript

Abstract

Chemotherapy and immunotherapy are two important modalities in cancer management. However, due to multiple reasons, a monotherapy is only partially effective. Hence, if used concurrently in targeted and stimuli-responsive manner, it could have been superior therapeutically. To facilitate co-delivery of chemotherapeutic and immunotherapeutic agent to the target cancer cells, engineered nanoparticles, i.e., a pH-responsive polymer PLGA-coated magnetic silica nanoparticles (Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs) encapsulating paclitaxel (PTX) and siRNA against programmed cell death ligand-1 (PD-L1) are synthesized and characterized. Developed nanoparticles demonstrated pH-sensitive sustained drug release up to 10 days. In vitro 4T1 cell line studies showed efficient cellular uptake, PD-L1 gene downregulation, and apoptosis. Further, in vivo efficacy studies carried out in the mice model demonstrated a significant reduction of tumor growth following treatment with dual-Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs as compared with monotherapy with Fe3O4-SiO2-PLGA-PDA-PTX NPs. The high therapeutic efficacy observed with dual-Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs was mainly due to the cytotoxic effect of PTX combined with targeted silencing of the gene of interest, i.e., PD-L1, which in turn improve CD8+ T cell-mediated cancer cell death as evident with increased proliferation of CD8+ T cells in co-culture experiments. Thereby, dual-Fe3O4-SiO2-PLGA-PDA-PTX-siRNA NPs may have a promising anti-cancer treatment potential against breast cancer; however, the beneficial effects of dual loading of PTX + PD-L1 siRNA may be corroborated against other cancer models such as lung and colorectal cancer models as well as in clinical trials.

Graphical Abstract

Nanoparticles targeting to the tumor site, pH-specific release of PTX and PD-L1 siRNA by dual-loaded fabricated nanoparticles within tumor cells for enhanced chemo-immunotherapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Data availability

All the materials used in the manuscript are commercially available; however, 4T1 cells can be requested from Prof. Avinash Bajaj, Regional Centre of Biotechnology, Faridabad, India. Data required for results and conclusion has included in the manuscript however, raw data will be provided by the corresponding author on reasonable request.

References

  1. Bailly C, Thuru X, Quesnel B. Combined cytotoxic chemotherapy and immunotherapy of cancer: modern times. NAR Cancer. 2020 Feb 17;2(1):zcaa002. https://doi.org/10.1093/narcan/zcaa002. PMID: 34316682; PMCID: PMC8209987.

  2. Marupudi NI, et al. Paclitaxel: a review of adverse toxicities and novel delivery strategies. Expert Opin Drug Saf. 2007;6(5):609–21.

    Article  CAS  PubMed  Google Scholar 

  3. Chou PL, Huang YP, Cheng MH, Rau KM, Fang YP. Improvement of Paclitaxel-Associated Adverse Reactions (ADRs) via the Use of Nano-Based Drug Delivery Systems: A Systematic Review and Network Meta-Analysis. Int J Nanomedicine. 2020 Mar 12;15:1731–1743. https://doi.org/10.2147/IJN.S231407. PMID: 32210563; PMCID: PMC7075337.

  4. Zhang H, et al. Iridium oxide nanoparticles-based theranostic probe for in vivo tumor imaging and synergistic chem/photothermal treatments of cancer cells. Chem Eng J. 2022;430:132675.

    Article  CAS  Google Scholar 

  5. Zhou Y, et al. Upconverting nanoparticles based nanodevice for DNAzymes amplified miRNAs detection and artificially controlled chemo-gene therapy. Biosens Bioelectron. 2022;214:114549.

    Article  CAS  PubMed  Google Scholar 

  6. Qiu L, et al. A targeted, self-delivered, and photocontrolled molecular beacon for mRNA detection in living cells. J Am Chem Soc. 2013;135(35):12952–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yan J, et al. Therapeutic DNAzymes: from structure design to clinical applications. Adv Mater. 2023: p. 2300374.

  8. Mueller SN, et al. PD-L1 has distinct functions in hematopoietic and nonhematopoietic cells in regulating T cell responses during chronic infection in mice. J Clin Investig. 2010;120(7):2508–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bartlett DW, Davis ME. Effect of siRNA nuclease stability on the in vitro and in vivo kinetics of siRNA-mediated gene silencing. Biotechnol Bioeng. 2007;97(4):909–21.

    Article  CAS  PubMed  Google Scholar 

  10. Volkov AA, et al. Selective protection of nuclease-sensitive sites in siRNA prolongs silencing effect. Oligonucleotides. 2009;19(2):191–202.

    Article  CAS  PubMed  Google Scholar 

  11. Lee JM, Yoon TJ, Cho YS. Recent developments in nanoparticle-based siRNA delivery for cancer therapy. Biomed Res Int. 2013;2013:782041. https://doi.org/10.1155/2013/782041. Epub 2013 Jun 17. PMID: 23844368; PMCID: PMC3703404.

  12. Tomar RS, Matta H, Chaudhary PM. Use of adeno-associated viral vector for delivery of small interfering RNA. Oncogene. 2003;22(36):5712–5.

    Article  CAS  PubMed  Google Scholar 

  13. Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003;4(5):346–58.

    Article  CAS  PubMed  Google Scholar 

  14. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res. 2015;3(5):436–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hanoteau A, Moser M. Chemotherapy and immunotherapy: a close interplay to fight cancer? Oncoimmunology. 2016;5(7):e1190061.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Attia MF, et al. An overview of active and passive targeting strategies to improve the nanocarriers efficiency to tumour sites. J Pharm Pharmacol. 2019;71(8):1185–98.

    Article  CAS  PubMed  Google Scholar 

  17. Zhang Y, Cao J, Yuan Z. Strategies and challenges to improve the performance of tumor-associated active targeting. J Mater Chem B. 2020;8(18):3959–71.

    Article  CAS  PubMed  Google Scholar 

  18. Thiramanas R, et al. Cellular uptake of siRNA-loaded nanocarriers to knockdown PD-L1: strategies to improve T-cell functions. Cells. 2020;9(9):2043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wu Y, et al. Silencing PD-1 and PD-L1 with nanoparticle-delivered small interfering RNA increases cytotoxicity of tumor-infiltrating lymphocytes. Nanomedicine. 2019;14(8):955–67.

    Article  CAS  PubMed  Google Scholar 

  20. Islam T, Josephson L. Current state and future applications of active targeting in malignancies using superparamagnetic iron oxide nanoparticles. Cancer Biomark. 2009;5(2):99–107.

    Article  CAS  PubMed  Google Scholar 

  21. Chen L, et al. Magnetic targeting combined with active targeting of dual-ligand iron oxide nanoprobes to promote the penetration depth in tumors for effective magnetic resonance imaging and hyperthermia. Acta Biomater. 2019;96:491–504.

    Article  CAS  PubMed  Google Scholar 

  22. Palanisamy S, Wang YM. Superparamagnetic iron oxide nanoparticulate system: synthesis, targeting, drug delivery and therapy in cancer. Dalton Trans. 2019;48(26):9490–515.

    Article  CAS  PubMed  Google Scholar 

  23. Zhang MW, et al. Controlled fabrication of iron oxide/mesoporous silica core-shell nanostructures. J Phys Chem C. 2013;117(41):21529–38.

    Article  CAS  Google Scholar 

  24. Gupta A, et al. Nanocarrier composed of magnetite core coated with three polymeric shells mediates LCS-1 delivery for synthetic lethal therapy of BLM-defective colorectal cancer cells. Biomacromol. 2018;19(3):803–15.

    Article  MathSciNet  CAS  Google Scholar 

  25. Ahmad A, et al. Hyperbranched polymer-functionalized magnetic nanoparticle-mediated hyperthermia and niclosamide bimodal therapy of colorectal cancer cells. ACS Biomater Sci Eng. 2020;6(2):1102–11.

    Article  CAS  PubMed  Google Scholar 

  26. Hariani PL, et al. Synthesis and properties of Fe3O4 nanoparticles by co-precipitation method to removal procion dye. Int J Environ Sci Dev. 2013;4(3):336–40.

    Article  CAS  Google Scholar 

  27. Tang H, et al. Facile synthesis of pH sensitive polymer-coated mesoporous silica nanoparticles and their application in drug delivery. Int J Pharm. 2011;421(2):388–96.

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  28. Saini K, Bandyopadhyaya R. Transferrin-conjugated polymer-coated mesoporous silica nanoparticles loaded with gemcitabine for killing pancreatic cancer cells. ACS Applied Nano Materials. 2019;3(1):229–40.

    Article  Google Scholar 

  29. Wei Y, et al. Polydopamine and peptide decorated doxorubicin-loaded mesoporous silica nanoparticles as a targeted drug delivery system for bladder cancer therapy. Drug Delivery. 2017;24(1):681–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chan A, et al. Remote and local control of stimuli responsive materials for therapeutic applications. Adv Drug Deliv Rev. 2013;65(4):497–514.

    Article  CAS  PubMed  Google Scholar 

  31. Hernández-Hernández AA, et al. Iron oxide nanoparticles: synthesis, functionalization, and applications in diagnosis and treatment of cancer. Chem Pap. 2020;74(11):3809–24.

    Article  Google Scholar 

  32. Makadia HK, Siegel SJ. Poly lactic–glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers. 2011;3(3):1377–97.

    Article  CAS  PubMed  Google Scholar 

  33. Goyal AK, et al. Application and perspective of pH-responsive nano drug delivery systems. In: Applications of Targeted Nano Drugs and Delivery Systems. Elsevier; 2019. p. 15–33.

    Chapter  Google Scholar 

  34. Khing TM, et al. The effect of paclitaxel on apoptosis, autophagy and mitotic catastrophe in AGS cells. Sci Rep. 2021;11(1):23490.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Callahan MK, Postow MA, Wolchok JD. Targeting T cell co-receptors for cancer therapy. Immunity. 2016;44(5):1069–78.

    Article  CAS  PubMed  Google Scholar 

  36. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. 2020;10(3):727.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Chen S, et al. Combination of 4–1BB agonist and PD-1 antagonist promotes antitumor effector/memory CD8 T cells in a poorly immunogenic tumor model. Cancer Immunol Res. 2015;3(2):149–60.

    Article  CAS  PubMed  Google Scholar 

  38. Liang D, Kuang G., Chen X., Lu J., Shang L., Sun W. Near-infrared light-responsive Nitric oxide microcarrier for multimodal tumor therapy. Smart Medicine. 2023, 2(3), e20230016. https://doi.org/10.1002/SMMD.20230016.

  39. Fernandes S, et al. Magnetic nanoparticle-based hyperthermia mediates drug delivery and impairs the tumorigenic capacity of quiescent colorectal cancer stem cells. ACS Appl Mater Interfaces. 2021;13(14):15959–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by ICMR grant-File no. 5/3/8/39/ITR-F/2019 and SERB NPDF grant-File no. PDF/2022/003287 which was received by Dr. Anuradha Gupta.

Author information

Authors and Affiliations

Authors

Contributions

Dr. JM, Dr. AG, and Dr. AKP have conceptualized and designed the study. Dr. AG has performed the experiments and has written the manuscript, MS performed hemolysis studies, Dr. VK performed flow cytometry experiments, KN performed the survival study, and HC performed the graphical abstract drawing and proof reading. All the authors have approved the manuscript for submission and publication.

Corresponding author

Correspondence to Jairam Meena.

Ethics declarations

Ethics approval

This study was performed as per the institutional animal ethical guideline after approval from the Animal Ethical Committee of National Institute of Immunology (IAEC#500/19), New Delhi, India.

Conflict of interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 17 KB)

Supplementary methods

Supplementary methods

Synthesis of pH-responsive PLGA-coated core-shell magnetic silica nanoparticle

In brief, iron oxide nanoparticles (Fe3O4) were prepared by co-precipitation of iron (II) and iron (III) chloride salts under N2 atmosphere using 2M NaOH [26]. Initially, the iron salts were dissolved in the 50 ml of water in the molar ratio of 2:1, then NaOH was added slowly and the reaction temperature was raised to 70 °C. After 3h, the reaction was cooled down and the Fe3O4 was collected with the help of magnet and vacuum dried.

In the second step, core-shell magnetic silica nanoparticles were prepared by modified sol-gel homogenous co-precipitation process [27]. Fe3O4 NPs were dispersed in a mixture of ethanol and water (80:20). Tetra ethyl ortho silicate (TEOS), 0.1 ml (0.1% w/v) was added to the magnetic nanoparticles and stirred for 10 min at room temperature. Then, 2M NaOH (1 ml) was added slowly and reaction mixture was stirred for 6 h at room temperature. The core-shell magnetic silica nanoparticles (Fe3O4-SiO2 NPs) were separated using an external magnet, washed with deoxygenated distilled water and vacuum dried. To achieve amine functionalization, Fe3O4-SiO2 NPs were dispersed in pure ethanol and 2 mmol, 3-aminopropyl triethoxysilane (APTES) was added to it, refluxed at 70 °C, washed with ethanol two-three times and vacuum dried.

In the third step, PLGA-coated Fe3O4-SiO2 nanoparticles were prepared by double emulsion solvent evaporation method [28]. Poly lactic co-glycolic acid (PLGA) was dissolved in dichloromethane (DCM). Fe3O4-SiO2 NPs were dispersed in 0.5 ml of 1% PVA solution and added to PLGA solution with probe sonication (30% amplitude, 20 duty cycles, 2 min) to form primary emulsion (W1/O). The ratio of Fe3O4-SiO2 nanoparticles to PLGA was optimized as 1:1. Thus formed primary emulsion was then added to 30 ml of 2% PVA solution with sonication (30% amplitude, 20 duty cycles, 3 min) to form double emulsion (W1/O/W2) and continuously stirred for 5 h to evaporate DCM. After 5 h, PLGA-coated nanoparticles designated as Fe3O4-SiO2-PLGA were collected using external magnet, washed and dried.

In the fourth step, in order to fabricate pH-sensitive release system, PLGA-coated Fe3O4-SiO2 nanoparticles (100 mg) were dispersed in 10 ml of dopamine hydrochloride solution in Tris-HCl buffer (10 mmol/L, pH 8.5) and continuous stirred for 6 h at room temperature [29]. After 6 h, poly dopamine (PDA) capped nanoparticles designated as Fe3O4-SiO2-PLGA-PDA NPs/bare NPs were separated with magnet, washed with water, lyophilized and stored for further use.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gupta, A., Niveria, K., Chandpa, H.H. et al. Stimuli-responsive magnetic silica-poly-lactic-co-glycolic acid hybrid nanoparticles for targeted cancer chemo-immunotherapy. Drug Deliv. and Transl. Res. (2024). https://doi.org/10.1007/s13346-024-01521-0

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s13346-024-01521-0

Keywords

Navigation