Skip to main content

Advertisement

Log in

Dual-Targeted Delivery of Nanoparticles Encapsulating Paclitaxel and Everolimus: a Novel Strategy to Overcome Breast Cancer Receptor Heterogeneity

  • Research Paper
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

The intratumoral heterogeneity observed in breast cancer (BC), in particular with regard to cell surface receptor expression, can hinder the success of many targeted cancer therapies. The development of novel therapeutic agents that target more than one receptor can overcome this inherent property of tumors and can facilitate their selective internalization in cancer cells. The goal of this study is to develop a drug combination-loaded nanoparticle (NP) formulation that is actively-targeted to HER2 and EGFR receptors on BC cells.

Methods

A polymeric NP formulation was prepared which co-encapsulated a synergistic combination of the chemotherapeutic agent, paclitaxel (PTX), and the mTOR inhibitor, everolimus (EVER), and is targeted to HER2 and EGFR receptors on BC cells using antibody Fab fragments as the targeting moieties. The physicochemical characteristics of the dual-targeted formulation (Dual-NP) were evaluated, along with its cytotoxic profile (in both, monolayer and 3D BC models), as well as the degree of cellular uptake in HER2high/EGFRmod and HER2neg/EGFRlow BC cells.

Results

Dual-NPs were found to have significantly higher cytotoxicity relative to HER2 mono-targeted (T-NPs) and untargeted NPs (UT-NPs) in HER2high/EGFRmod monolayer BC cells after 72 h exposure, while no significant difference was observed in HER2neg/EGFRlow cells. However, in the HER2high/EGFRmod spheroids, the cytotoxicity of Dual-NPs was comparable to that of T-NPs. This was thought to be attributed to the previously reported downregulation of EGFR in 3D in comparison to 2D BC models. Dual-NPs had significantly higher cellular uptake relative to UT-NPs and T-NPs in HER2high/EGFRmod BC cells after 24 h exposure, whereas in the HER2neg/EGFRlow cells, the increase in cellular uptake of the Dual-NPs was not as high as the level achieved in the HER2high/EGFRmod cells. Blocking HER2 and EGFR significantly reduced the uptake of T-NPs and Dual-NPs in the HER2high/EGFRmod BC cells, demonstrating specific binding to both EGFR and HER2.

Conclusions

The dual-targeting strategy developed in this study in conjunction with a potentially promising delivery vector for a synergistic combination therapy can overcome receptor heterogeneity, yielding significant improvements in the cytotoxicity and cellular uptake in BC cells.

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

Similar content being viewed by others

References

  1. Ayoub JPM, Verma S. Advances in the management of metastatic breast cancer: options beyond first-line chemotherapy. Curr Oncol. 2012;19(2):91–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gallardo A, Lerma E, Escuin D, Tibau A, Muñoz J, Ojeda B, et al. Increased signalling of EGFR and IGF1R, and deregulation of PTEN/PI3K/Akt pathway are related with trastuzumab resistance in HER2 breast carcinomas. Br J Cancer. 2012;106(8):1367–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. DiGiovanna MP, Stern DF, Edgerton SM, Whalen SG, Moore D, Thor AD. Relationship of epidermal growth factor receptor expression to ErbB-2 signaling activity and prognosis in breast cancer patients. J Clin Oncol. 2005;23(6):1152–60.

    Article  CAS  PubMed  Google Scholar 

  4. Kontermann RE. Dual targeting strategies with bispecific antibodies. MAbs. 2012;4(2):182–97.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Liu Y, Sun J, Lian H, Cao W, Wang Y, He Z. Folate and CD44 receptors dual-targeting hydrophobized hyaluronic acid paclitaxel-loaded polymeric micelles for overcoming multidrug resistance and improving tumor distribution. J Pharm Sci. 2014;103(5):1538–47.

    Article  CAS  PubMed  Google Scholar 

  6. Ajabnoor GMA, Crook T, Coley HM. Paclitaxel resistance is associated with switch from apoptotic to autophagic cell death in MCF-7 breast cancer cells. Cell Death Dis. 2012;3(1):e260. https://doi.org/10.1038/cddis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Murray S, Briasoulis E, Linardou H, Bafaloukos D, Papadimitriou C. Taxane resistance in breast cancer: mechanisms, predictive biomarkers and circumvention strategies. Cancer Treat Rev. 2012;38(7):890–903.

    Article  CAS  PubMed  Google Scholar 

  8. Gelderblom H, Verweij J, Nooter K, Sparreboom A, Cremophor EL. The drawbacks and advantages of vehicle selection for drug formulation. Eur J Cancer. 2001;37:1590–8.

    Article  CAS  PubMed  Google Scholar 

  9. Houdaihed L, Evans JC, Allen C. Codelivery of paclitaxel and Everolimus at the optimal synergistic ratio: a promising solution for the treatment of breast cancer. Mol Pharm. 2018;15(9):3672–81.

    Article  CAS  PubMed  Google Scholar 

  10. Chapman AP. PEGylated antibodies and antibody fragments for improved therapy: a review. Adv Drug Deliv Rev. 2002;54(4):531–45.

    Article  CAS  PubMed  Google Scholar 

  11. Hoang B, Ekdawi SN, Reilly RM, Allen C. Active targeting of block copolymer micelles with trastuzumab fab fragments and nuclear localization signal leads to increased tumor uptake and nuclear localization in HER2-overexpressing xenografts. Mol Pharm. 2013;10(11):4229–41.

    Article  CAS  PubMed  Google Scholar 

  12. Ahn J, Miura Y, Yamada N, Chida T, Liu X, Kim A, et al. Antibody fragment-conjugated polymeric micelles incorporating platinum drugs for targeted therapy of pancreatic cancer. Biomaterials. 2015;39:23–30.

    Article  CAS  PubMed  Google Scholar 

  13. Fiandra L, Mazzucchelli S, De Palma C, Colombo M, Allevi R, Sommaruga S, et al. Assessing the in vivo targeting efficiency of multifunctional nanoconstructs bearing antibody-derived ligands. ACS Nano. 2013;7(7):6092–102.

    Article  CAS  PubMed  Google Scholar 

  14. Quarta A, Bernareggi D, Benigni F, Luison E, Nano G, Nitti S, et al. Targeting FR-expressing cells in ovarian cancer with fab-functionalized nanoparticles: a full study to provide the proof of principle from in vitro to in vivo. Nanoscale. 2015;7(6):2336–51.

    Article  CAS  PubMed  Google Scholar 

  15. Munster P, Krop IE, LoRusso P, Ma C, Siegel BA, Shields AF, et al. Safety and pharmacokinetics of MM-302, a HER2-targeted antibody–liposomal doxorubicin conjugate, in patients with advanced HER2-positive breast cancer: a phase 1 dose-escalation study. Br J Cancer. 2018;119(9):1086–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Miller K, Cortes J, Hurvitz SA, Krop IE, Tripathy D, Verma S, et al. HERMIONE: a randomized phase 2 trial of MM-302 plus trastuzumab versus chemotherapy of physician’s choice plus trastuzumab in patients with previously treated, anthracycline-naïve, HER2-positive, locally advanced/metastatic breast cancer. BMC Cancer. 2016;16(1):1–11.

    Article  CAS  Google Scholar 

  17. Duan D, Wang A, Ni L, Zhang L, Yan X, Jiang Y, et al. Trastuzumab- and fab’ fragment-modified curcumin PEG -PLGA nanoparticles: preparation and evaluation in vitro and in vivo. Int J Nanomedicine. 2018;13:1831–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, et al. The expression patterns of ER, PR, HER2, CK5/6, EGFR, KI-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer Basic Clin Res. 2010;4(1):35–41.

    Google Scholar 

  19. Chung I, Reichelt M, Shao L, Akita RW, Koeppen H, Rangell L, et al. High cell-surface density of HER2 deforms cell membranes. Nat Commun. 2016;7:1–11.

    CAS  Google Scholar 

  20. Yunokawa M, Koizumi F, Kitamura Y, Katanasaka Y, Okamoto N, Kodaira M, et al. Efficacy of everolimus, a novel mTOR inhibitor, against basal-like triple-negative breast cancer cells. Cancer Sci. 2012;103(9):1665–71.

    Article  CAS  PubMed  Google Scholar 

  21. Hoang B, Reilly RM, Allen C. Block copolymer micelles target auger Electron radiotherapy to the nucleus of HER2-positive breast cancer cells. Biomacromolecules. 2012;13(2):455–65.

    Article  CAS  PubMed  Google Scholar 

  22. Scollard DA, Chan C, Holloway CMB, Reilly RM. A kit to prepare 111In-DTPA-trastuzumab (Herceptin) Fab fragments injection under GMP conditions for imaging or radioimmunoguided surgery of HER2-positive breast cancer. Nucl Med Biol. 2011;38(1):129–36.

    Article  CAS  PubMed  Google Scholar 

  23. Gao J, Kou G, Wang H, Chen H, Li B, Dai J, et al. PE38KDEL-loaded anti-HER2 nanoparticles inhibit breast tumor progression with reduced toxicity and immunogenicity. Breast Cancer Res Treat. 2009;115:29–41.

    Article  CAS  PubMed  Google Scholar 

  24. Eetezadi S. Nanomedicines and combination therapy of doxorubicin and Olaparib for treatment of ovarian Cancer. 2016.

    Google Scholar 

  25. Zhao DZ, Zhao X, Zu Y, Li J, Zhang Y, Jiang R, et al. Preparation, characterization, and in vitro targeted delivery of folate-decorated paclitaxel-loaded bovine serum albumin nanoparticles. Int J Nanomedicine. 2010:669–77.

  26. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12(4):207–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Pickl M, Ries CH. Comparison of 3D and 2D tumor models reveals enhanced HER2 activation in 3D associated with an increased response to trastuzumab. Oncogene. 2009;28:461–46.

    Article  CAS  PubMed  Google Scholar 

  28. Wang K, Kievit FM, Florczyk SJ, Stephen ZR, Zhang M. 3D porous chitosan–alginate scaffolds as an in vitro model for evaluating nanoparticle–mediated tumor targeting and gene delivery to prostate cancer. Biomacromolecules. 2015;16(10):3362–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cheng Z, Al Zaki A, Hui JZ, Muzykantov VR, Tsourkas A. Multifunctional nanoparticles: cost versus benefit of adding targeting and imaging capabilities. Science. 2012;338(6109):903–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Goodman T, Olive P, Pun S. Increased nanoparticle penetration in collagenase- treated multicellular spheroids. J Struct Chem. 2007;2(2):265–74.

    CAS  Google Scholar 

  31. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:2–25.

    Article  CAS  PubMed  Google Scholar 

  32. Cabral H, Matsumoto Y, Mizuno K, Chen Q, Murakami M, Kimura M, et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat Nanotechnol. 2011;6(12):815–23.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments And Disclosures

L.H. is the recipient of the CIHR Doctoral Research Award and the Centre for Pharmaceutical Oncology Scholarship. C.A. acknowledges research support from CIHR (Grant MOP325013). The authors have no conflict of interest to declare.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Christine Allen.

Additional information

Guest Editor: Joshua Reineke

Publisher’s Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Houdaihed, L., Evans, J.C. & Allen, C. Dual-Targeted Delivery of Nanoparticles Encapsulating Paclitaxel and Everolimus: a Novel Strategy to Overcome Breast Cancer Receptor Heterogeneity. Pharm Res 37, 39 (2020). https://doi.org/10.1007/s11095-019-2684-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11095-019-2684-6

Key words

Navigation