86
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Nanoparticle-Mediated Co-Delivery of Notch-1 Antibodies and ABT-737 as a Potent Treatment Strategy for Triple-Negative Breast Cancer

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Triple-negative breast cancer (TNBC) accounts for nearly one-quarter of all breast cancer cases, but effective targeted therapies for this disease remain elusive because TNBC cells lack expression of the three most common receptors seen on other subtypes of breast cancer. Here, we exploit TNBC cells’ overexpression of Notch-1 receptors and Bcl-2 anti-apoptotic proteins to provide an effective targeted therapy. Prior studies have shown that the small molecule drug ABT-737, which inhibits Bcl-2 to reinstate apoptotic signaling, is a promising candidate for TNBC therapy. However, ABT-737 is poorly soluble in aqueous conditions, and its orally bioavailable derivative causes severe thrombocytopenia. To enable targeted delivery of ABT-737 to TNBC and enhance its therapeutic efficacy, we encapsulated the drug in poly (lactic-co-glycolic acid) nanoparticles (NPs) that were functionalized with Notch-1 antibodies to produce N1-ABT-NPs. The antibodies in this NP platform enable both TNBC cell-specific binding and suppression of Notch signaling within TNBC cells by locking the Notch-1 receptors in a ligand unresponsive state. This Notch inhibition potentiates the effect of ABT-737 by up-regulating Noxa, resulting in effective killing of TNBC cells. We present the results of in vitro studies that demonstrate N1-ABT-NPs can preferentially bind TNBC cells versus noncancerous breast epithelial cells to effectively regulate Bcl-2 and Notch signaling to induce cell death. Further, we show that N1-ABT-NPs can accumulate in subcutaneous TNBC xenograft tumors in mice following systemic administration to reduce tumor burden and extend animal survival. Together, these findings demonstrate that NP-mediated co-delivery of Notch-1 antibodies and ABT-737 is a potent treatment strategy for TNBC that may improve patient outcomes with further development and implementation.

          Graphical Abstract

          Related collections

          Author and article information

          Journal
          101313589
          34787
          ACS Nano
          ACS Nano
          ACS nano
          1936-0851
          1936-086X
          12 March 2020
          26 February 2020
          24 March 2020
          24 March 2021
          : 14
          : 3
          : 3378-3388
          Affiliations
          Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
          Department of Biomedical Engineering and Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States; Helen F. Graham Cancer Center and Research Institute, Newark, Delaware 19713, United States
          Author notes
          Corresponding Author: Emily S. Day – Department of Biomedical Engineering and Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States; Helen F. Graham Cancer Center and Research Institute, Newark, Delaware 19713, United States; emilyday@ 123456udel.edu
          Author information
          http://orcid.org/0000-0002-8707-826X
          Article
          PMC7098846 PMC7098846 7098846 nihpa1574628
          10.1021/acsnano.9b09263
          7098846
          32083466
          cc866cd3-2dab-4ce0-83c7-96e3ed7cb354
          History
          Categories
          Article

          Notch signaling,gene regulation,nanocarrier,targeted cancer nanomedicine,signal cascade interference,multivalency,drug delivery

          Comments

          Comment on this article