1. Academic Validation
  2. Lipophilic siRNA targets albumin in situ and promotes bioavailability, tumor penetration, and carrier-free gene silencing

Lipophilic siRNA targets albumin in situ and promotes bioavailability, tumor penetration, and carrier-free gene silencing

  • Proc Natl Acad Sci U S A. 2017 Aug 8;114(32):E6490-E6497. doi: 10.1073/pnas.1621240114.
Samantha M Sarett 1 Thomas A Werfel 1 Linus Lee 1 Meredith A Jackson 1 Kameron V Kilchrist 1 Dana Brantley-Sieders 2 Craig L Duvall 3
Affiliations

Affiliations

  • 1 Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235.
  • 2 Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232.
  • 3 Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235; [email protected].
Abstract

Clinical translation of therapies based on small interfering RNA (siRNA) is hampered by siRNA's comprehensively poor pharmacokinetic properties, which necessitate molecule modifications and complex delivery strategies. We sought an alternative approach to commonly used nanoparticle carriers by leveraging the long-lived endogenous serum protein albumin as an siRNA carrier. We synthesized siRNA conjugated to a diacyl lipid moiety (siRNA-L2), which rapidly binds albumin in situ. siRNA-L2, in comparison with unmodified siRNA, exhibited a 5.7-fold increase in circulation half-life, an 8.6-fold increase in bioavailability, and reduced renal accumulation. Benchmarked against leading commercial siRNA nanocarrier in vivo jetPEI, siRNA-L2 achieved 19-fold greater tumor accumulation and 46-fold increase in per-tumor-cell uptake in a mouse orthotopic model of human triple-negative breast Cancer. siRNA-L2 penetrated tumor tissue rapidly and homogeneously; 30 min after i.v. injection, siRNA-L2 achieved uptake in 99% of tumor cells, compared with 60% for jetPEI. Remarkably, siRNA-L2 achieved a tumor:liver accumulation ratio >40:1 vs. <3:1 for jetPEI. The improved pharmacokinetic properties of siRNA-L2 facilitated significant tumor gene silencing for 7 d after two i.v. doses. Proof-of-concept was extended to a patient-derived xenograft model, in which jetPEI tumor accumulation was reduced fourfold relative to the same formulation in the orthotopic model. The siRNA-L2 tumor accumulation diminished only twofold, suggesting that the superior tumor distribution of the conjugate over nanoparticles will be accentuated in clinical situations. These data reveal the immense promise of in situ albumin targeting for development of translational, carrier-free RNAi-based Cancer therapies.

Keywords

RNAi; albumin binding; pharmacokinetics; siRNA delivery; tumor delivery.

Figures
Products