Pharmaceutics (Oct 2020)

Viral load Reduction in SHIV-Positive Nonhuman Primates via Long-Acting Subcutaneous Tenofovir Alafenamide Fumarate Release from a Nanofluidic Implant

  • Fernanda P. Pons-Faudoa,
  • Nicola Di Trani,
  • Antons Sizovs,
  • Kathryn A. Shelton,
  • Zoha Momin,
  • Lane R. Bushman,
  • Jiaqiong Xu,
  • Dorothy E. Lewis,
  • Sandra Demaria,
  • Trevor Hawkins,
  • James F. Rooney,
  • Mark A. Marzinke,
  • Jason T. Kimata,
  • Peter L. Anderson,
  • Pramod N. Nehete,
  • Roberto C. Arduino,
  • K. Jagannadha Sastry,
  • Alessandro Grattoni

DOI
https://doi.org/10.3390/pharmaceutics12100981
Journal volume & issue
Vol. 12, no. 10
p. 981

Abstract

Read online

HIV-1 is a chronic disease managed by strictly adhering to daily antiretroviral therapy (ART). However, not all people living with HIV-1 have access to ART, and those with access may not adhere to treatment regimens increasing viral load and disease progression. Here, a subcutaneous nanofluidic implant was used as a long-acting (LA) drug delivery platform to address these issues. The device was loaded with tenofovir alafenamide (TAF) and implanted in treatment-naïve simian HIV (SHIV)-positive nonhuman primates (NHP) for a month. We monitored intracellular tenofovir-diphosphate (TFV-DP) concentration in the target cells, peripheral blood mononuclear cells (PBMC). The concentrations of TFV-DP were maintained at a median of 391.0 fmol/106 cells (IQR, 243.0 to 509.0 fmol/106 cells) for the duration of the study. Further, we achieved drug penetration into lymphatic tissues, known for persistent HIV-1 replication. Moreover, we observed a first-phase viral load decay of −1.14 ± 0.81 log10 copies/mL (95% CI, −0.30 to −2.23 log10 copies/mL), similar to −1.08 log10 copies/mL decay observed in humans. Thus, LA TAF delivered from our nanofluidic implant had similar effects as oral TAF dosing with a lower dose, with potential as a platform for LA ART.

Keywords