Abstract

Antiretroviral therapy is currently only capable of controlling HIV replication rather than completely eradicating virus from patients. This is due in part to the establishment of a latent virus reservoir in resting CD4+ T cells, which persists even in the presence of HAART. It is thought that forced activation of latently infected cells could induce virus production, allowing targeting of the cell by the immune response. A variety of molecules are able to stimulate HIV from latency. However no tested purging strategy has proven capable of eliminating the infection completely or preventing viral rebound if therapy is stopped. Hence novel latency activation approaches are required. Nanoparticles can offer several advantages over more traditional drug delivery methods, including improved drug solubility, stability, and the ability to simultaneously target multiple different molecules to particular cell or tissue types. Here we describe the development of a novel lipid nanoparticle with the protein kinase C activator bryostatin-2 incorporated (LNP-Bry). These particles can target and activate primary human CD4+ T-cells and stimulate latent virus production from human T-cell lines in vitro and from latently infected cells in a humanized mouse model ex vivo. This activation was synergistically enhanced by the HDAC inhibitor sodium butyrate. Furthermore, LNP-Bry can also be loaded with the protease inhibitor nelfinavir (LNP-Bry-Nel), producing a particle capable of both activating latent virus and inhibiting viral spread. Taken together these data demonstrate the ability of nanotechnological approaches to provide improved methods for activating latent HIV and provide key proof-of-principle experiments showing how novel delivery systems may enhance future HIV therapy.

Highlights

  • Active antiretroviral therapy (HAART) can powerfully suppress HIV replication but does not clear virus from infected individuals

  • Bryostatin has been demonstrated to activate latent HIV at nanomolar concentrations in vitro [9,10,11]. This molecule is believed to induce expression of HIV by activating NFkB [12], which is required for optimal transcription of viral mRNA from the HIV long terminal repeat promoter (LTR)

  • Bryostatin-1 has been tested in clinical trials for its anti-cancer properties [13,14,15,16,17,18] there are no published studies using this class of compounds in clinical HIV purging strategies

Read more

Summary

Introduction

Active antiretroviral therapy (HAART) can powerfully suppress HIV replication but does not clear virus from infected individuals. Complete eradication of HIV from infected individuals will probably require further improvements in latency activators and the development of a more optimized virus purging strategy. One focus of latency research is the identification of factors that can activate HIV from latency and have the potential to be used in a clinical setting. Such factors have included histone deacetylase inhibitors, agonistic anti-CD3 antibodies, and cytokines such as interleukin (IL)-2 and IL-7 [2,4,5,6,7,8]. Bryostatin has been demonstrated to activate latent HIV at nanomolar concentrations in vitro [9,10,11]. Bryostatin-2 was used in our studies and has slightly different properties than the parent compound bryostatin-1, both can activate PKC at nanomolar concentrations [13]

Objectives
Methods
Results
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call