Limitations in disease outcomes for HIV/AIDS are influenced by poor adherence to antiretroviral therapy (ART) and inadequate drug penetrance into viral reservoirs (gut, lymph node and central nervous system). The need for lifelong daily regimens, pill fatigue, co-morbid diseases, substance abuse and inherent drug toxicities often affect adherence to drug regimens. When suboptimal adherence occurs, it results in accelerated rates of human immunodeficiency virus type one (HIV-1) mutation and consequent disease progression. We posit that the emergence of long-acting nanoformulated ART (nanoART) can positively impact these limitations. In order to move our work beyond the laboratory bench to clinical use, we propose a series of next step investigations to firmly establish the cell-based carriage system for nanoART that began four years ago. Such a system for drug delivery has been demonstrated to be a highly efficient means to generate ART depots that limit tissue and cell toxicities positively affecting immune and viral parameters. These were now demonstrated for Cabotegravir, a long-acting injectable integrase inhibitor made by GlaxoSmithKline Pharmaceuticals that is being tested in phase II investigations for a once every two-month dose. We posit that this and other antiretrovirals can be packaged into targeted nanoparticles to both improve drug biodistribution, dose frequency and concentration for up to six months. Indeed, preliminary data supports this contention and its promise seen in animal models of HIV/AIDS. These include rodents and rhesus macaques. We posit that such a drug delivery system can inevitably revolutionize ART treatment and potentially eliminate conventional therapeutic regimens. To such ends, we have built a highly integrative and cross-disciplinary research program. We have shown previously that a single dose of targeted nanoART can produce high-sustained tissue and plasma drug levels in the reticuloendothelial system (RES; lymphoid tissues, liver and brain). Notably, such formulations have shown long-lived drug depots in monocyte-macrophages within the RES. NanoART can be taken up within minutes by circulating monocytes and tissue macrophages and released in tissues over a period of months. Our partners in the University of Nebraska Medical Center College of Pharmacy (Project 1. T. Bronich and Core B, Y. Alnouti) will be joined with UNMC College of Medicine Departments of Radiology (Project 3, M. Boska) and Pharmacology and Experimental Neuroscience (Project 2, H. Gendelman; Core C, L. Poluektova and H. Fox), and The Scripps Research Institute (Project 3, G. Siuzdak) to optimize the formulations and better appreciate its toxicology profiles. This can now be facilitated through integrated cell biologic, pharmacologic, virologic, and molecular testing to translate the technologies to the patient bedside (Core A, H. Gendelman, R.L. Mosley and J. McMillan). The research takes advantage of the newly minted Nebraska Nanomedicine Production Plant with broad resources in medicinal and polymer chemistry, molecular bioimaging, and pharmaceutics.

Public Health Relevance

Antiretroviral therapy (ART) is commonly associated with untoward side effects, failures of drug reservoir (gut, lymphoid tissues and brain) penetrance, limited adherence to daily regimens and viral mutation. All or any of these can lead to treatment failures which are common in the drug abusing population. The goal of this program is to improve medicine access and improve clinical outcomes for HIV/AIDS.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Program Projects (P01)
Project #
5P01DA028555-08
Application #
9304046
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Rapaka, Rao
Project Start
2010-07-15
Project End
2020-06-30
Budget Start
2017-07-01
Budget End
2018-06-30
Support Year
8
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Nebraska Medical Center
Department
Pharmacology
Type
Schools of Medicine
DUNS #
168559177
City
Omaha
State
NE
Country
United States
Zip Code
68198
Beyer, Brittney A; Fang, Mingliang; Sadrian, Benjamin et al. (2018) Metabolomics-based discovery of a metabolite that enhances oligodendrocyte maturation. Nat Chem Biol 14:22-28
Herskovitz, Jonathan; Gendelman, Howard E (2018) HIV and the Macrophage: From Cell Reservoirs to Drug Delivery to Viral Eradication. J Neuroimmune Pharmacol :
Kevadiya, Bhavesh D; Woldstad, Christopher; Ottemann, Brendan M et al. (2018) Multimodal Theranostic Nanoformulations Permit Magnetic Resonance Bioimaging of Antiretroviral Drug Particle Tissue-Cell Biodistribution. Theranostics 8:256-276
McMillan, JoEllyn; Szlachetka, Adam; Slack, Lara et al. (2018) Pharmacokinetics of a Long-Acting Nanoformulated Dolutegravir Prodrug in Rhesus Macaques. Antimicrob Agents Chemother 62:
Ottemann, Brendan M; Helmink, Austin J; Zhang, Wenting et al. (2018) Bioimaging predictors of rilpivirine biodistribution and antiretroviral activities. Biomaterials 185:174-193
Zhou, Tian; Su, Hang; Dash, Prasanta et al. (2018) Creation of a nanoformulated cabotegravir prodrug with improved antiretroviral profiles. Biomaterials 151:53-65
Kevadiya, Bhavesh D; Ottemann, Brendan M; Thomas, Midhun Ben et al. (2018) Neurotheranostics as personalized medicines. Adv Drug Deliv Rev :
Gautam, Nagsen; Lin, Zhiyi; Banoub, Mary G et al. (2018) Simultaneous quantification of intracellular lamivudine and abacavir triphosphate metabolites by LC-MS/MS. J Pharm Biomed Anal 153:248-259
Zhou, Tian; Lin, Zhiyi; Puligujja, Pavan et al. (2018) Optimizing the preparation and stability of decorated antiretroviral drug nanocrystals. Nanomedicine (Lond) 13:871-885
Kiyota, Tomomi; Machhi, Jatin; Lu, Yaman et al. (2018) Granulocyte-macrophage colony-stimulating factor neuroprotective activities in Alzheimer's disease mice. J Neuroimmunol 319:80-92

Showing the most recent 10 out of 141 publications