We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Gold nanoparticles to improve HIV drug delivery

    Carolina Garrido

    Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7005, USA

    ,
    Carrie A Simpson

    Department of Chemistry, University of Colorado, Boulder, CO 80309, USA

    ,
    Noelle P Dahl

    Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7005, USA

    ,
    Jamee Bresee

    Department of Chemistry, University of Colorado, Boulder, CO 80309, USA

    ,
    Daniel C Whitehead

    Department of Chemistry, North Carolina State University, Raleigh, NC 27607, USA

    ,
    Erick A Lindsey

    Department of Chemistry, North Carolina State University, Raleigh, NC 27607, USA

    ,
    Tyler L Harris

    Department of Chemistry, North Carolina State University, Raleigh, NC 27607, USA

    ,
    Candice A Smith

    Department of Chemistry, University of Colorado, Boulder, CO 80309, USA

    ,
    Carly J Carter

    Department of Chemistry, University of Colorado, Boulder, CO 80309, USA

    ,
    Daniel L Feldheim

    Department of Chemistry, University of Colorado, Boulder, CO 80309, USA

    ,
    Christian Melander

    Department of Chemistry, North Carolina State University, Raleigh, NC 27607, USA

    &
    David M Margolis

    *Author for correspondence:

    E-mail Address: dmargo@med.unc.edu

    Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7005, USA

    Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA

    Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, USA

    Published Online:https://doi.org/10.4155/fmc.15.57

    Background: Antiretroviral therapy (ART) has improved lifespan and quality of life of patients infected with the HIV-1. However, ART has several potential limitations, including the development of drug resistance and suboptimal penetration to selected anatomic compartments. Improving the delivery of antiretroviral molecules could overcome several of the limitations of current ART. Results & Conclusion: Two to ten nanometer diameter inorganic gold crystals serve as a base scaffold to combine molecules with an array of properties in its surface. We show entry into different cell types, antiviral activity of an HIV integrase inhibitor conjugated in a gold nanoparticle and penetration into the brain in vivo without toxicity. Herein, gold nanoparticles prove to be a promising tool to use in HIV therapy.

    Papers of special note have been highlighted as: • of interest

    References

    • 1 Lucas GM, Chaisson RE, Moore RD. Highly active antiretroviral therapy in a large urban clinic: risk factors for virologic failure and adverse drug reactions. Ann. Intern. Med. 131(2), 81–87 (1999).
    • 2 Yazdanpanah Y, Fagard C, Descamps D et al. High rate of virologic suppression with raltegravir plus etravirine and darunavir/ritonavir among treatment-experienced patients infected with multidrug-resistant HIV: results of the ANRS 139 TRIO trial. Clin. Inf. Dis. 49(9), 1441–1449 (2009).
    • 3 Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clin. Pharmacol. Ther. 83(5), 761–769 (2007).• Comprehensive review of the state of the art in nanoparticle use for therapeutics.
    • 4 Mahajan SD, Aalinkeel R, Law WC et al. Anti-HIV-1 nanotherapeutics: promises and challenges for the future. Int. J. Nanomedicine 7, 5301–5314 (2012).
    • 5 Nowacek A, Mcmillan J, Miller R, Anderson A, Rabinow B, Gendelman H. Nanoformulated antiretroviral drug combinations extend drug release and antiretroviral responses in HIV-1-infected macrophages: implications for neuroaids therapeutics. J. Neuroimmune Pharmacol. 5(4), 592–601 (2010).• Interesting approach of antiretroviral nanoformulations using a cell-based delivery system to improve distribution.
    • 6 Shibata A, Pham A, Belshan M et al. Polymeric nanoparticles containing combination antiretroviral drugs for HIV type 1 treatment. AIDS Res. Hum. Retroviruses 29(5), 746–754 (2013).
    • 7 Rosi NL, Giljohann DA, Thaxton CS, Lytton-Jean AKR, Han MS, Mirkin CA. Oligonucleotide-modified gold nanoparticles for intracellular gene regulation. Science 312(5776), 1027–1030 (2006).
    • 8 Thaxton CS, Georganopoulou DG, Mirkin CA. Gold nanoparticle probes for the detection of nucleic acid targets. Clin. Chim. Acta 363(1–2), 120–126 (2006).
    • 9 Loo C, Hirsch L, Lee M-H et al. Gold nanoshell bioconjugates for molecular imaging in living cells. Opt. Lett. 30(9), 1012–1014 (2005).
    • 10 Gibson JD, Khanal BP, Zubarev ER. Paclitaxel-functionalized gold nanoparticles. J. Am. Chem. Soc. 129(37), 11653–11661 (2007).
    • 11 Libutti SK, Paciotti GF, Byrnes AA et al. Phase I and pharmacokinetic studies of CYT-6091, a novel pegylated colloidal gold-rh TNF nanomedicine. Clin. Cancer Res. 16(24), 6139–6149 (2010).
    • 12 Bresee J, Maier KE, Melander C, Feldheim DL. Identification of antibiotics using small molecule variable ligand display on gold nanoparticles. Chem. Comm. 46, 7516–7518 (2010).
    • 13 Bowman M-C, Ballard TE, Ackerson CJ, Feldheim DL, Margolis DM, Melander C. Inhibition of HIV fusion with multivalent gold nanoparticles. J. Am. Chem. Soc. 130(22), 6896–6897 (2008).• Reports the use of the same gold nanoparticles of the present study conjugated with a different antiretroviral drug.
    • 14 Simpson CA, Huffman BJ, Gerdon AE, Cliffel DE. Unexpected toxicity of monolayer protected gold clusters eliminated by PEG-thiol place exchange reactions. Chem. Res. Toxicol. 23(10), 1608–1616 (2010).
    • 15 Simpson CA, Salleng KJ, Cliffel DE, Feldheim DL. In vivo toxicity, biodistribution, and clearance of glutathione-coated gold nanoparticles. Nanomedicine 9(2), 257–263 (2013).
    • 16 Chithrani BD, Ghazani AA, Chan WC. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett. 6(4), 662–668 (2006).• Studies the importance of gold nanoparticles’ size in their ability to enter cells.
    • 17 Kim JA, Aberg C, Salvati A, Dawson KA. Role of cell cycle on the cellular uptake and dilution of nanoparticles in a cell population. Nat. Nano 7(1), 62–68 (2012).
    • 18 Tkachenko AG, Xie H, Liu Y et al. Cellular trajectories of peptide-modified gold particle complexes: comparison of nuclear localization signals and peptide transduction domains. Bioconjug. Chem. 15(3), 482–490 (2004).
    • 19 Baumann D, Hofmann D, Nullmeier S et al. Complex encounters: nanoparticles in whole blood and their uptake into different types of white blood cells. Nanomedicine 8(5), 699–713 (2013).
    • 20 Solmaz SR, Chauhan R, Blobel G, Melčák I. Molecular architecture of the transport channel of the nuclear pore complex. Cell 147(3), 590–602 (2011).
    • 21 Feldherr CM, Lanford RE, Akin D. Signal-mediated nuclear transport in simian virus 40-transformed cells is regulated by large tumor antigen. Proc. Natl Acad. Sci. USA 89(22), 11002–11005 (1992).
    • 22 De La Fuente JM, Berry CC. Tat peptide as an efficient molecule to translocate gold nanoparticles into the cell nucleus. Bioconjug. Chem. 16(5), 1176–1180 (2005).
    • 23 Syed S, Zubair A, Frieri M. Immune response to nanomaterials: Implications for medicine and literature review. Curr. Allergy Asthma Rep. 13(1), 50–57 (2013).
    • 24 Bresee J, Bond CM, Worthington RJ et al. Nanoscale structure-activity relationships, mode of action, and biocompatibility of gold nanoparticle antibiotics. J. Am. Chem. Soc. 136(14), 5295–5300 (2014).
    • 25 Ryan JA, Overton KW, Speight ME et al. Cellular uptake of gold nanoparticles passivated with BSA-SV40 large T antigen conjugates. Anal. Chem. 79(23), 9150–9159 (2007).
    • 26 Tkachenko AG, Xie H, Liu Y et al. Cellular trajectories of peptide-modified gold particle complexes: comparison of nuclear localization signals and peptide transduction domains. Bioconjug. Chem. 15(3), 482–490 (2004).
    • 27 Bender AR, Von Briesen H, Kreuter J, Duncan IB, Rübsamen-Waigmann H. Efficiency of nanoparticles as a carrier system for antiviral agents in human immunodeficiency virus-infected human monocytes/macrophages in vitro. Antimicrob. Agents Chemother. 40(6), 1467–1471 (1996).
    • 28 Shah L, Amiji M. Intracellular delivery of saquinavir in biodegradable polymeric nanoparticles for HIV/aids. Pharm. Res. 23(11), 2638–2645 (2006).
    • 29 Chattopadhyay N, Zastre J, Wong H-L, Wu X, Bendayan R. Solid lipid nanoparticles enhance the delivery of the HIV protease inhibitor, atazanavir, by a human brain endothelial cell line. Pharm. Res. 25(10), 2262–2271 (2008).
    • 30 Shiang Y-C, Ou C-M, Chen S-J et al. Highly efficient inhibition of human immunodeficiency virus type 1 reverse transcriptase by aptamers functionalized gold nanoparticles. Nanoscale 5(7), 2756–2764 (2013).
    • 31 Di Gianvincenzo P, Marradi M, Martínez-Ávila OM, Bedoya LM, Alcamí J, Penadés S. Gold nanoparticles capped with sulfate-ended ligands as anti-HIV agents. Bioorg. Med. Chem. Lett. 20(9), 2718–2721 (2010).
    • 32 Clifford DB, Ances BM. HIV-associated neurocognitive disorder. Lancet Infect. Dis. 13(11), 976–986 (2013).
    • 33 Gray LR, Roche M, Flynn JK, Wesselingh SL, Gorry PR, Churchill MJ. Is the central nervous system a reservoir of HIV-1? Curr. Opin. HIV AIDS 9(6), 552–558 (2014).
    • 34 De Jong WH, Hagens WI, Krystek P, Burger MC, Sips AJ, Geertsma RE. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials 29(12), 1912–1919 (2008).
    • 35 Sonavane G, Tomoda K, Makino K. Biodistribution of colloidal gold nanoparticles after intravenous administration: effect of particle size. Colloids Surf. B Biointerfaces 66(2), 274–280 (2008).
    • 36 Yim YS, Choi JS, Kim GT et al. A facile approach for the delivery of inorganic nanoparticles into the brain by passing through the blood-brain barrier (bbb). Chem. Commun. (Camb). 48(1), 61–63 (2012).
    • 37 Weiss CK, Kohnle MV, Landfester K et al. The first step into the brain: uptake of NIO-PBCA nanoparticles by endothelial cells in vitro and in vivo, and direct evidence for their blood-brain barrier permeation. ChemMedChem. 3(9), 1395–1403 (2008).
    • 38 Simpson CA, Agrawal AC, Balinski A, Harkness KM, Cliffel DE. Short-chain PEG mixed monolayer protected gold clusters increase clearance and red blood cell counts. ACS nano 5(5), 3577–3584 (2011).
    • 39 De Boer AG, Gaillard PJ. Drug targeting to the brain. Annu. Rev. Pharmacol. Toxicol. 47, 323–355 (2007).
    • 40 Rapoport SI, Ohno K, Fredericks WR, Pettigrew KD. Regional cerebrovascular permeability to [14c]sucrose after osmotic opening of the blood-brain barrier. Brain Res. 150(3), 653–657 (1978).
    • 41 Etame AB, Diaz RJ, O'reilly MA et al. Enhanced delivery of gold nanoparticles with therapeutic potential into the brain using MRI-guided focused ultrasound. Nanomedicine 8(7), 1133–1142 (2012).