403
Views
28
CrossRef citations to date
0
Altmetric
Review

Lipid-based carriers for controlled delivery of nitric oxide

, , &
Pages 1341-1353 | Received 29 Sep 2016, Accepted 19 Jan 2017, Published online: 06 Feb 2017

References

  • Choi DW, Koh JY, Peters S. Pharmacology of glutamate neurotoxicity in cortical cell culture: attenuation by NMDA antagonists. J Neurosci. 1988;8:185–196.
  • Ignarro LJ, Buga GM, Wood KS, et al. Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci U S A. 1987;84:9265–9269.
  • Ignarro LJ. Endothelium-derived nitric oxide: actions and properties. Faseb J. 1989;3:31–36.
  • Griffith OW, Stuehr DJ. Nitric oxide synthases: properties and catalytic mechanism. Annu Rev Physiol. 1995;57:707–736.
  • Elnaggar MA, Seo SH, Gobaa S, et al. Nitric oxide releasing coronary stent: a new approach using layer-by-layer coating and liposomal encapsulation. Small. 2016;12:6012–6023.
  • Arnal JF, Dinh-Xuan AT, Pueyo M, et al. Endothelium-derived nitric oxide and vascular physiology and pathology. Cell Mol Life Sci. 1999;55:1078–1087.
  • Loscalzo J. Nitric oxide insufficiency, platelet activation, and arterial thrombosis. Circ Res. 2001;88:756–762.
  • Rudic RD, Shesely EG, Maeda N, et al. Direct evidence for the importance of endothelium-derived nitric oxide in vascular remodeling. J Clin Invest. 1998;101:731–736.
  • Garry PS, Ezra M, Rowland MJ, et al. The role of the nitric oxide pathway in brain injury and its treatment – from bench to bedside. Exp Neurol. 2015;263:235–243.
  • Napoli C, Paolisso G, Casamassimi A, et al. Effects of nitric oxide on cell proliferation: novel insights. J Am Coll Cardiol. 2013;62:89–95.
  • Tanner FC, Meier P, Greutert H, et al. Nitric oxide modulates expression of cell cycle regulatory proteins: a cytostatic strategy for inhibition of human vascular smooth muscle cell proliferation. Circulation. 2000;101:1982–1989.
  • Kapadia MR, Eng JW, Jiang Q, et al. Nitric oxide regulates the 26S proteasome in vascular smooth muscle cells. Nitric Oxide. 2009;20:279–288.
  • Gooch KJ, Dangler CA, Frangos JA. Exogenous, basal, and flow-induced nitric oxide production and endothelial cell proliferation. J Cell Physiol. 1997;171:252–258.
  • Ishida A, Sasaguri T, Kosaka C, et al. Induction of the cyclin-dependent kinase inhibitor p21(Sdi1/Cip1/Waf1) by nitric oxide-generating vasodilator in vascular smooth muscle cells. J Biol Chem. 1997;272:10050–10057.
  • Taylor EL, Li JT, Tupper JC, et al. GEA 3162, a peroxynitrite donor, induces Bcl-2-sensitive, p53-independent apoptosis in murine bone marrow cells. Biochem Pharmacol. 2007;74:1039–1049.
  • Mocellin S, Bronte V, Nitti D. Nitric oxide, a double edged sword in cancer biology: searching for therapeutic opportunities. Med Res Rev. 2007;27:317–352.
  • Hakim TS, Sugimori K, Camporesi EM, et al. Half-life of nitric oxide in aqueous solutions with and without haemoglobin. Physiol Meas. 1996;17:267.
  • Keefer LK. Fifty years of diazeniumdiolate research. From laboratory curiosity to broad-spectrum biomedical advances. ACS Chem Biol. 2011;6:1147–1155.
  • Tan A, Farhatnia Y, De Mel A, et al. Inception to actualization: next generation coronary stent coatings incorporating nanotechnology. J Biotechnol. 2013;164:151–170.
  • Elnaggar MA, Joung YK, Han DK. Advanced Stents for Cardiovascular Applications. In: Jo H, Jun H-W, Shin J, et al., editors. Biomedical Engineering: Frontier Research and Converging Technologies. Cham: Springer International Publishing; 2016. p. 407–426.
  • Wan A, Gao Q, Li H. Preparation and characterization of diazeniumdiolate releasing ethylcellulose films. J Mater Sci: Mater Med. 2009;20:321–327.
  • Frost MC, Reynolds MM, Meyerhoff ME. Polymers incorporating nitric oxide releasing/generating substances for improved biocompatibility of blood-contacting medical devices. Biomaterials. 2005;26:1685–1693.
  • Yoo J-W, Lee J-S, Lee CH. Characterization of nitric oxide-releasing microparticles for the mucosal delivery. J Biomed Mater Res. 2010;92A:1233–1243.
  • Kushwaha M, Anderson JM, Bosworth CA, et al. A nitric oxide releasing, self-assembled peptide amphiphile matrix that mimics native endothelium for coating implantable cardiovascular devices. Biomaterials. 2010;31:1502–1508.
  • Tai LA, Wang YC, Yang CS. Heat-activated sustaining nitric oxide release from zwitterionic diazeniumdiolate loaded in thermo-sensitive liposomes. Nitric Oxide. 2010;23:60–64.
  • Ostrowski AD, Lin BF, Tirrell MV, et al. Liposome encapsulation of a photochemical no precursor for controlled nitric oxide release and simultaneous fluorescence imaging. Mol Pharm. 2012;9:2950–2955.
  • Antimisiaris SG, Siablis D, Liatsikos E, et al. Liposome-coated metal stents: an in vitro evaluation of controlled-release modality in the ureter. J Endourology /Endourological Soc. 2000;14:743–747.
  • Koromila G, Michanetzis GP, Missirlis YF, et al. Heparin incorporating liposomes as a delivery system of heparin from PET-covered metallic stents: effect on haemocompatibility. Biomaterials. 2006;27:2525–2533.
  • Kisel MA, Kulik LN, Tsybovsky IS, et al. Liposomes with phosphatidylethanol as a carrier for oral delivery of insulin: studies in the rat. Int J Pharm. 2001;216:105–114.
  • Kanaoka E, Takahashi K, Yoshikawa T, et al. A novel and simple type of liposome carrier for recombinant interleukin-2. J Pharm Pharmacol. 2001;53:295–302.
  • Li H, Song J-H, Park J-S HK. Polyethylene glycol-coated liposomes for oral delivery of recombinant human epidermal growth factor. Int J Pharm. 2003;258:11–19.
  • Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4:145–160.
  • Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4:307–320.
  • DiTizio V, Ferguson GW, Mittelman MW, et al. A liposomal hydrogel for the prevention of bacterial adhesion to catheters. Biomaterials. 1998;19:1877–1884.
  • Britton GL, Kim H, Kee PH, et al. In vivo therapeutic gas delivery for neuroprotection with echogenic liposomes. Circulation. 2010;122:1578–1587.
  • Peng T, Britton GL, Kim H, et al. Therapeutic time window and dose dependence of xenon delivered via echogenic liposomes for neuroprotection in stroke. CNS Neurosci Ther. 2013;19:773–784.
  • Unnikrishnan S, Klibanov AL. Microbubbles as ultrasound contrast agents for molecular imaging: preparation and application. AJR Am J Roentgenol. 2012;199:292–299.
  • Paini M, Daly SR, Aliakbarian B, et al. An efficient liposome based method for antioxidants encapsulation. Colloids Surf B Biointerfaces. 2015;136:1067–1072.
  • Huang SL, McPherson DD, Macdonald RC. A method to co-encapsulate gas and drugs in liposomes for ultrasound-controlled drug delivery. Ultrasound Med Biol. 2008;34:1272–1280.
  • Tardy I, Pochon S, Theraulaz M, et al. In vivo ultrasound imaging of thrombi using a target-specific contrast agent. Acad Radiol. 2002;9(Suppl 2):S294–6.
  • Chen X, Wang X, Wang Y, et al. Improved tumor-targeting drug delivery and therapeutic efficacy by cationic liposome modified with truncated bFGF peptide. J Control Release. 2010;145:17–25.
  • Chen CC, Borden MA. Ligand conjugation to bimodal poly(ethylene glycol) brush layers on microbubbles. Langmuir. 2010;26:13183–13194.
  • Kwan JJ, Kaya M, Borden MA, et al. Theranostic oxygen delivery using ultrasound and microbubbles. Theranostics. 2012;2:1174–1184.
  • Tong J, Ding J, Shen X, et al. Mesenchymal stem cell transplantation enhancement in myocardial infarction rat model under ultrasound combined with nitric oxide microbubbles. PLoS One. 2013;8:e80186.
  • Eisenbrey JR, Albala L, Kramer MR, et al. Development of an ultrasound sensitive oxygen carrier for oxygen delivery to hypoxic tissue. Int J Pharm. 2015;478:361–367.
  • Cavalli R, Bisazza A, Rolfo A, et al. Ultrasound-mediated oxygen delivery from chitosan nanobubbles. Int J Pharm. 2009;378:215–217.
  • Cavalli R, Bisazza A, Giustetto P, et al. Preparation and characterization of dextran nanobubbles for oxygen delivery. Int J Pharm. 2009;381:160–165.
  • Borden MA, Longo ML. Dissolution behavior of lipid monolayer-coated, air-filled microbubbles: effect of lipid hydrophobic chain length. Langmuir. 2002;18:9225–9233.
  • Van Liew HD, Burkard ME. Bubbles in circulating blood: stabilization and simulations of cyclic changes of size and content. J Appl Physiol (1985). 1995;79:1379–1385.
  • Fix SM, Borden MA, Dayton PA. Therapeutic gas delivery via microbubbles and liposomes. J Control Release. 2015;209:139–149.
  • Drago RS, Ragsdale RO, Eyman DP. A mechanism for the reaction of diethylamine with nitric oxide. J Am Chem Soc. 1961;83:4337–4339.
  • Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature. 1987;327:524–526.
  • Garthwaite J, Charles SL, Chess-Williams R. Endothelium-derived relaxing factor release on activation of NMDA receptors suggests role as intercellular messenger in the brain. Nature. 1988;336:385–388.
  • Marletta MA, Yoon PS, Iyengar R, et al. Macrophage oxidation of L-arginine to nitrite and nitrate: nitric oxide is an intermediate. Biochemistry. 1988;27:8706–8711.
  • Hibbs JB Jr., Taintor RR, Vavrin Z, et al. Nitric oxide: a cytotoxic activated macrophage effector molecule. Biochem Biophys Res Commun. 1988;157:87–94.
  • Moncada S, Radomski MW, Palmer RM. Endothelium-derived relaxing factor. Identification as nitric oxide and role in the control of vascular tone and platelet function. Biochem Pharmacol. 1988;37:2495–2501.
  • Hrabie JA, Keefer LK. Chemistry of the nitric oxide-releasing diazeniumdiolate (“nitrosohydroxylamine”) functional group and its oxygen-substituted derivatives. Chem Rev. 2002;102:1135–1154.
  • Hetrick EM, Schoenfisch MH. Reducing implant-related infections: active release strategies. Chem Soc Rev. 2006;35:780–789.
  • Nablo BJ, Prichard HL, Butler RD, et al. Inhibition of implant-associated infections via nitric oxide release. Biomaterials. 2005;26:6984–6990.
  • Riccio DA, Dobmeier KP, Hetrick EM, et al. Nitric oxide-releasing S-nitrosothiol-modified xerogels. Biomaterials. 2009;30:4494–4502.
  • Shin JH, Metzger SK, Schoenfisch MH. Synthesis of nitric oxide-releasing silica nanoparticles. J Am Chem Soc. 2007;129:4612–4619.
  • Shin JH, Schoenfisch MH. Inorganic/organic hybrid silica nanoparticles as a nitric oxide delivery scaffold. Chem Mater. 2008;20:239–249.
  • Riccio DA, Nugent JL, Schoenfisch MH. Stober synthesis of nitric oxide-releasing s-nitrosothiol-modified silica particles. Chem Mater. 2011;23:1727–1735.
  • Stasko NA, Schoenfisch MH. Dendrimers as a scaffold for nitric oxide release. J Am Chem Soc. 2006;128:8265–8271.
  • Stasko NA, Fischer TH, Schoenfisch MH. S-nitrosothiol-modified dendrimers as nitric oxide delivery vehicles. Biomacromolecules. 2008;9:834–841.
  • Mowery KA, Schoenfisch MH, Saavedra JE, et al. Preparation and characterization of hydrophobic polymeric films that are thromboresistant via nitric oxide release. Biomaterials. 2000;21:9–21.
  • Parzuchowski PG, Frost MC, Meyerhoff ME. Synthesis and characterization of polymethacrylate-based nitric oxide donors. J Am Chem Soc. 2002;124:12182–12191.
  • Coneski PN, Rao KS, Schoenfisch MH. Degradable nitric oxide-releasing biomaterials via post-polymerization functionalization of cross-linked polyesters. Biomacromolecules. 2010;11:3208–3215.
  • Coneski PN, Nash JA, Schoenfisch MH. Nitric oxide-releasing electrospun polymer microfibers. ACS Appl Mater Interfaces. 2011;3:426–432.
  • Suchyta DJ, Schoenfisch MH. Encapsulation of N-diazeniumdiolates within liposomes for enhanced nitric oxide donor stability and delivery. Mol Pharm. 2015;12:3569–3574.
  • Szoka F Jr., Papahadjopoulos D. Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation. Proc Natl Acad Sci U S A. 1978;75:4194–4198.
  • O’Callaghan DS, Savale L, Montani D, et al. Treatment of pulmonary arterial hypertension with targeted therapies. Nat Reviews Cardiol. 2011;8:526–538.
  • Barst RJ, Channick R, Ivy D, et al. Clinical perspectives with long-term pulsed inhaled nitric oxide for the treatment of pulmonary arterial hypertension. Pulm Circ. 2012;2:139–147.
  • Weinberger B, Fakhrzadeh L, Heck DE, et al. Inhaled nitric oxide primes lung macrophages to produce reactive oxygen and nitrogen intermediates. Am J Respir Crit Care Med. 1998;158:931–938.
  • Clutton-Brock J. Two cases of poisoning by contamination of nitrous oxide with higher oxides of nitrogen during anaesthesia. Br J Anaesth. 1967;39:388–392.
  • Hagan G, Pepke-Zaba J. Pulmonary hypertension, nitric oxide and nitric oxide-releasing compounds. Expert Rev Respir Med. 2011;5:163–171.
  • Miller OI, Tang SF, Keech A, et al. Rebound pulmonary hypertension on withdrawal from inhaled nitric oxide. Lancet (London, England). 1995;346:51–52.
  • Nahar K, Rashid J, Absar S, et al. Liposomal aerosols of nitric oxide (no) donor as a long-acting substitute for the ultra-short-acting inhaled NO in the treatment of PAH. Pharm Res. 2016;33:1696–1710.
  • Keefer LK, Nims RW, Davies KM, et al. “NONOates” (1-substituted diazen-1-ium-1,2-diolates) as nitric oxide donors: convenient nitric oxide dosage forms. Methods Enzymol. 1996;268:281–293.
  • Lam CF, Van Heerden PV, Ilett KF, et al. Two aerosolized nitric oxide adducts as selective pulmonary vasodilators for acute pulmonary hypertension. Chest. 2003;123:869–874.
  • Lam CF, Caterina P, Filion P, et al. The safety of aerosolized diethylenetriamine nitric oxide adduct after single-dose administration to anesthetized piglets and multiple-dose administration to conscious rats. Toxicol Appl Pharmacol. 2003;190:65–71.
  • Kliment CR, Tobolewski JM, Manni ML, et al. Extracellular superoxide dismutase protects against matrix degradation of heparan sulfate in the lung. Antioxid Redox Signal. 2008;10:261–268.
  • Urakami T, Jarvinen TA, Toba M, et al. Peptide-directed highly selective targeting of pulmonary arterial hypertension. Am J Pathol. 2011;178:2489–2495.
  • Götz F. Staphylococcus and biofilms. Mol Microbiol. 2002;43:1367–1378.
  • Singhal D, Foreman A, Jervis-Bardy J, et al. Staphylococcus aureus biofilms: nemesis of endoscopic sinus surgery. Laryngoscope. 2011;121:1578–1583.
  • Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent infections. Science. 1999;284:1318–1322.
  • Ha KR, Psaltis AJ, Butcher AR, et al. In vitro activity of mupirocin on clinical isolates of Staphylococcus aureus and its potential implications in chronic rhinosinusitis. Laryngoscope. 2008;118:535–540.
  • Jardeleza C, Rao S, Thierry B, et al. A novel nitric oxide-based therapeutic agent against Staphylococcus aureus biofilms. PLo One. 2014;9:e92117.
  • Chrysant SG, Glasser SP, Bittar N, et al. Efficacy and safety of extended-release isosorbide mononitrate for stable effort angina pectoris. Am J Cardiol. 1993;72:1249–1256.
  • Barraud N, Storey MV, Moore ZP, et al. Nitric oxide-mediated dispersal in single- and multi-species biofilms of clinically and industrially relevant microorganisms. Microb Biotechnol. 2009;2:370–378.
  • Hetrick EM, Shin JH, Paul HS, et al. Anti-biofilm efficacy of nitric oxide-releasing silica nanoparticles. Biomaterials. 2009;30:2782–2789.
  • Carpenter AW, Schoenfisch MH. Nitric oxide release part II. therapeutic applications. Chem Soc Rev. 2012;41:3742–3752.
  • Jones ML, Ganopolsky JG, Labbé A, et al. Antimicrobial properties of nitric oxide and its application in antimicrobial formulations and medical devices. Appl Microbiol Biotechnol. 2010;88:401–407.
  • Wink DA, Vodovotz Y, Laval J, et al. The multifaceted roles of nitric oxide in cancer. Carcinogenesis. 1998;19:711–721.
  • Hirst D, Robson T. Nitric oxide in cancer therapeutics: interaction with cytotoxic chemotherapy. Curr Pharm Des. 2010;16:411–420.
  • Jardeleza C, Thierry B, Rao S, et al. An in vivo safety and efficacy demonstration of a topical liposomal nitric oxide donor treatment for Staphylococcus aureus biofilm-associated rhinosinusitis. Transl Res. 2015;166(6):683–692.
  • Chen ZF, Qin QP, Qin JL, et al. Water-soluble ruthenium(ii) complexes with chiral 4-(2,3-dihydroxypropyl)-formamide oxoaporphine (FOA): in vitro and in vivo anticancer activity by stabilization of g-quadruplex DNA, inhibition of telomerase activity, and induction of tumor cell apoptosis. J Med Chem. 2015;58:4771–4789.
  • Hummer AA, Heffeter P, Berger W, et al. X-ray absorption near edge structure spectroscopy to resolve the in vivo chemistry of the redox-active indazolium trans-[Tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019). J Med Chem. 2013;56:1182–1196.
  • Cardoso CR, Lima MV, Cheleski J, et al. Luminescent ruthenium complexes for theranostic applications. J Med Chem. 2014;57:4906–4915.
  • Caruso F, Rossi M, Benson A, et al. Ruthenium-arene complexes of curcumin: X-ray and density functional theory structure, synthesis, and spectroscopic characterization, in vitro antitumor activity, and DNA docking studies of (p-cymene)Ru(curcuminato)chloro. J Med Chem. 2012;55:1072–1081.
  • Schluga P, Hartinger CG, Egger A, et al. Redox behavior of tumor-inhibiting ruthenium(III) complexes and effects of physiological reductants on their binding to GMP. Dalton Trans. 2006;(14):1796–1802.
  • Frik M, Martinez A, Elie BT, et al. In vitro and in vivo evaluation of water-soluble iminophosphorane ruthenium(II) compounds. A potential chemotherapeutic agent for triple negative breast cancer. J Med Chem. 2014;57:9995–10012.
  • Aman F, Hanif M, Siddiqui WA, et al. Anticancer ruthenium(η6-p-cymene) complexes of nonsteroidal anti-inflammatory drug derivatives. Organometallics. 2014;33:5546–5553.
  • Pettinari R, Marchetti F, Condello F, et al. Ruthenium(II)–arene rapta type complexes containing curcumin and bisdemethoxycurcumin display potent and selective anticancer activity. Organometallics. 2014;33:3709–3715.
  • Robles-Escajeda E, Martinez A, Varela-Ramirez A, et al. Analysis of the cytotoxic effects of ruthenium-ketoconazole and ruthenium-clotrimazole complexes on cancer cells. Cell Biol Toxicol. 2013;29:431–443.
  • Kurzwernhart A, Kandioller W, Enyedy EA, et al. 3-Hydroxyflavones vs. 3-hydroxyquinolinones: structure-activity relationships and stability studies on Ru(II)(arene) anticancer complexes with biologically active ligands. Dalton Trans. 2013;42:6193–6202.
  • Kurzwernhart A, Kandioller W, Bachler S, et al. Structure-activity relationships of targeted RuII(eta6-p-cymene) anticancer complexes with flavonol-derived ligands. J Med Chem. 2012;55:10512–10522.
  • Castonguay A, Doucet C, Juhas M, et al. New ruthenium(II)-letrozole complexes as anticancer therapeutics. J Med Chem. 2012;55:8799–8806.
  • Muhlgassner G, Bartel C, Schmid WF, et al. Biological activity of ruthenium and osmium arene complexes with modified paullones in human cancer cells. J Inorg Biochem. 2012;116:180–187.
  • Rubner G, Bensdorf K, Wellner A, et al. Synthesis and biological activities of transition metal complexes based on acetylsalicylic acid as neo-anticancer agents. J Med Chem. 2010;53:6889–6898.
  • Carneiro ZA, De Moraes JC, Rodrigues FP, et al. Photocytotoxic activity of a nitrosyl phthalocyanine ruthenium complex – a system capable of producing nitric oxide and singlet oxygen. J Inorg Biochem. 2011;105:1035–1043.
  • Heinrich TA, Tedesco AC, Fukuto JM, et al. Production of reactive oxygen and nitrogen species by light irradiation of a nitrosyl phthalocyanine ruthenium complex as a strategy for cancer treatment. Dalton Trans. 2014;43:4021–4025.
  • De Lima RG, Silva BR, Da Silva RS, et al. Ruthenium complexes as NO donors for vascular relaxation induction. Molecules. 2014;19:9628–9654.
  • Pereira Ade C, Ford PC, Da Silva RS, et al. Ruthenium-nitrite complex as pro-drug releases NO in a tissue and enzyme-dependent way. Nitric Oxide. 2011;24:192–198.
  • Rodrigues FP, Carneiro ZA, Mascharak P, et al. Incorporation of a ruthenium nitrosyl complex into liposomes, the nitric oxide released from these liposomes and HepG2 cell death mechanism. Coord Chem Rev. 2016;306:701–707.
  • Bordini J, Ford PC, Tfouni E. Photochemical release of nitric oxide from a regenerable, sol-gel encapsulated Ru-salen-nitrosyl complex. Chem Commun (Camb). 2005;(33):4169–4171.
  • Mitchell-Koch JT, Reed TM, Borovik AS. Light-activated transfer of nitric oxide from a porous material. Angew Chem Int Ed Engl. 2004;43:2806–2809.
  • Gomes AJ, Barbougli PA, Espreafico EM, et al. trans-[Ru(NO)(NH3)4(py)](BF4)3.H2O encapsulated in PLGA microparticles for delivery of nitric oxide to B16-F10 cells: cytotoxicity and phototoxicity. J Inorg Biochem. 2008;102:757–766.
  • Halpenny GM, Mascharak PK. Accelerated photorelease of NO from {Ru-NO}6 nitrosyls containing carboxamido-N and carboxylato-O donors: syntheses, structures, and photochemistry. Inorg Chem. 2009;48:1490–1497.
  • Rodrigues FP, Pestana CR, Polizello AC, et al. Release of NO from a nitrosyl ruthenium complex through oxidation of mitochondrial NADH and effects on mitochondria. Nitric Oxide. 2012;26:174–181.
  • Heinecke J, Ford PC. Formation of cysteine sulfenic acid by oxygen atom transfer from nitrite. J Am Chem Soc. 2010;132:9240–9243.
  • Kremer JM, Esker MW, Pathmamanoharan C, et al. Vesicles of variable diameter prepared by a modified injection method. Biochemistry. 1977;16:3932–3935.
  • Deamer DW. Preparation and properties of ether-injection liposomes. Ann N Y Acad Sci. 1978;308:250–258.
  • Machy P, Leserman LD. Small liposomes are better than large liposomes for specific drug delivery in vitro. Biochim Biophys Acta. 1983;730:313–320.
  • Heath TD, Lopez NG, Papahadjopoulos D. The effects of liposome size and surface charge on liposome-mediated delivery of methotrexate-gamma-aspartate to cells in vitro. Biochim Biophys Acta. 1985;820:74–84.
  • Nakanishi K, Koshiyama T, Iba S, et al. Lipophilic ruthenium salen complexes: incorporation into liposome bilayers and photoinduced release of nitric oxide. Dalton Trans. 2015;44:14200–14203.
  • Pescador P, Brodersen N, Scheidt HA, et al. Microtubes self-assembled from a cholesterol-modified nucleoside. Chem Commun (Camb). 2010;46:5358–5360.
  • Bangham AD, Standish MM, Watkins JC. Diffusion of univalent ions across the lamellae of swollen phospholipids. J Mol Biol. 1965;13:238–252.
  • De Leo M, Ford PC. Reversible photolabilization of no from chromium(iii)-coordinated nitrite. New strategy nitric oxide delivery. J Am Chem Society. 1999;121:1980–1981.
  • DeLeo MA, Ford PC. Photoreactions of coordinated nitrite ion. reversible nitric oxide labilization from the chromium(III) complex [trans-Cr(cyclam)(ONO)2]+. Coord Chem Rev. 2000;208:47–59.
  • DeRosa F, Bu X, Ford PC. Chromium(III) complexes for photochemical nitric oxide generation from coordinated nitrite: synthesis and photochemistry of macrocyclic complexes with pendant chromophores, trans-[Cr(L)(ONO)2]BF4. Inorg Chem. 2005;44:4157–4165.
  • Ostrowski AD, Absalonson RO, Leo MAD, et al. Photochemistry of trans-Cr(cyclam)(ONO)2+, a nitric oxide precursor. Inorg Chem. 2011;50:4453–4462.
  • Ostrowski AD, Deakin SJ, Azhar B, et al. Nitric oxide photogeneration from trans-Cr(cyclam)(ONO)(2)(+) in a reducing environment. activation of soluble guanylyl cyclase and arterial vasorelaxation. J Med Chem. 2010;53:715–722.
  • Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58.
  • Brozik A, Hegedus C, Erdei Z, et al. Tyrosine kinase inhibitors as modulators of ATP binding cassette multidrug transporters: substrates, chemosensitizers or inducers of acquired multidrug resistance? Expert Opin Drug Metab Toxicol. 2011;7:623–642.
  • Agarwal S, Sane R, Oberoi R, et al. Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain. Expert Rev Mol Med. 2011;13:e17.
  • Amiri-Kordestani L, Basseville A, Kurdziel K, et al. Targeting MDR in breast and lung cancer: discriminating its potential importance from the failure of drug resistance reversal studies. Drug Resist Updat. 2012;15:50–61.
  • Shaffer BC, Gillet JP, Patel C, et al. Drug resistance: still a daunting challenge to the successful treatment of AML. Drug Resist Updat. 2012;15:62–69.
  • Riganti C, Miraglia E, Viarisio D, et al. Nitric oxide reverts the resistance to doxorubicin in human colon cancer cells by inhibiting the drug efflux. Cancer Res. 2005;65:516–525.
  • Fruttero R, Crosetti M, Chegaev K, et al. Phenylsulfonylfuroxans as modulators of multidrug-resistance-associated protein-1 and P-glycoprotein. J Med Chem. 2010;53:5467–5475.
  • Chegaev K, Riganti C, Lazzarato L, et al. Nitric oxide donor doxorubicins accumulate into Doxorubicin-resistant human colon cancer cells inducing cytotoxicity. ACS Med Chem Lett. 2011;2:494–497.
  • Apetoh L, Mignot G, Panaretakis T, et al. Immunogenicity of anthracyclines: moving towards more personalized medicine. Trends Mol Med. 2008;14:141–151.
  • Pedrini I, Gazzano E, Chegaev K, et al. Liposomal nitrooxy-doxorubicin: one step over caelyx in drug-resistant human cancer cells. Mol Pharm. 2014;11:3068–3079.
  • Markman M. Pegylated liposomal doxorubicin in the treatment of cancers of the breast and ovary. Expert Opin Pharmacother. 2006;7:1469–1474.
  • Leonard RC, Williams S, Tulpule A, et al. Improving the therapeutic index of anthracycline chemotherapy: focus on liposomal doxorubicin (Myocet). Breast (Edinburgh, Scotland). 2009;18:218–224.
  • McEwan C, Owen J, Stride E, et al. Oxygen carrying microbubbles for enhanced sonodynamic therapy of hypoxic tumours. J Control Release. 2015;203:51–56.
  • Kheir JN, Scharp LA, Borden MA, et al. Oxygen gas-filled microparticles provide intravenous oxygen delivery. Sci Transl Med. 2012;4:140ra88.
  • Swanson EJ, Mohan V, Kheir J, et al. Phospholipid-stabilized microbubble foam for injectable oxygen delivery. Langmuir. 2010;26:15726–15729.
  • Feshitan JA, Legband ND, Borden MA, et al. Systemic oxygen delivery by peritoneal perfusion of oxygen microbubbles. Biomaterials. 2014;35:2600–2606.
  • Legband ND, Feshitan JA, Borden MA, et al. Evaluation of peritoneal microbubble oxygenation therapy in a rabbit model of hypoxemia. IEEE Trans Biomed Eng. 2015;62:1376–1382.
  • Wang C, Yang F, Xu Z, et al. Intravenous release of NO from lipidic microbubbles accelerates deep vein thrombosis resolution in a rat model. Thromb Res. 2013;131:e31–8.
  • Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. N Engl J Med. 2008;358:2765–2775.
  • Alban S. Adverse effects of heparin. Handb Exp Pharmacol. 2012;207:211–263.
  • Panovsky R, Meluzin J, Janousek S, et al. Cell therapy in patients with left ventricular dysfunction due to myocardial infarction. Echocardiography. 2008;25:888–897.
  • Teupe C, Richter S, Fisslthaler B, et al. Vascular gene transfer of phosphomimetic endothelial nitric oxide synthase (S1177D) using ultrasound-enhanced destruction of plasmid-loaded microbubbles improves vasoreactivity. Circulation. 2002;105:1104–1109.
  • Zen K, Okigaki M, Hosokawa Y, et al. Myocardium-targeted delivery of endothelial progenitor cells by ultrasound-mediated microbubble destruction improves cardiac function via an angiogenic response. J Mol Cell Cardiol. 2006;40:799–809.
  • Hirst D, Robson T. Targeting nitric oxide for cancer therapy. J Pharm Pharmacol. 2007;59:3–13.
  • Lee SY, Rim Y, McPherson DD, et al. A novel liposomal nanomedicine for nitric oxide delivery and breast cancer treatment. Biomed Mater Eng. 2014;24:61–67.
  • Kim H, Britton GL, Peng T, et al. Nitric oxide-loaded echogenic liposomes for treatment of vasospasm following subarachnoid hemorrhage. Int J Nanomedicine. 2014;9:155–165.
  • Sutton JT, Raymond JL, Verleye MC, et al. Pulsed ultrasound enhances the delivery of nitric oxide from bubble liposomes to ex vivo porcine carotid tissue. Int J Nanomedicine. 2014;9:4671–4683.
  • Huang SL, Kee PH, Kim H, et al. Nitric oxide-loaded echogenic liposomes for nitric oxide delivery and inhibition of intimal hyperplasia. J Am Coll Cardiol. 2009;54:652–659.
  • Pluta RM. Delayed cerebral vasospasm and nitric oxide: review, new hypothesis, and proposed treatment. Pharmacol Ther. 2005;105:23–56.
  • Hanggi D, Steiger HJ. Nitric oxide in subarachnoid haemorrhage and its therapeutics implications. Acta Neurochir (Wien). 2006;148:605–613. discussion 13.
  • Tsao PS, McEvoy LM, Drexler H, et al. Enhanced endothelial adhesiveness in hypercholesterolemia is attenuated by L-arginine. Circulation. 1994;89:2176–2182. Epub 1994/05/01.
  • Subbotin VM. Analysis of arterial intimal hyperplasia: review and hypothesis. Theor Biol Med Model. 2007;4:41.
  • Jagadeesha DK, Miller FJ Jr., Bhalla RC. Inhibition of apoptotic signaling and neointimal hyperplasia by tempol and nitric oxide synthase following vascular injury. J Vasc Res. 2009;46:109–118.
  • Lima ES, Bonini MG, Augusto O, et al. Nitrated lipids decompose to nitric oxide and lipid radicals and cause vasorelaxation. Free Radic Biol Med. 2005;39:532–539.
  • Faine LA, Cavalcanti DM, Rudnicki M, et al. Bioactivity of nitrolinoleate: effects on adhesion molecules and CD40-CD40L system. J Nutr Biochem. 2010;21:125–132.
  • Schopfer FJ, Baker PRS, Giles G, et al. Fatty acid transduction of nitric oxide signaling: nitrolinoleic acid is a hydrophobically stabilized nitric oxide donor. J Biol Chem. 2005;280:19289–19297.
  • Napolitano A, Camera E, Picardo M, et al. Acid-promoted reactions of ethyl linoleate with nitrite ions: formation and structural characterization of isomeric nitroalkene, nitrohydroxy, and novel 3-nitro-1,5-hexadiene and 1,5-dinitro-1,3-pentadiene products. J Org Chem. 2000;65:4853–4860.
  • O’Donnell VB, Eiserich JP, Bloodsworth A, et al. [47] Nitration of unsaturated fatty acids by nitric oxide-derived reactive species. Methods Enzymol. 1999;301:454–470. Academic Press.
  • Bhowmick D, Mugesh G. Insights into the catalytic mechanism of synthetic glutathione peroxidase mimetics. Org Biomol Chem. 2015;13:10262–10272.
  • Yang Z, Yang Y, Xiong K, et al. Nitric oxide producing coating mimicking endothelium function for multifunctional vascular stents. Biomaterials. 2015;63:80–92.
  • Shaaban S, Negm A, Ashmawy AM, et al. Combinatorial synthesis, in silico, molecular and biochemical studies of tetrazole-derived organic selenides with increased selectivity against hepatocellular carcinoma. Eur J Med Chem. 2016;122:55–71.
  • Chen M-X, Li B-K, Yin D-K, et al. Layer-by-layer assembly of chitosan stabilized multilayered liposomes for paclitaxel delivery. Carbohydr Polym. 2014;111:298–304.
  • Gibis M, Zeeb B, Weiss J. Formation, characterization, and stability of encapsulated hibiscus extract in multilayered liposomes. Food Hydrocoll. 2014;38:28–39.
  • Jain S, Kumar D, Swarnakar NK, et al. Polyelectrolyte stabilized multilayered liposomes for oral delivery of paclitaxel. Biomaterials. 2012;33:6758–6768.
  • Luo R, Liu Y, Yao H, et al. Copper-incorporated collagen/catechol film for in situ generation of nitric oxide. ACS Biomater Sci Eng. 2015;1:771–779.
  • Weng Y, Song Q, Zhou Y, et al. Immobilization of selenocystamine on TiO2 surfaces for in situ catalytic generation of nitric oxide and potential application in intravascular stents. Biomaterials. 2011;32:1253–1263.
  • Gallo A, Mani G. A stent for co-delivering paclitaxel and nitric oxide from abluminal and luminal surfaces: preparation, surface characterization, and in vitro drug release studies. Appl Surf Sci. 2013;279:216–232.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.