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

Surfactants: their critical role in enhancing drug delivery to the lungs

    Javier O Morales

    Division of Pharmaceutics, College of Pharmacy, University of Texas at Austin, 2409 West University Avenue, PHR 4.214, Austin, TX 78712, USA

    ,
    Jay I Peters

    Department of Pulmonary Care, College of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA

    &
    Published Online:https://doi.org/10.4155/tde.11.15

    For local lung conditions and diseases, pulmonary drug delivery has been widely used for more than 50 years now. A more recent trend involves the pulmonary route as a systemic drug-delivery target. Advantages such as avoidance of the gastrointestinal environment, different enzyme content compared with the intestine, and avoidance of first-pass metabolism make the lung an alternative route for the systemic delivery of actives. However, the lung offers barriers to absorption such as a surfactant layer, epithelial surface lining fluid, epithelial monolayer, interstitium and basement membrane, and capillary endothelium. Many delivery strategies have been developed in order to overcome these limitations. The use of surfactants is one of these approaches and their role in enhancing pulmonary drug delivery is reviewed in this article. A systematic review of the literature relating to the effect of surfactants on formulations for pulmonary delivery was conducted. Specifically, research reporting enhancement of in vivo performance was focused on. The effect of the addition of surfactants such as phospholipids, bile salts, non-ionic, fatty acids, and liposomes as phospholipid-containing carriers on the enhancement of therapeutic outcomes of drugs for pulmonary delivery was compiled. The main use attributed to surfactants in pulmonary drug delivery is as absorption enhancers by mechanisms of action not yet fully understood. Furthermore, surfactants have been used to improve the delivery of inhaled drugs in various additional strategies discussed herein.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Hoover JL, Rush BD, Wilkinson KF et al. Peptides are better absorbed from the lung than the gut in the rat. Pharm. Res.9(8),1103–1106 (1992).
    • Banga AK. Pulmonary and other mucosal delivery of therapeutics peptides and proteins. In: Therapeutic Peptides and Proteins Formulation, Processing, and Delivery Systems (2nd Edition). CRC/Taylor & Francis, Boca Raton, FL, USA 291–326 (2006).
    • Patton JS. Mechanisms of macromolecule absorption by the lungs. Adv. Drug Deliv. Rev.19(1),3–36 (1996).
    • Thurlbeck WM. The internal surface area of nonemphysematous lungs. Am. Rev. Respir. Dis.95(5),765–773 (1967).
    • Weibel ER. Morphological basis of alveolar-capillary gas exchange. Physiol. Rev.53(2),419 (1973).
    • Agu U, Ugwoke I, Armand M, Kinget R, Verbeke N. The lung as a route for systemic delivery of therapeutic proteins and peptides. Respir. Res.2(4),198 (2001).▪▪ Excellent article on the mechanisms for drug absorption in the lungs with a detailed description of the lung physiology.
    • Clark AR, Shire SJ. Formulation of proteins for pulmonary drug delivery. In: Protein Formulation and Delivery. McNally EJ (Ed.). M Dekker, NY, USA 201–234 (2000).
    • Groneberg DA, Witt C, Wagner U, Chung KF, Fischer A. Fundamentals of pulmonary drug delivery. Respir. Med.97(4),382–387 (2003).
    • Yu J, Chien YW. Pulmonary drug delivery: physiologic and mechanistic aspects. Crit. Rev. Ther. Drug Carrier Syst.14(4),395 (1997).
    • 10  Newman SP, Chan HK. In vitro/in vivo comparisons in pulmonary drug delivery. J. Aerosol Med. Pulm. Drug Deliv.21(1),77–84 (2008).
    • 11  Taylor KMG, McCallion ONM. Ultrasonic nebulisers for pulmonary drug delivery. Int. J. Pharm.153(1),93–104 (1997).
    • 12  Dennis J, Nerbrink O. New nebulizer technology. In: Drug Delivery to the Lung. Bisgaard H, O’Callaghan C, Smaldone GC (Eds). Marcel Dekker, NY, USA (2002).
    • 13  Rubin BK, Fink JB. Optimizing aerosol delivery by pressurized metered-dose inhalers. Respir. care50(9),1191–1200 (2005).
    • 14  McDonald KJ, Martin GP. Transition to CFC-free metered dose inhalers – into the new millennium. Int. J. Pharm.201(1),89–107 (2000).
    • 15  Bell J, Newman S. The rejuvenated pressurised metered dose inhaler. Expert Opin. Drug Deliv.4(3),215–234 (2007).
    • 16  Newman SP, Busse WW. Evolution of dry powder inhaler design, formulation, and performance. Respir. Med.96(5),293–304 (2002).
    • 17  Atkins PJ. Dry powder inhalers: an overview. Respir. Care50(10),1304–1312 (2005).
    • 18  Ilowite JS, Gorvoy JD, Smaldone GC. Quantitative deposition of aerosolized gentamicin in cystic fibrosis. Am. Rev. Respir. Dis.136(6),1445–1449 (1987).
    • 19  Collis GG, Cole CH, Le Souef PN. Dilution of nebulised aerosols by air entrainment in children. Lancet336(8711),341–343 (1990).
    • 20  Köhler E, Sollich V, Schuster-Wonka R, Hühnerbein J. Lung deposition in cystic fibrosis patients using an ultrasonic or a jet nebulizer. J. Aerosol Med.16(1),37–46 (2003).
    • 21  Le Brun PPH, de Boer AH, Gjaltema D et al. Inhalation of tobramycin in cystic fibrosis: part 2: optimization of the tobramycin solution for a jet and an ultrasonic nebulizer. Int. J. Pharm.189(2),215–225 (1999).
    • 22  Geller DE, Pitlick WH, Nardella PA, Tracewell WG, Ramsey BW. Pharmacokinetics and bioavailability of aerosolized tobramycin in cystic fibrosis. Chest122(1),219–226 (2002).
    • 23  Dhand R. Nebulizers that use a vibrating mesh or plate with multiple apertures to generate aerosol. Respir. Care47(12),1406–1418 (2002).
    • 24  Byrne NM, Keavey PM, Perry JD, Gould FK, Spencer DA. Comparison of lung deposition of colomycin using the HaloLite and the Pari LC Plus nebulisers in patients with cystic fibrosis. Arch. Dis. Child.88(8),715–718 (2003).
    • 25  Newman SP, Flora M, Hirst PH et al. Pharmacoscintigraphy of TOBI in the PARI LC PLUS, and the AeroDose inhaler. J. Aerosol Med.14,388 (2001).
    • 26  Coates AL, Green M, Leung K et al. The challenges of quantitative measurement of lung deposition using 99mTc-DTPA from delivery systems with very different delivery times. J. Aerosol Med.20(3),320–330 (2007).
    • 27  Nikander K, Prince I, Coughlin S, Warren S, Taylor G. Lung deposition of 99mTc-DTPA delivered with the I-neb® adaptive aerosol delivery (AAD®) System. J. Aerosol Med.20,192–193 (2007).
    • 28  Meltzer EO. Formulation considerations of intranasal corticosteroids for the treatment of allergic rhinitis. Ann. Allergy, Asthma Immunol.98(1),12–21 (2007).
    • 29  Newman S. Principles of metered-dose inhaler design. Respir. Care50(9),1177–1190 (2005).
    • 30  Crowder TM, Louey MD, Sethuraman VV, Smyth HDC, Hickey AJ. 2001: an odyssey in inhaler formulations and design. Pharm. Technol.25(7),99–113 (2001).
    • 31  Olmsted SS, Padgett JL, Yudin AI et al. Diffusion of macromolecules and virus-like particles in human cervical mucus. Biophys. J.81(4),1930–1937 (2001).
    • 32  Saltzman W, Radomsky M, Whaley K, Cone R. Antibody diffusion in human cervical mucus. Biophys. J.66(2),508–515 (1994).
    • 33  Lai SK, O’Hanlon DE, Harrold S et al. Rapid transport of large polymeric nanoparticles in fresh undiluted human mucus. Proc. Natl Acad. Sci. USA104(5),1482–1487 (2007).
    • 34  DeFelippis MR, Sukumar M, Rajagopalan N. Peptides and proteins: non-invasive delivery. In: Encyclopedia of Pharmaceutical Technology (3rd Edition). Swarbrick J (Ed.). Informa Healthcare, NY, USA, 2692–2712 (2006).
    • 35  Hussain A, Arnold JJ, Khan MA, Ahsan F. Absorption enhancers in pulmonary protein delivery. J. Control. Release94(1),15–24 (2004).▪▪ Comprehensive review of the various absorption enhancers utilized in lung-delivery formulations.
    • 36  Myers MA, Thomas DA, Straub L et al. Pulmonary effects of chronic exposure to liposome aerosols in mice. Exp. Lung Res.19(1),1–19 (1993).
    • 37  Thomas D, Myers M, Wichert B, Schreier H, Gonzalez-Rothi R. Acute effects of liposome aerosol inhalation on pulmonary function in healthy human volunteers. Chest99(5),1268–1270 (1991).
    • 38  Waldrep J, Knight C, Black M et al. Pulmonary delivery of beclomethasone liposome aerosol in volunteers. Chest111(2),316–323 (1997).
    • 39  Possmayer F, Nag K, Rodriguez K, Qanbar R, Schürch S. Surface activity in vitro: role of surfactant proteins. Comp. Biochem. Physiol. A Mol. Integr. Physiol.129(1),209–220 (2001).
    • 40  FDA Guidance for Industry. Guidance on the Labeling of Certain Uses of Lecithin Derived from Soy Under Section 403(w) of the Federal Food, Drug, and Cosmetic Act. US FDA, Rockville, MD, USA (2006).
    • 41  Rawat M, Singh D, Saraf S, Saraf S. Lipid carriers: a versatile delivery vehicle for proteins and peptides. Yakugaku Zasshi128(2),269–280 (2008).
    • 42  Sebti T, Amighi K. Preparation and in vitro evaluation of lipidic carriers and fillers for inhalation. Eur. J. Pharm. Biopharm.63(1),51–58 (2006).
    • 43  Desai TR, Hancock REW, Finlay WH. Delivery of liposomes in dry powder form: aerodynamic dispersion properties. Eur. J. Pharm. Sci.20(4–5),459–467 (2003).
    • 44  Sebti T, Pilcer G, Van Gansbeke B et al. Pharmacoscintigraphic evaluation of lipid dry powder budesonide formulations for inhalation. Eur. J. Pharm. Biopharm.64(1),26–32 (2006).
    • 45  Hirst P, Newman S, Clark D, Hertog M. Lung deposition of budesonide from the novel dry powder inhaler Airmax™. Respir. Med.96(6),389–396 (2002).
    • 46  Vanbever R, Mintzes JD, Wang J et al. Formulation and physical characterization of large porous particles for inhalation. Pharm. Res.16(11),1735–1742 (1999).
    • 47  Wang J, Ben-Jebria A, Edwards D. Inhalation of estradiol for sustained systemic delivery. J. Aerosol Med.12(1),27–36 (1999).
    • 48  Bosquillon C, Préat V, Vanbever R. Pulmonary delivery of growth hormone using dry powders and visualization of its local fate in rats. J. Control. Release96(2),233–244 (2004).
    • 49  Lindmark T, Kimura Y, Artursson P. Absorption enhancement through intracellular regulation of tight junction permeability by medium chain fatty acids in Caco-2 cells. J. Pharm. Exper. Ther.284(1),362–369 (1998).
    • 50  Codrons V, Vanderbist F, Ucakar B, Preat V, Vanbever R. Impact of formulation and methods of pulmonary delivery on absorption of parathyroid hormone (1–34) from rat lungs. J. Pharm. Sci.93(5),1241–1252 (2004).
    • 51  Mitra R, Pezron I, Li Y, Mitra AK. Enhanced pulmonary delivery of insulin by lung lavage fluid and phospholipids. Int. J. Pharm.217(1–2),25–31 (2001).
    • 52  Ungaro F, Giovino C, Coletta C et al. Engineering gas-foamed large porous particles for efficient local delivery of macromolecules to the lung. Eur. J. Pharm. Sci.41(1),60–70 (2010).
    • 53  Dellamary L, Tarara T, Smith D et al. Hollow porous particles in metered dose inhalers. Pharm. Res.17(2),168–174 (2000).
    • 54  Duddu S, Sisk S, Walter Y et al. Improved lung delivery from a passive dry powder inhaler using an engineered PulmoSphere® powder. Pharm. Res.19(5),689–695 (2002).
    • 55  Newhouse M, Hirst P, Duddu S et al. Inhalation of a dry powder tobramycin pulmosphere formulation in healthy volunteers. Chest124(1),360–366 (2003).
    • 56  Hadinoto K, Phanapavudhikul P, Kewu Z, Tan RB. Dry powder aerosol delivery of large hollow nanoparticulate aggregates as prospective carriers of nanoparticulate drugs: effects of phospholipids. Int. J. Pharm.333(1–2),187–198 (2007).
    • 57  Grenha A, Seijo B, Remuñán-López C. Microencapsulated chitosan nanoparticles for lung protein delivery. Eur. J. Pharm. Sci.25(4–5),427–437 (2005).
    • 58  Hadinoto K, Phanapavudhikul P, Kewu Z, Tan RBH. Novel formulation of large hollow nanoparticles aggregates as potential carriers in inhaled delivery of nanoparticulate drugs. Ind. Eng. Chem. Res.45(10),3697–3706 (2006).▪ Report on the development of large hollow nanoparticle aggregates and in vitro characterization.
    • 59  Tsapis N, Bennett D, Jackson B, Weitz DA, Edwards DA. Trojan particles: large porous carriers of nanoparticles for drug delivery. Proc. Natl Acad. Sci. USA99(19),12001–12005 (2002).
    • 60  Evora C, Soriano I, Rogers RA et al. Relating the phagocytosis of microparticles by alveolar macrophages to surface chemistry: the effect of 1,2-dipalmitoylphosphatidylcholine. J. Control. Release51(2–3),143–152 (1998).
    • 61  Jones BG, Dickinson PA, Gumbleton M, Kellaway IW. Lung surfactant phospholipids inhibit the uptake of respirable microspheres by the alveolar macrophage NR8383. J. Pharm. Pharmacol.54,1065–1072 (2002).
    • 62  Van Oort M, Sacchetti M. Spray-drying and supercritical fluid particle generation techniques. In: Inhalation Aerosols: Physical and Biological Basis for Therapy (2nd Edition). Hickey AJ (Ed.). Informa Healthcare, Hoboken, NJ, USA 307–346 (2007).
    • 63  Bosquillon C, Lombry C, Préat V, Vanbever R. Influence of formulation excipients and physical characteristics of inhalation dry powders on their aerosolization performance. J. Control. Release70(3),329–339 (2001).
    • 64  Malik S, Washington C, Purewal TS. Solution and adsorption behavior of lecithin surfactants in CFC suspensions: a light scattering study in aerosol propellants. Int. J. Pharm.186(1),63–69 (1999).
    • 65  Saso Y, Kondo S, Seki T, Morimoto K. Formulation design and pharmaceutical evaluation of an HFA 227-based furosemide metered dose inhaler. STP Pharma Sci.14(2),135–140 (2004).▪ Report on the use of lecithin as a coating agent of furosemide particles to be dispersed in an hydrofluoroalkane propellant-based metered dose inhaler.
    • 66  Tarara T, Hartman M, Gill H, Kennedy A, Weers J. Characterization of suspension-based metered dose inhaler formulations composed of spray-dried budesonide microcrystals dispersed in HFA-134a. Pharm. Res.21(9),1607–1614 (2004).
    • 67  Saso Y, Seki T, Fukuchi R, Chono S, Morimoto K. Effect of lecithin coating on the pulmonary absorption of furosemide in rats. Bio. Pharm. Bull.29(7),1445–1448 (2006).
    • 68  Sommerville ML, Cain JB, Johnson CS Jr, Hickey AJ. Lecithin inverse microemulsions for the pulmonary delivery of polar compounds utilizing dimethylether and propane as propellants. Pharm. Devel. Technol.5(2),219–230 (2000).
    • 69  Hiranita T, Nakamura S, Kawachi M et al. Miscibility behavior of dipalmitoylphosphatidylcholine with a single-chain partially fluorinated amphiphile in Langmuir monolayers. J. Colloid Interface Sci.265(1),83–92 (2003).
    • 70  Courrier HM, Krafft MP, Butz N et al. Evaluation of cytotoxicity of new semi-fluorinated amphiphiles derived from dimorpholinophosphate. Biomaterials24(4),689–696 (2003).
    • 71  Courrier H, Pons F, Lessinger J et al. In vivo evaluation of a reverse water-in-fluorocarbon emulsion stabilized with a semifluorinated amphiphile as a drug delivery system through the pulmonary route. Int. J. Pharm.282(1–2),131–140 (2004).
    • 72  Saso Y, Seki T, Chono S, Morimoto K. An HFA 227-based metered dose inhaler containing furosemide coated with lecithin. J. Drug Deliv. Sci. Technol.16(2),147–150 (2006).
    • 73  Bi R, Zhang N. Liposomes as a carrier for pulmonary delivery of peptides and proteins. J. Biomed. Nanotechnol.3(4),332–341 (2007).
    • 74  Pilcer G, Amighi K. Formulation strategy and use of excipients in pulmonary drug delivery. Int. J. Pharm.392(1–2),1–19 (2010).▪▪ Comprehensive survey of excipients used in the formulations of nebulizers, metered dose inhalers and dry powder inhalers with major focus on the latter.
    • 75  Iacono A, Smaldone G, Keenan R et al. Dose-related reversal of acute lung rejection by aerosolized cyclosporine. Am. J. Respir. Crit. Care Med.155(5),1690–1698 (1997).
    • 76  Mitruka SN, Pham SM, Zeevi A et al. Aerosol cyclosporine prevents acute allograft rejection in experimental lung transplantation. J. Thorac. Cardiovasc. Surg.115(1),28–37 (1998).
    • 77  Letsou GV, Safi HJ, Reardon MJ et al. Pharmacokinetics of liposomal aerosolized cyclosporine A for pulmonary immunosuppression. Ann. Thorac. Surg.68(6),2044–2048 (1999).
    • 78  Lo Y, Tsai J, Kuo J. Liposomes and disaccharides as carriers in spray-dried powder formulations of superoxide dismutase. J. Control. Release94(2–3),259–272 (2004).
    • 79  Chono S, Fukuchi R, Seki T, Morimoto K. Aerosolized liposomes with dipalmitoyl phosphatidylcholine enhance pulmonary insulin delivery. J. Control. Release137(2),104–109 (2009).
    • 80  Kobayashi S, Kondo S, Juni K. Study on pulmonary delivery of salmon calcitonin in rats: effects of protease inhibitors and absorption enhancers. Pharm. Res.11(9),1239–1243 (1994).
    • 81  Kobayashi S, Kondo S, Juni K. Pulmonary delivery of salmon calcitonin dry powders containing absorption enhancers in rats. Pharm. Res.13(1),80–83 (1996).
    • 82  Zaru M, Mourtas S, Klepetsanis P, Fadda AM, Antimisiaris SG. Liposomes for drug delivery to the lungs by nebulization. Eur. J. Pharm. Biopharm.67(3),655–666 (2007).
    • 83  Bridges PA, Taylor KM. An investigation of some of the factors influencing the jet nebulisation of liposomes. Int. J. Pharm.204(1–2),69–79 (2000).
    • 84  Gürsoy A, Kut E, ÖzkIrImlI S. Co-encapsulation of isoniazid and rifampicin in liposomes and characterization of liposomes by derivative spectroscopy. Int. J. Pharm.271(1–2),115–123 (2004).
    • 85  Zaru M, Manca M, Fadda AM, Antimisiaris SG. Chitosan-coated liposomes for delivery to lungs by nebulisation. Colloids Surf. B Biointerfaces71(1),88–95 (2009).
    • 86  Darwis Y, Kellaway IW. Nebulisation of rehydrated freeze-dried beclomethasone dipropionate liposomes. Int. J. Pharm.215(1–2),113–121 (2001).
    • 87  Smola M, Vandamme T, Sokolowski A. Nanocarriers as pulmonary drug delivery systems to treat and to diagnose respiratory and non respiratory diseases. Int. J. Nanomedicine3(1),1–19 (2008).
    • 88  Nicolazzo JA, Reed BL, Finnin BC. Buccal penetration enhancers – how do they really work? J. Control. Release105(1–2),1–15 (2005).
    • 89  Johansson F, Hjertberg E, Eirefelt S, Tronde A, Hultkvist Bengtsson U. Mechanisms for absorption enhancement of inhaled insulin by sodium taurocholate. Eur. J. Pharm. Sci.17(1–2),63–71 (2002).
    • 90  Heinemann L, Klappoth W, Rave K et al. Intra-individual variability of the metabolic effect of inhaled insulin together with an absorption enhancer. Diabetes Care23(9),1343–1347 (2000).
    • 91  Morimoto K, Uehara Y, Iwanaga K, Kakemi M. Effects of sodium glycocholate and protease inhibitors on permeability of TRH and insulin across rabbit trachea. Pharm. Acta Helv.74(4),411–415 (2000).
    • 92  Yamamoto A, Okumura S, Fukuda Y et al. Improvement of the pulmonary absorption of (Asu1,7)-eel calcitonin by various absorption enhancers and their pulmonary toxicity in rats. J. Pharm. Sci.86(10),1144–1147 (1997).
    • 93  Mesiha MS, Ponnapula S, Plakogiannis F. Oral absorption of insulin encapsulated in artificial chyles of bile salts, palmitic acid and [α]-tocopherol dispersions. Int. J. Pharm.249(1–2),1–5 (2002).
    • 94  Komada F, Iwakawa S, Yamamoto N, Sakakibara H, Okumura K. Intratracheal delivery of peptide and protein agents: absorption from solution and dry powder by rat lung. J. Pharm. Sci.83(6),863–867 (1994).
    • 95  Erjavec Z, Woolthuis GMH, Vries-Hospers HG et al. Tolerance and efficacy of amphotericin B inhalations for prevention of invasive pulmonary aspergillosis in haematological patients. Eur. J. Clin. Microbiol. Infect. Dis.16(5),364–368 (1997).
    • 96  Drew RH, Dodds Ashley E, Benjamin DK Jr et al. Comparative safety of amphotericin B lipid complex and amphotericin B deoxycholate as aerosolized antifungal prophylaxis in lung-transplant recipients. Transplantation77(2),232–237 (2004).
    • 97  Wollmer P, Bäckström K, Zhao H, Nilsson PG, Jonson B. Surface active agents as enhancers of alveolar absorption. Pharm. Res.17(1),38–41 (2000).
    • 98  Freeman DJ, Niven RW. The influence of sodium glycocholate and other additives on the in vivo transfection of plasmid DNA in the lungs. Pharm. Res.13(2),202–209 (1996).
    • 99  Li HY, Seville PC, Williamson IJ, Birchall JC. The use of absorption enhancers to enhance the dispersibility of spray-dried powders for pulmonary gene therapy. J. Gene Med.7(8),1035–1043 (2005).▪ Investigation of the influence of absorption enhancers in the dispersion properties of spray-dried powders and their biological activity in vitro.
    • 100  Ungaro F, De Rosa G, Miro A, Quaglia F, La Rotonda MI. Cyclodextrins in the production of large porous particles: development of dry powders for the sustained release of insulin to the lungs. Eur. J. Pharm. Sci.28(5),423–432 (2006).
    • 101  Niven RW, Byron PR. Solute absorption from the airways of the isolated rat lung. II. Effect of surfactants on absorption of fluorescein. Pharm. Res.7(1),8–13 (1990).
    • 102  Takatsuka S, Morita T, Horikiri Y, Yamahara H, Saji H. Absorption enhancement of poorly absorbed hydrophilic compounds from various mucosal sites by combination of mucolytic agent and non-ionic surfactant. Int. J. Pharm.338(1–2),87–93 (2007).
    • 103  Hosoya K, Kubo H, Natsume H et al. The structural barrier of absorptive mucosae: site difference of the permeability of fluorescein isothiocyanate-labelled dextran in rabbits. Biopharm. Drug Dispos.14(8),685–695 (1993).
    • 104  Yamamoto A, Iseki T, Ochi-Sugiyama M et al. Absorption of water-soluble compounds with different molecular weights and [Asu1.7]-eel calcitonin from various mucosal administration sites. J. Control. Release76(3),363–374 (2001).
    • 105  Machida M, Hayashi M, Awazu S. The effects of absorption enhancers on the pulmonary absorption of recombinant human granulocyte colony-stimulating factor (rhG-CSF) in rats. Biol. Pharm. Bull.23(1),84–86 (2000).
    • 106  Okumura K, Iwakawa S, Yoshida T, Seki T, Komada F. Intratracheal delivery of insulin absorption from solution and aerosol by rat lung. Int. J. Pharm.88(1–3),63–73 (1992).
    • 107  McConville JT, Overhoff KA, Sinswat P et al. Targeted high lung concentrations of itraconazole using nebulized dispersions in a murine model. Pharm. Res.23(5),901–911 (2006).
    • 108  Vaughn JM, Wiederhold NP, McConville JT et al. Murine airway histology and intracellular uptake of inhaled amorphous itraconazole. Int. J. Pharm.338(1–2),219–224 (2007).
    • 109  Fiegel J, Fu J, Hanes J. Poly(ether-anhydride) dry powder aerosols for sustained drug delivery in the lungs. J. Control. Release96(3),411–423 (2004).
    • 110  Fu J, Fiegel J, Krauland E, Hanes J. New polymeric carriers for controlled drug delivery following inhalation or injection. Biomaterials23(22),4425–4433 (2002).
    • 111  Rawat A, Majumder Q, Ahsan F. Inhalable large porous microspheres of low molecular weight heparin: in vitro and in vivo evaluation. J. Control. Release128(3),224–232 (2008).
    • 112  Steckel H, Brandes HG. A novel spray-drying technique to produce low density particles for pulmonary delivery. Int. J. Pharm.278(1),187–195 (2004).
    • 113  Wang L, Zhang Y, Tang X. Characterization of a new inhalable thymopentin formulation. Int. J. Pharm.375(1–2),1–7 (2009).
    • 114  Adler M, Unger M, Lee G. Surface composition of spray-dried particles of bovine serum albumin/trehalose/surfactant. Pharm. Res.17(7),863–870 (2000).
    • 115  Johnson KA. Preparation of peptide and protein powders for inhalation. Adv. Drug Deliv. Rev.26(1),3–15 (1997).
    • 116  Chemistry, Manufacturing and Controls Documentation. FDA Guidance for Industry: metered dose inhaler (MDI) and dry powder inhaler (DPI) drug products. US Dept of Health and Human Sciences, FDA Center for Drug Evaluation and Research (CDER). (1998).
    • 117  Whitehead K, Karr N, Mitragotri S. Discovery of synergistic permeation enhancers for oral drug delivery. J. Control. Release128(2),128–133 (2008).▪ Investigation of various combinations of permeation enhancers and discovery of synergism in several cases.