We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Biocomposite-based nanostructured delivery systems for the treatment and control of inflammatory lung diseases

    Dhrubojyoti Mukherjee

    Department of Pharmaceutics, Faculty of Pharmacy, Ramaiah University of Applied Sciences, Bengaluru, Karnataka, 560054, India

    &
    Shvetank Bhatt

    *Author for correspondence:

    E-mail Address: shvetankbhatt@gmail.com

    Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, Madhya Pradesh, 474005, India

    Published Online:https://doi.org/10.2217/nnm-2021-0425

    Diseases related to the lungs are among the most prevalent medical problems threatening human life. The treatment options and therapeutics available for these diseases are hindered by inadequate drug concentrations at pathological sites, a dearth of cell-specific targeting and different biological barriers in the alveoli or conducting airways. Nanostructured delivery systems for lung drug delivery have been significant in addressing these issues. The strategies used include surface engineering by altering the material structure or incorporation of specific ligands to reach prespecified targets. The unique characteristics of nanoparticles, such as controlled size and distribution, surface functional groups and therapeutic release triggering capabilities, are tailored to specific requirements to overcome the major therapeutic barriers in pulmonary diseases. In the present review, the authors intend to deliver significant up-to-date research in nanostructured therapies in inflammatory lung diseases with an emphasis on biocomposite-based nanoparticles.

    Plain language summary

    Lung-related disorders such as asthma, chronic obstructive pulmonary diseases and pulmonary fibrosis are the result of inflammatory processes in the human body. The causes of these lung diseases can vary from unknown to specific. Chronic obstructive pulmonary disease is most commonly caused by smoking, whereas asthma may be caused by allergens, infections, cold air or smoke. Targeting these lung-related diseases with biologically degradable polymeric nanoparticles has recently been proposed as an effective treatment. These nanoparticles can be made by combining different materials to form biocomposite nanoparticles. Different drugs can be loaded into nanoparticles, and the surface of nanoparticles can be modified to change their properties; for example, to make them target diseased parts of the lung. In this article, the authors discuss current trends in nanoparticle treatment of inflammatory lung diseases, including the significance of biologically degradable materials.

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

    References

    • 1. Chen L, Deng H, Cui H et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 9(6), 7204–7218 (2018).
    • 2. Subbarao P, Mandhane PJ, Sears MR. Asthma: epidemiology, etiology and risk factors. CMAJ 181(9), e181–e190 (2009).
    • 3. Hall S, Agrawal DK. Key mediators in the immunopathogenesis of allergic asthma. Int. Immunopharmacol. 23(1), 316–329 (2014).
    • 4. Kudo M, Ishigatsubo Y, Aoki I. Pathology of asthma. Front. Microbiol. 4(263), 1–16 (2013).
    • 5. Rawat D, Sharma S. Case study: 60-year-old female presenting with shortness of breath. In: StatPearls. StatPearls Publishing LLC, FL, USA (2021).
    • 6. Hikichi M, Mizumura K, Maruoka S, Gon Y. Pathogenesis of chronic obstructive pulmonary disease (COPD) induced by cigarette smoke. J. Thorac. Dis. 11(Suppl. 17), S2129–S2140 (2019).
    • 7. Brode SK, Ling SC, Chapman KR. Alpha-1 antitrypsin deficiency: a commonly overlooked cause of lung disease. CMAJ 184(12), 1365–1371 (2012).
    • 8. Buist AS. Similarities and differences between asthma and chronic obstructive pulmonary disease: treatment and early outcomes. Eur. Respir. J. Suppl. 39, 30s–35s (2003).
    • 9. Wilson MS, Wynn TA. Pulmonary fibrosis: pathogenesis, etiology and regulation. Mucosal Immunol. 2(2), 103–121 (2009).
    • 10. Qiao Q et al. Nanomedicine for acute respiratory distress syndrome: the latest application, targeting strategy, and rational design. Acta Pharm. Sin. B 11(10), 3060–3091 (2021).
    • 11. Han Y et al. Ag NPs on chitosan–alginate-coated magnetite for synthesis of indazolo[2,1-b]phthalazines and human lung protective effects against α-guttiferin. Int. J. Biol. Macromol. 164, 2974–2986 (2020).
    • 12. Dharmage SC, Perret JL, Custovic A. Epidemiology of asthma in children and adults. Front. Pediatr. 7, 246 (2019).
    • 13. Matucci A et al. Is IgE or eosinophils the key player in allergic asthma pathogenesis? Are we asking the right question? Respir. Res. 19(1), 113 (2018).
    • 14. Stone KD, Prussin C, Metcalfe DD. IgE, mast cells, basophils, and eosinophils. J. Allergy Clin. Immunol. 125(2 Suppl. 2), S73–S80 (2010).
    • 15. Krystel-Whittemore M, Dileepan KN, Wood JG. Mast cell: a multi-functional master cell. Front Immunol. 6(620), 1–12 (2016).
    • 16. McBrien CN, Menzies-Gow A. The biology of eosinophils and their role in asthma. Front. Med. (Lausanne) 4, 93 (2017).
    • 17. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 15(3), 178–196 (2014).
    • 18. Venge P. The eosinophil and airway remodelling in asthma. Clin. Respir. J. 4(Suppl. 1), 15–19 (2010).
    • 19. Barnes PJ et al. Chronic obstructive pulmonary disease. Nat. Rev. Dis. Primers 1, 15076 (2015).
    • 20. Tetley TD. Inflammatory cells and chronic obstructive pulmonary disease. Curr. Drug Targets Inflamm. Allergy 4(6), 607–618 (2005).
    • 21. Bajpai J et al. Study of oxidative stress biomarkers in chronic obstructive pulmonary disease and their correlation with disease severity in north Indian population cohort. Lung India 34(4), 324–329 (2017).
    • 22. Pandey KC, De S, Mishra PK. Role of proteases in chronic obstructive pulmonary disease. Front. Pharmacol. 8, 512 (2017).
    • 23. Cukic V et al. Asthma and chronic obstructive pulmonary disease (COPD) – differences and similarities. Mater. Sociomed. 24(2), 100–105 (2012).
    • 24. Bhatt S, Kanoujia J, Nagappa AN. Targeting molecular and cellular mechanisms in chronic obstructive pulmonary disease. In: Targeting Cellular Signalling Pathways in Lung Diseases. Dua KNagappa ANMalheiros Luzo ÂC (Eds). Springer Singapore, Singapore, Republic of Singapore, 127–146 (2021).
    • 25. Navaratnam V et al. The rising incidence of idiopathic pulmonary fibrosis in the U.K. Thorax 66(6), 462–467 (2011).
    • 26. Wuyts WA et al. The pathogenesis of pulmonary fibrosis: a moving target. Eur. Respir. J. 41(5), 1207–1218 (2013).
    • 27. Song Q, Chen P, Liu XM. The role of cigarette smoke-induced pulmonary vascular endothelial cell apoptosis in COPD. Respir. Res. 22(1), 39 (2021). • Describes the effect of smoking on the deeper part of the lungs.
    • 28. Laniado-Laborín R. Smoking and chronic obstructive pulmonary disease (COPD). Parallel epidemics of the 21 century. Int. J. Environ. Res. Public Health 6(1), 209–224 (2009).
    • 29. Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv. Drug Deliv. Rev. 56(11), 1649–1659 (2004).
    • 30. Nguyen HX. Targeted delivery of surface-modified nanoparticles: modulation of inflammation for acute lung injury. In: Surface Modification of Nanoparticles for Targeted Drug Delivery. Pathak YV (Ed.). Springer International Publishing, Cham, Switzerland, 331–353 (2019).
    • 31. Saini R, Bajpai AK, Jain E. Advances in bionanocomposites for biomedical applications. Biodegradable and Biocompatible Polymer Composites. 1–21 (2017).
    • 32. Patra JK et al. Nano based drug delivery systems: recent developments and future prospects. J. Nanobiotechnology 16(1), 71 (2018).
    • 33. Bonner JC. Nanoparticles as a potential cause of pleural and interstitial lung disease. Proc. Am. Thorac. Soc. 7(2), 138–141 (2010). •• Discusses the channelling of nanoparticles into the deeper part of the lungs, which could explain the importance and strategic significance of nanoparticle targeting in the lungs.
    • 34. Suk JS et al. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 99(Pt A), 28–51 (2016).
    • 35. Mikušová V, Mikuš P. Advances in chitosan-based nanoparticles for drug delivery. Int. J. Mol. Sci. 22(17), 9652 (2021).
    • 36. Labiris NR, Dolovich MB. Pulmonary drug delivery. Part I: physiological factors affecting therapeutic effectiveness of aerosolized medications. Br. J. Clin. Pharmacol. 56(6), 588–599 (2003).
    • 37. Mangum JB et al. Single-walled carbon nanotube (SWCNT)-induced interstitial fibrosis in the lungs of rats is associated with increased levels of PDGF mRNA and the formation of unique intercellular carbon structures that bridge alveolar macrophages in situ. Part. Fibre Toxicol. 3, 15 (2006).
    • 38. Pontes JF, Grenha A. Multifunctional nanocarriers for lung drug delivery. Nanomaterials (Basel) 10(2), 1–24 (2020).
    • 39. Luo MX, Hua S, Shang QY. Application of nanotechnology in drug delivery systems for respiratory diseases (review). Mol. Med. Rep. 23(5), 325 (2021).
    • 40. Qiao H et al. The transport and deposition of nanoparticles in respiratory system by inhalation. J. Nanomaterials 2015, 394507 (2015). •• Details the effect of nanomaterial particle size on the pulmonary system.
    • 41. Bharadwaz A, Jayasuriya AC. Recent trends in the application of widely used natural and synthetic polymer nanocomposites in bone tissue regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 110, 110698 (2020).
    • 42. Kaur G et al. Advances in pulmonary delivery of nanoparticles. Artif. Cells Blood Substit. Immobil. Biotechnol. 40(1–2), 75–96 (2012).
    • 43. Samani DA et al. Electrospun polycaprolactone/graphene/baghdadite composite nanofibres with improved mechanical and biological properties. Fibers Polym. 20(5), 982–990 (2019).
    • 44. Bagheri M, Mahmoodzadeh A. Polycaprolactone/graphene nanocomposites: synthesis, characterization and mechanical properties of electrospun nanofibers. J. Inorg. Organomet. Polym. Mater. 30(5), 1566–1577 (2020).
    • 45. Ely L et al. Effervescent dry powder for respiratory drug delivery. Eur. J. Pharm. Biopharm. 65(3), 346–353 (2007).
    • 46. Al-Hallak KMHD et al. Secondary cytotoxicity mediated by alveolar macrophages: a contribution to the total efficacy of nanoparticles in lung cancer therapy. Eur. J. Pharm. Biopharm. 76(1), 112–119 (2010).
    • 47. Loch-Neckel G et al. Orally administered chitosan-coated polycaprolactone nanoparticles containing curcumin attenuate metastatic melanoma in the lungs. J. Pharm. Sci. 104(10), 3524–3534 (2015).
    • 48. Ilyas RA et al. Natural fiber-reinforced polycaprolactone green and hybrid biocomposites for various advanced applications. Polymers 14(1), 182 (2022).
    • 49. Tahriri M et al. Biodegradation properties of PLGA/nano-fluorhydroxyapatite composite microsphere-sintered scaffolds. Dent. Mater. 32, e49–e50 (2016).
    • 50. Masaeli R et al. Efficacy of the biomaterials 3wt%-nanostrontium-hydroxyapatite-enhanced calcium phosphate cement (nanoSr-CPC) and nanoSr-CPC-incorporated simvastatin-loaded poly(lactic-co-glycolic-acid) microspheres in osteogenesis improvement: an explorative multi-phase experimental in vitro/vivo study. Mater. Sci. Eng. C Mater. Biol. Appl. 69, 171–183 (2016).
    • 51. Wang Y et al. Poly(lactide-co-glycolide)/titania composite microsphere-sintered scaffolds for bone tissue engineering applications. J. Biomed. Mater. Res. B Appl. Biomater. 93(1), 84–92 (2010).
    • 52. Eslami H, Solati-Hashjin M, Tahriri M. The comparison of powder characteristics and physicochemical, mechanical and biological properties between nanostructure ceramics of hydroxyapatite and fluoridated hydroxyapatite. Mater. Sci. Eng. C Mater. Biol. Appl. 29(4), 1387–1398 (2009).
    • 53. Lv Q, Nair L, Laurencin CT. Fabrication, characterization, and in vitro evaluation of poly(lactic acid glycolic acid)/nano-hydroxyapatite composite microsphere-based scaffolds for bone tissue engineering in rotating bioreactors. J. Biomed Mater Res A. 91A(3), 679–691 (2009).
    • 54. Emami F, Mostafavi Yazdi SJ, Na DH. Poly(lactic acid)/poly(lactic-co-glycolic acid) particulate carriers for pulmonary drug delivery. J. Pharm. Investig. 49(4), 427–442 (2019).
    • 55. Nishimura S, Takami T, Murakami Y. Porous PLGA microparticles formed by ‘one-step’ emulsification for pulmonary drug delivery: the surface morphology and the aerodynamic properties. Colloids Surf. B Biointerfaces 159, 318–326 (2017).
    • 56. Rogueda PG, Traini D. The nanoscale in pulmonary delivery. Part 1: deposition, fate, toxicology and effects. Expert Opin. Drug Deliv. 4(6), 595–606 (2007).
    • 57. Hillaireau H, Couvreur P. Nanocarriers' entry into the cell: relevance to drug delivery. Cell. Mol. Life Sci. 66(17), 2873–2896 (2009).
    • 58. Ungaro F et al. Engineered PLGA nano- and micro-carriers for pulmonary delivery: challenges and promises. J. Pharm. Pharmacol. 64(9), 1217–1235 (2012).
    • 59. Harush-Frenkel O et al. A safety and tolerability study of differently charged nanoparticles for local pulmonary drug delivery. Toxicol. Appl. Pharmacol. 246(1–2), 83–90 (2010).
    • 60. Zhao D et al. Poly(lactic-co-glycolic acid)-based composite bone-substitute materials. Bioact. Mater. 6(2), 346–360 (2021).
    • 61. Hak S et al. The effect of nanoparticle polyethylene glycol surface density on ligand-directed tumor targeting studied in vivo by dual modality imaging. ACS Nano 6(6), 5648–5658 (2012).
    • 62. Kamnoore D et al. Hydroxyapatite nanoparticle-enriched thiolated polymer-based biocompatible scaffold can improve skin tissue regeneration. J. Mater. Res. 36(21), 4287–4306 (2021). • Discusses the importance of tissue regeneration in understanding the effect of bio-nanocomposites on pulmonary targeting and healing.
    • 63. Zhang CY et al. pH-responsive nanoparticles targeted to lungs for improved therapy of acute lung inflammation/injury. ACS Appl. Mater. Interfaces 11(18), 16380–16390 (2019).
    • 64. Xu G et al. Transferrin and tocopheryl-polyethylene glycol-succinate dual ligands decorated, cisplatin loaded nano-sized system for the treatment of lung cancer. Biomed. Pharmacother. 99, 354–362 (2018).
    • 65. Sikka MP, Midha VK. The role of biopolymers and biodegradable polymeric dressings in managing chronic wounds. In: Advanced Textiles for Wound Care (2nd Edition). Rajendran S (Ed.). Woodhead Publishing, 463–488 (2019).
    • 66. Lopes MS, Jardini AL, Filho RM. Poly (lactic acid) production for tissue engineering applications. Procedia Eng. 42, 1402–1413 (2012).
    • 67. Scaffaro R et al. Preparation of three-layered porous PLA/PEG scaffold: relationship between morphology, mechanical behavior and cell permeability. J. Mech. Behav. Biomed. Mater. 54, 8–20 (2016).
    • 68. Salerno A et al. Bio-safe fabrication of PLA scaffolds for bone tissue engineering by combining phase separation, porogen leaching and scCO2 drying. J. Supercrit. Fluids 97, 238–246 (2015).
    • 69. Das SC et al. Nanomedicine in pulmonary delivery. In: Theory and Applications of Nonparenteral Nanomedicines. Kesharwani PTaurin SGreish K (Eds). Academic Press, 319–354 (2021).
    • 70. Buhecha MD et al. Development and characterization of PLA nanoparticles for pulmonary drug delivery: co-encapsulation of theophylline and budesonide, a hydrophilic and lipophilic drug. J. Drug Deliv. Sci. Technol. 53, 101128 (2019).
    • 71. Ganguly P, Breen A, Pillai SC. Toxicity of nanomaterials: exposure, pathways, assessment, and recent advances. ACS Biomater. Sci. Eng. 4(7), 2237–2275 (2018).
    • 72. Shahrousvand M, Sadeghi GMM, Salimi A. Artificial extracellular matrix for biomedical applications: biocompatible and biodegradable poly (tetramethylene ether) glycol/poly (ε-caprolactone diol)-based polyurethanes. J. Biomater. Sci. Polym. Ed. 27(17), 1712–1728 (2016).
    • 73. Paul DR, Robeson LM. Polymer nanotechnology: nanocomposites. Polymer 49(15), 3187–3204 (2008).
    • 74. Iyer R et al. Glutathione-responsive biodegradable polyurethane nanoparticles for lung cancer treatment. J. Control. Release 321, 363–371 (2020).
    • 75. Gabriel LP et al. Bio-based polyurethane for tissue engineering applications: how hydroxyapatite nanoparticles influence the structure, thermal and biological behavior of polyurethane composites. Nanomedicine 13(1), 201–208 (2017).
    • 76. da Silva GR et al. Biodegradation of polyurethanes and nanocomposites to non-cytotoxic degradation products. Polym. Degrad. Stab. 95(4), 491–499 (2010).
    • 77. Bhat SS et al. Thiolated polymer nanocarrier reinforced with glycyrrhetinic acid for targeted delivery of 5-fluorouracil in hepatocellular carcinoma. Drug Deliv. Transl. Res. 11(5), 2252–2269 (2021).
    • 78. Mukherjee D et al. Improvement of bone microarchitecture in methylprednisolone induced rat model of osteoporosis by using thiolated chitosan-based risedronate mucoadhesive film. Drug Dev. Ind. Pharm. 44(11), 1845–1856 (2018).
    • 79. Yi H et al. Biofabrication with chitosan. Biomacromolecules 6(6), 2881–2894 (2005).
    • 80. Mostafa M et al. Insight into chitosan/zeolite-A nanocomposite as an advanced carrier for levofloxacin and its anti-inflammatory properties; loading, release, and anti-inflammatory studies. Int. J. Biol. Macromol. 179, 206–216 (2021).
    • 81. Jatal R et al. Lung targeted electrosprayed chitosan nanocomposite microparticles boost the cytotoxic activity of magnolol. Carbohydr. Polym. Technol. Appl. 2, 100169 (2021).
    • 82. Jin H et al. Nasal delivery of hesperidin/chitosan nanoparticles suppresses cytokine storm syndrome in a mouse model of acute lung injury. 11(1786), 592238 (2021).
    • 83. Rizeq BR et al. Synthesis, bioapplications, and toxicity evaluation of chitosan-based nanoparticles. Int. J. Mol. Sci. 20(22), (2019).
    • 84. Campbell JJ et al. Development of three-dimensional collagen scaffolds with controlled architecture for cell migration studies using breast cancer cell lines. Biomaterials 114, 34–43 (2017).
    • 85. Sarker B et al. Collagen/silica nanocomposites and hybrids for bone tissue engineering. Nanotechnol. Rev. 2(4), 427–447 (2013).
    • 86. Li S et al. Preparation and biocompatibility studies of collagen/hyaluronic acid/oligomeric proanthocyanidins composites. Mater. Chem. Phys. 272, 124959 (2021).
    • 87. Azarpira N, Kaviani M, Sarvestani FS. Incorporation of VEGF-and bFGF-loaded alginate oxide particles in acellular collagen-alginate composite hydrogel to promote angiogenesis. Tissue Cell 72, 101539 (2021).
    • 88. Wolinsky JB et al. Prevention of in vivo lung tumor growth by prolonged local delivery of hydroxycamptothecin using poly(ester-carbonate)–collagen composites. J. Control. Release 144(3), 280–287 (2010).
    • 89. Shalaby M et al. Collagen polymer and magnetic collagen nanocomposite recycled from waste to reduce polluted water toxicity. Polym. Polym. Compos. 29(9), 1515–1527 (2020).
    • 90. Johari N, Hosseini HRM, Samadikuchaksaraei A. Novel fluoridated silk fibroin/TiO2 nanocomposite scaffolds for bone tissue engineering. Mater. Sci. Eng. C Mater. Biol. Appl. 82, 265–276 (2018).
    • 91. Kapoor S, Kundu SC. Silk protein-based hydrogels: promising advanced materials for biomedical applications. Acta Biomater. 31, 17–32 (2016).
    • 92. Datta A, Ghosh AK, Kundu SC. Purification and characterization of fibroin from the tropical saturniid silkworm, Antheraea mylitta. Insect Biochem. Mol. Biol. 31(10), 1013–1018 (2001).
    • 93. Patra C et al. Silk protein fibroin from Antheraea mylitta for cardiac tissue engineering. Biomaterials 33(9), 2673–2680 (2012).
    • 94. Zuluaga-Vélez A et al. Silk fibroin nanocomposites as tissue engineering scaffolds – a systematic review. Biomed. Pharmacother. 141, 111924 (2021).
    • 95. Ma X et al. Chitosan/polydopamine layer by layer self-assembled silk fibroin nanofibers for biomedical applications. Carbohydr. Polym. 251, 117058 (2021).
    • 96. Passi M, Kumar V, Packirisamy G. Theranostic nanozyme: silk fibroin based multifunctional nanocomposites to combat oxidative stress. Mater. Sci. Eng. C Mater. Biol. Appl. 107, 110255 (2020).
    • 97. Beladi F, Saber-Samandari S, Saber-Samandari S. Cellular compatibility of nanocomposite scaffolds based on hydroxyapatite entrapped in cellulose network for bone repair. Mater. Sci. Eng. C Mater. Biol. Appl. 75, 385–392 (2017).
    • 98. Saber-Samandari S et al. Synthesis, characterization and application of cellulose-based nano-biocomposite hydrogels. J. Macromol. Sci. A 50(11), 1133–1141 (2013).
    • 99. Kalia S et al. Cellulose-based bio- and nanocomposites: a review. Int. J. Polym. Sci. 2011, 837875 (2011).
    • 100. Patil TV et al. Nanocellulose, a versatile platform: from the delivery of active molecules to tissue engineering applications. Bioact. Mater. 9, 566–589 (2022).
    • 101. Hamouda RA et al. Characterization and anticancer activity of biosynthesized Au/cellulose nanocomposite from Chlorella vulgaris. Polymers (Basel) 13(19), 3340 (2021).
    • 102. Joseph B et al. Cellulose nanocomposites: fabrication and biomedical applications. J. Bioresour. Bioprod. 5(4), 223–237 (2020).
    • 103. He L et al. Alginate-based platforms for cancer-targeted drug delivery. Biomed Res. Int. 2020, 1487259 (2020). • Demonstrates the efficacy of alginates for drug delivery of anticancer drugs in lung cancer.
    • 104. Abasalizadeh F et al. Alginate-based hydrogels as drug delivery vehicles in cancer treatment and their applications in wound dressing and 3D bioprinting. J. Biol. Eng. 14(1), 8 (2020).
    • 105. Hill M et al. Alginate/chitosan particle-based drug delivery systems for pulmonary applications. Pharmaceutics 11(8), 379 (2019).
    • 106. Kloster GA et al. Alginate-based nanocomposites with magnetic properties. Compos. A Appl. Sci. Manuf. 135, 105936 (2020).
    • 107. Gaber M et al. Protein–lipid nanohybrids as emerging platforms for drug and gene delivery: challenges and outcomes. J. Control. Release 254, 75–91 (2017).
    • 108. Elhissi A. Liposomes for pulmonary drug delivery: the role of formulation and inhalation device design. Curr. Pharm. Des. 23(3), 362–372 (2017).
    • 109. Abdelaziz HM et al. Inhalable particulate drug delivery systems for lung cancer therapy: nanoparticles, microparticles, nanocomposites and nanoaggregates. J. Control. Release 269, 374–392 (2018).
    • 110. Rudokas M et al. Liposome delivery systems for inhalation: a critical review highlighting formulation issues and anticancer applications. Med. Princ. Pract. 25(Suppl. 2), 60–72 (2016).
    • 111. Romøren K et al. Transfection efficiency and cytotoxicity of cationic liposomes in salmonid cell lines of hepatocyte and macrophage origin. Biochim. Biophys. Acta 1663(1–2), 127–134 (2004).
    • 112. Inglut CT et al. Immunological and toxicological considerations for the design of liposomes. Nanomaterials (Basel) 10(2), 190 (2020).
    • 113. Bochicchio S, Lamberti G, Barba AA. Polymer–lipid pharmaceutical nanocarriers: innovations by new formulations and production technologies. Pharmaceutics 13(2), 198 (2021).
    • 114. Polack FP, SJ Thomas, N Kitchin et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N. Engl. J. Med. 383(27), 2603–2615 (2020).
    • 115. Walsh EE et al. Safety and immunogenicity of two RNA-based COVID-19 vaccine candidates. N. Engl. J. Med. 383(25), 2439–2450 (2020).
    • 116. Persano F, Gigli G, Leporatti S. Lipid–polymer hybrid nanoparticles in cancer therapy: current overview and future directions. Nano Express 2(1), 012006 (2021).
    • 117. Roy I, Vij N. Nanodelivery in airway diseases: challenges and therapeutic applications. Nanomedicine 6(2), 237–244 (2010).
    • 118. Patil JS, Sarasija S. Pulmonary drug delivery strategies: a concise, systematic review. Lung India 29(1), 44–49 (2012).
    • 119. Yıldız-Peköz A, Ehrhardt C. Advances in pulmonary drug delivery. Pharmaceutics 12(10), 911 (2020).
    • 120. Yao H, de Boer WI, Rahman I. Targeting lung inflammation: novel therapies for the treatment of COPD. Curr. Respir. Med. Rev. 4(1), 57–68 (2008).
    • 121. Han S et al. Efficient delivery of antitumor drug to the nuclei of tumor cells by amphiphilic biodegradable poly(L-aspartic acid-co-lactic acid)/DPPE co-polymer nanoparticles. Small 8(10), 1596–1606 (2012).
    • 122. Kenyon NJ et al. Self-assembling nanoparticles containing dexamethasone as a novel therapy in allergic airways inflammation. PLOS ONE 8(10), e77730 (2013).
    • 123. Kumar M et al. Chitosan IFN-gamma-pDNA nanoparticle (CIN) therapy for allergic asthma. Genet. Vaccines Ther. 1(1), 3 (2003).
    • 124. Pandey R et al. Poly (DL-lactide-co-glycolide) nanoparticle-based inhalable sustained drug delivery system for experimental tuberculosis. J. Antimicrob. Chemother. 52(6), 981–986 (2003).
    • 125. Roa WH et al. Inhalable nanoparticles, a non-invasive approach to treat lung cancer in a mouse model. J. Control. Release 150(1), 49–55 (2011).
    • 126. Kamat CD et al. Poly(β-amino ester) nanoparticle delivery of TP53 has activity against small cell lung cancer in vitro and in vivo. Mol. Cancer Ther. 12(4), 405–415 (2013).
    • 127. Chen Y et al. Nanoparticles modified with tumor-targeting scFv deliver siRNA and miRNA for cancer therapy. Mol. Ther. 18(9), 1650–1656 (2010).