Saturday, June 26, 2021

Design Considerations for a Successful Nanoparticle Delivery System

 Nanoparticles (NPs) are thought to have potential as novel intravascular probes for both diagnostic (e.g., imaging) and therapeutic purposes (e.g., drug delivery)

Critical issues for successful nanoparticle delivery include the ability:

  • To target specific tissues and cell types 
    • Endocytosis (uptake into the cells)
    • Targeting agent (address tags)
  • To escape from the biological particulate filter 
    • Mononuclear Phagocyte System (MPS; clearance of unwanted particulate material)
    • Protection from Clearance by MPS 
      • Main NP research question—how is particulate material recognized and cleared?
In this article, we will cover the above five highlighted areas.  Note that the below topics are parts of the design considerations, but will not discussed here.
  • Reduction in toxicity while maintaining therapeutic effects
  • Greater safety and biocompatibility


Figure 1.  Endocytosis (Created by Mariana Ruiz Villarreal LadyofHats)

Endocytosis—Uptake into the Cells


Nanoparticles enter the cells by endocytosis. Endocytosis is a process of the uptake substances into the cell.  It is a form of active transport which includes:[92]
  • Pinocytosis (cell drinking) 
    • The uptake of small (<0.5 μm) particles into vesicles
  • Phagocytosis (cell eating)
    • The uptake of larger particles (>0.5 μm) by a process involving actin polymerization. 
Both can be receptor mediated. 

Figure 2.  Schematic representation of qdot targeting. (source: [95])
Intravenous delivery of qdots into specific tissues of the mouse. (Upper)
Design of peptide-coated qdots. (Lower)
Qdots were coated with either peptides only or with peptides and PEG.
PEG helps the qdots maintain solubility in aqueous solvents and minimize nonspecific binding.

Targeting Agent (Address Tags)


Biodegradable nanoparticles have been studied extensively for the fabrication of drug delivery systems.  The design considerations include controlling the release of drugs, stabilizing labile molecules (e.g., proteins, peptides, or DNA) from degradation, and site-specific drug targeting.

Nanoparticles (NPs) can be linked to biological molecules that can act as address tags.  Common address tags are:

Address tags can direct NPs to:

  • Specific sites within the body[90]
  • Specific organelles within the cell[95]
  • Follow specifically the movement of individual protein or RNA molecules in living cells[96]

For example, scientists were able to show that (See Figure 1):[91]

ZnS-capped CdSe qdots coated with a lung-targeting peptide accumulate in the lungs of mice after i.v. injection, whereas two other peptides specifically direct qdots to blood vessels or lymphatic vessels in tumors. 
Adding polyethylene glycol (PEG) to the qdot coating prevents nonselective accumulation of qdots in reticuloendothelial tissues.


These targeting agents should ideally be covalently linked to the nanoparticle and should be present in a controlled number per nanoparticle:

  • Multivalent nanoparticles
    • Bearing multiple targeting groups, can cluster receptors, which can activate cellular signaling pathways, and give stronger anchoring
  • Monovalent nanoparticles
    • Bearing a single binding site,[97-99] avoid clustering and so are preferable for tracking the behavior of individual proteins.

Figure 3.  Nanoparticle Uptake: The Phagocyte Problem (Source: [111])

Mononuclear Phagocyte System


Initially, Reticuloendothelial System (RES) denotes a system of specialized cells that effectively clear colloidal vital stains (so called because they stain living cells) from the blood circulation.  
The capture and clearance of unwanted particulate material from blood and lymph were considered to be the major function of the RES. 
As knowledge accumulated, the term RES was regarded as insufficient to describe resident phagocytes and their antecedents.  In 1969, a group of prominent pathologists/immunologists proposed the term Mononuclear Phagocyte System (MPS) as a more accurate term.[100]

The MPS is part of both humoral and cell-mediated immunity.  The trinity of MPS include:
which are distinguished on the basis of their morphology, function, and origin, yet collectively constitute the MPS.

Phagocytes and Nanoparticles


Issues plaguing nanomaterials circulation and targeting in humans can be attributed to:[104-110]
  • Rapid vascular filtration and clearance of therapeutics and diagnostics
  • Induction of host inflammatory responses due to non-specific recognition
  • Uptake of nanoconstructs by macrophages in vivo
Phagocytes are key cellular participants determining important aspects of host exposure to nanomaterials, initiating clearance, biodistribution and the tenuous balance between host tolerance and adverse nanotoxicity.[111] 
Macrophages in particular are believed to be among the first and primary cell types that process nanoparticles, mediating host inflammatory and immunological biological responses. 
These processes occur ubiquitously throughout tissues where nanomaterials are present, including the host mononuclear phagocytic system (MPS) residents in dedicated host filtration organs (i.e., liver, kidney spleen, and lung)

Nanoparticle delivery vehicles designed to either avoid or specifically harness this host recognition system could improve payload delivery, reduce inflammatory effects and improve imaging and drug efficacy. 

Figure 4.  Adsorbed protein conformation can be affected by different particle surface properties.
Surface curvature, topography, hydrophilic/hydrophobic chemistry and
polymer coating steric barriers on nanoparticle surfaces are shown to
alter amounts and conformations of proteins adsorbed to surfaces. (Source: [111])


Protection from Clearance by MPS


The nanoparticle size and surface properties can be manipulated to avoid rapid clearance by phagocytic cells.  
Nanoparticle association with the host highly evolved mononuclear phagocytic system (MPS) is a function of particle opsonization upon contact with blood and rapid recognition of these opsonins via the MPS.[113,115] 
Surface-adsorbed proteins (opsonins) influence macrophage recognition and uptake of nanoparticles.[112-114]  Additionally, conformational protein rearrangements on nanoparticle surfaces alters protein epitope exposures to phagocytes.[116,117]  Certain epitopes have the capacity to activate macrophages.[113] 

Protein adsorption and subsequent phagocytic uptake appear dependent upon (see Figure 4):[111] 
  • Particle surface curvature[118,119]
  • Particle surface topography[120]
  • Particle surface energies (e.g., hydrophilicity/hydrophobicity)
  • Polymer coating steric barriers on nanoparticle surfaces
For example, hydrophilic polyethylene glycol (PEG) has often been immobilized in many forms and approaches to provide a brush-like steric barrier that is shown to reduce protein adsorption and is correlated with increased blood circulation times for some particles.[121] Dextran layers employed on commercial iron oxide MRI agent nanoparticles (i.e., Feridexmay serve the same role.[121] 
Surface modification of NPs with polyethylene glycol (PEG) have improved drug delivery properties and prolonged circulation life time with enhanced protection from clearance by mononuclear phagocytic systems.[101-103]

References

  1. Nanoparticles—a historical perspective
  2. Preparation and Characterization of Cross-Linked Polymeric Nanoparticles for Enhanced Oil Recovery Applications (pdf)
  3. Polyacrylamide and its deriv ylamide and its derivatives for oil r es for oil recovery (pdf)
  4. Nanosizing of drugs: Effect on dissolution rate
  5. Development and functional characterization of alginate dressing as potential protein delivery system for wound healing
  6. Designing hydrogels for controlled drug delivery (good)
  7. Novavax addresses urgent global public health needs with innovative technology
  8. How the Novavax Vaccine Works
  9. Widder KJ, Senyel AE, Scarpelli GD. Magnetic microspheres: a model system of site specific drug delivery in vivo. Proc Soc Exp Biol Med. 1978;158:141–6.
  10. Nuclear molecular imaging with nanoparticles: radiochemistry, applications and translation
  11. Methods for Synthesis of Nanoparticles and Fabrication of Nanocomposites
  12. NANOPARTICULATE DRUG DELIVERY SYSTEM
  13. Switchable Release of Entrapped Nanoparticles from Alginate Hydrogels.[Adv Healthc Mater. 2015]
  14. Nanosized cationic hydrogels for drug delivery: preparation, properties and interactions with cells. Vinogradov SV, Bronich TK, Kabanov AV. Adv Drug Deliv Rev. 2002 Jan 17; 54(1):135-47.
  15. Applications of Targeted Nano Drugs and Delivery Systems
  16. Langer R. Drug delivery and targeting. Nature. 1998;392:5–10.
  17. Hoare TR, Kohane DS. Hydrogels in drug delivery: Progress and challenges. Polymer. 2008;49:1993–2007.
  18. Liechty WB, Kryscio DR, Slaughter BV, Peppas NA. Polymers for drug delivery systems. Ann Rev Chem Biomol Eng. 2010;1:149–173.
  19. Cohen J. IL-12 deaths: explanation and a puzzle. Science. 1995;270:908–908.
  20. Florence AT, Jani PU. Novel oral drug formulations. Drug Safety. 1994;10:233–266.
  21. Ashley GW, Henise J, Reid R, Santi DV. Hydrogel drug delivery system with predictable and tunable drug release and degradation rates. Proc Natl Acad Sci USA. 2013;110:2318–2323.
  22. Tiwari G, et al. Drug delivery systems: An updated review. Int J Pharm Investig. 2012;2:2–11.
  23. Tibbitt MW, Dahlman JE, Langer R. Emerging frontiers in drug delivery. J Am Chem Soc.
  24. Singh, BN; Prateeksha, Gupta VK; Chen, J; Atanasov, AG (2017). "Organic Nanoparticle-Based Combinatory Approaches for Gene Therapy". Trends Biotechnol. 35 (12): 1121–1124. 2016;138:704–717.
  25. Wang, Zhenming; Wang, Zhefeng; Lu, William Weijia; Zhen, Wanxin; Yang, Dazhi; Peng, Songlin (October 2017). "Novel biomaterial strategies for controlled growth factor delivery for biomedical applications"
  26. Mitragotri S, Lahann J. Physical approaches to biomaterial design. Nat Mater. 2009;8:15–23.
  27. Euliss LE, DuPont JA, Gratton S, DeSimone J. Imparting size, shape, and composition control of materials for nanomedicine. Chem Soc Rev. 2006;35:1095–1104.
  28. Gratton SE, et al. The effect of particle design on cellular internalization pathways. Proc Natl Acad Sci USA. 2008;105:11613–11618.
  29. Geng H, Song H, Qi J, Cui D. Sustained release of VEGF from PLGA nanoparticles embedded thermo-sensitive hydrogel in full-thickness porcine bladder acellular matrix. Nanoscale Res Lett. 2011;6:1–8.
  30. Oh JK, Drumright R, Siegwart DJ, Matyjaszewski K. The development of microgels/nanogels for drug delivery applications. Prog Polym Sci. 2008;33:448–477.
  31. Crommelin DJ, Storm G. Liposomes: from the bench to the bed. J Liposome Res. 2003;13:33–6.
  32. Metselaar JM, Storm G. Liposomes in the treatment of inflammatory disorders. Expert Opin Drug Deliv. 2005;2:465–76.
  33. Noh SY, Nash A, Notman R (2020). "The aggregation of striped nanoparticles in mixed phospholipid bilayers". Nanoscale. 12 (8): 4868–81.
  34. Nanotechnologies: 6. What are potential harmful effects of nanoparticles?
  35. Thake, T.H.F; Webb, J.R; Nash, A.; Rappoport, J.Z.; Notman, R. (2013). "Permeation of polystyrene nanoparticles across model lipid bilayer membranes". Soft Matter. 9 (43): 10265 10274.
  36. Greulich, C.; Diendorf, J.; Simon, T.; Eggeler, G.; Epple, M.; Köller, M. (January 2011). "Uptake and intracellular distribution of silver nanoparticles in human mesenchymal stem cells". Acta Biomaterialia. 7 (1): 347–354.
  37. Compartmentalization Technologies via Self-Assembly and Cross-Linking of Amphiphilic Random Block Copolymers in Water
  38. Drug delivery and nanoparticles: Applications and hazards (good)
  39. The pKa Table Is Your Friend (important)
  40. Nanoparticles as a Targeted Drug Delivery System
  41. Self-Assembled Hydrogel Nanoparticles for Drug Delivery Applications
  42. Gupta M, Gupta AK. In vitro cytotoxicity studies of hydrogel pullulan nanoparticles prepared by AOT/N-hexane micellar system. J Pharm Pharmaceut Sci. 2004;7:38–46.
  43. Biocompatibility and drug delivery systems (good)
  44. S. R. Little and D. S. Kohane, Polymers for intracellular delivery of nucleic acids, J. Mater. Chem., 2008, 18, 832–841
  45. Polymeric Nucleic Acid Delivery for Immunoengineering
  46. Dubernet C., Couvreur P. Nanoparticles in cancer therapy and diagnosis. Adv. Drug Delivery Rev. 2002;54:631–651.
  47. Panyam J., Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv. Drug Delivery Rev. 2003;55:329–347.Moghimi S.M., Hunter A.C., Murray J.C. Long-circulating and target-specific nanoparticles: Theory to practice. Pharmacol. Rev. 2001;53:283–318.
  48. Moghimi S.M., Hunter A.C., Murray J.C. Long-circulating and target-specific nanoparticles: Theory to practice. Pharmacol. Rev. 2001;53:283–318.
  49. Sun T.-M., Du J.-Z., Yan L.-F., Mao H.-Q., Wang J. Self-assembled biodegradable micellar nanoparticles of amphiphilic and cationic block copolymer for siRNA delivery. Biomaterials. 2008;29:4348–4355.
  50. Jain A.K., Goyal A.K., Gupta P.N., Khatri K., Mishra N., Mehta A., Mangal S., Vyas S.P. Synthesis, characterization and evaluation of novel triblock copolymer based nanoparticles for vaccine delivery against hepatitis B. J. Control. Release. 2009;136:161–169.
  51. Niidome T, Yamagata M, Okamoto Y, et al. PEG-modified gold nanorods with a stealth character for in vivo applications. J Control Release. 2006;114:343–7.
  52. M. Goldberg, R. Langer and X. Jia, Nanostructured materials for applications in drug delivery and tissue engineering, J. Biomater. Sci., Polym. Ed., 2007, 18, 241–268.
  53. D. S. Kohane, Microparticles and nanoparticles for drug delivery, Biotechnol. Bioeng., 2007, 96, 203–209
  54. What Are Lipid Nanoparticles in mRNA Vaccines? 
  55. Plard J.P., Bazile D. Comparison of the safety profiles of PLA(50) and Me.PEG-PLA(50) nanoparticles after single dose intravenous administration to rat. Colloids Surf. B. 1999;16:173–183.
  56. Peracchia M.T., Fattal E., Desmaele D., Besnard M., Noel J.P., Gomis J.M., Appel M., d'Angelo J., Couvreur P. Stealth PEGylated polycyanoacrylate nanoparticles for intravenous administration and splenic targeting. J. Control. Release. 1999;60:121–128.
  57. Matsumura Y., Maeda H. A New Concept for macromolecular therapeutics in cancer chemotherapy: Mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs1. Cancer Res. 1986;46:6387–6392.
  58. Brannon-Peppas L., Blanchette J.O. Nanoparticle and targeted systems for cancer therapy. Adv. Drug Delivery Rev. 2004;56:1649–1659.
  59. Moghimi S.M., Hunter A.C., Murray J.C. Nanomedicine: Current status and future prospects. FASEB J. 2005;19:311–330.
  60. Fang C, Shi B, Pei YY, et al. In vivo tumor targeting of tumor necrosis factor-alpha-loaded stealth nanoparticles: Effect of MePEG molecular weight and particle size. Eur J Pharm Sci. 2006;27:27–36.
  61. Shim J, Seok Kang H, Park WS, et al. Transdermal delivery of minoxidil with block copolymer nanoparticles. J Control Release. 2004;97:477–84.
  62. Zhang L, Hu Y, Jiang X, et al. Camptothecin derivative-loaded poly(caprolactone-co-lactide)-b-PEG-b-poly(caprolactone-co-lactide) nanoparticles and their biodistribution in mice. J Control Release. 2004;96:135–48.
  63. A programmable chemical switch based on triggerable Michael acceptors
  64. SELF-IMMOLATIVE CHEMISTRY: Structural Features and Applications in Designing Smart Materials (pdf)
  65. Measuring particle size distribution of nanoparticle enabled medicinal products, the joint view of EUNCL and NCI-NCL. A step by step approach combining orthogonal measurements with increasing complexity
  66. D’Mello SR, Cruz CN, Chen ML, Kapoor M, Lee SL, Tyner KM. The evolving landscape of drug products containing nanomaterials in the United States. Nat. Nanotechnol. 12(6), 523–529 (2017).
  67. US FDA. Guidance Document: Drug Products, Including Biological Products, that Contain Nanomaterials – Guidance for Industry.
  68. EMA. Development of Block-Copolymer-Micelle Medicinal Products (Reference Number EMA/CHMP/130299/2013).
  69. EMA (Committee for Medicinal Products for Human Use). Reflection Paper on the Data Requirements for Intravenous Iron-Based Nano-Colloidal Products Developed with Reference to an Innovator Medicinal Product (2012).
  70. Committee for Human Medicinal Products. Reflection Paper on the Data Requirements for Intravenous Liposomal Products Developed with Reference to Aninnovator Liposomal Product (2013).
  71. US FDA. Liposome Drug Products: Chemistry, Manufacturing, and Controls; Human Pharmacokinetics and Bioavailability; and Labeling Documentation (2018).
  72. Nano-enabled medicinal products: time for an international advanced community?
  73. El-Sayed, M. A. Some interesting properties of metals confined in time and nanometer space of different shapes. Accounts Chem. Res. 34, 257–264 (2001).
  74. Trindade, T., O'Brien, P. & Pickett, N. L. Nanocrystalline semiconductors: synthesis, properties and perspectives. Chem. Mater. 13, 3843–3858 (2001).
  75. Kelly, K. L., Coronado, E., Zhao, L. L. & Schatz, G. C. The optical properties of metal nanoparticles: the influence of size, shape, and dielectric environment. J. Phys. Chem. B. 107, 668–677 (2003).
  76. Seo, W. S. et al. Size-dependent magnetic properties of colloidal Mn3O4 and MnO nanoparticles. Angew. Chem. Int. Ed. 43, 1115–1117 (2004).
  77. Mohanty, A., Garg, N. & Jin, R. A universal approach to the synthesis of noble metal nanodendrites and their catalytic properties. Angew. Chem. Int. Ed. 49, 4962–4966 (2010).
  78. Rao, C. N. R., Kulkarni, G. U., Thomas, P. J. & Edwards, P. P. Size-dependent chemistry: properties of nanocrystals. Chem. Eur. J. 8, 29–35 (2002).
  79. Boal, A. K. et al. Self-assembly of nanoparticles into structured spherical and network aggregates. Nature 404, 746–748 (2000).
  80. Li, P., Kaslan, M., Lee, S. H., Yao, J. & Gao, Z. Progress in exosome isolation techniques. Theranostics 7, 789 (2017).
  81. Lane, R. E., Korbie, D., Anderson, W., Vaidyanathan, R. & Trau, M. Analysis of exosome purification methods using a model liposome system and tunable-resistive pulse sensing. Sci. Rep. 5, 7639 (2015).
  82. Batrakova, E. V. & Kim, M. S. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J. Control. Rel. 219, 396–405 (2015).
  83. Prime, KL; Whitesides, GM (1991). "Self-assembled organic monolayers: model systems for studying adsorption of proteins at surfaces". Science. 252 (5009): 1164–7.
  84. Liu, Wenhao; Greytak, Andrew B.; Lee, Jungmin; Wong, Cliff R.; Park, Jongnam; Marshall, Lisa F.; Jiang, Wen; Curtin, Peter N.; Ting, Alice Y.; Nocera, Daniel G.; Fukumura, Dai; Jain, Rakesh K.; Bawendi, Moungi G. (20 January 2010). 
  85. "Compact Biocompatible Quantum Dots via RAFT-Mediated Synthesis of Imidazole-Based Random Copolymer Ligand". Journal of the American Chemical Society. 132 (2): 472–483.
  86. Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. Biomaterials. 2005;26:3995–4021.
  87. "Nanoparticles play at being red blood cells". Archived from the original on 1 July 2011. Retrieved 1 July 2011.
  88. Surface-Energy Control and Characterization of Nanoparticle Coatings
  89. Core–shell nanostructures: perspectives towards drug delivery applications
  90. Cancer Cell Membrane-Coated Nanoparticles for Anticancer Vaccination and Drug Delivery
  91. Akerman ME, Chan WC, Laakkonen P, Bhatia SN, Ruoslahti E (2002). "Nanocrystal targeting in vivo". Proceedings of the National Academy of Sciences of the United States of America. 99(20): 12617–12621.
  92. Praaning-van Dalen, DP; Brouwer, A; Knook, DL (December 1981). "Clearance capacity of rat liver Kupffer, Endothelial, and parenchymal cells". Gastroenterology. 81 (6): 1036–44.
  93. Anderson, CL (December 2015). "The liver sinusoidal endothelium reappears after being eclipsed by the Kupffer cell: a 20th century biological delusion corrected". Journal of Leukocyte Biology. 98(6): 875–6.
  94. Ganesan, LP; Mohanty, S; Kim, J; Clark, KR; Robinson, JM; Anderson, CL (September 2011). "Rapid and efficient clearance of blood-borne virus by liver sinusoidal endothelium". PLoS Pathogens. 7 (9): e1002281.
  95. Hoshino, A; Fujioka, K; Oku, T; Nakamura, S; Suga, M; Yamaguchi, Y; Suzuki, K; Yasuhara, M; Yamamoto, K (2004). "Quantum dots targeted to the assigned organelle in living cells". Microbiology and Immunology. 48 (12): 985–94.
  96. Suzuki, KG; Fujiwara, TK; Edidin, M; Kusumi, A (2007). "Dynamic recruitment of phospholipase C at transiently immobilized GPI-anchored receptor clusters induces IP3 Ca2+ signaling: single-molecule tracking study 2". The Journal of Cell Biology. 177 (4): 731–42.
  97. Sung, KM; Mosley, DW; Peelle, BR; Zhang, S; Jacobson, JM (2004). "Synthesis of monofunctionalized gold nanoparticles by fmoc solid-phase reactions". Journal of the American Chemical Society. 126 (16): 5064–5.
  98. Fu, A; Micheel, CM; Cha, J; Chang, H; Yang, H; Alivisatos, AP (2004). "Discrete nanostructures of quantum dots/Au with DNA". Journal of the American Chemical Society. 126 (35): 10832–3.
  99. Howarth, M; Liu, W; Puthenveetil, S; Zheng, Y; Marshall, LF; Schmidt, MM; Wittrup, KD; Bawendi, MG; Ting, AY (2008). "Monovalent, reduced-size quantum dots for imaging receptors on living cells". Nature Methods. 5 (5): 397–9.
  100. van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL. [Mononuclear phagocytic system: new classification of macrophages, monocytes and of their cell line]. Bull World Health Organ (1972) 47:651–8.
  101. Jokerst, V. J.; Lobovkina, T.; Zare, N. R.; Gambhir, S. S. Nanoparticle PEGylation for imaging and therapy. Nanomedicine 2012, 6, 715– 728.
  102. Hak, S.; Helgesen, E.; Hektoen, H. H.; Huuse, E. M.; Jarzyna, P. A.; Mulder, W. J. M.; Haraldseth, O.; de Lange Davies, C. L. The Effect of Nanoparticle Polyethylene Glycol Surface Density on Ligand-Directed Tumor Targeting Studied in Vivo by Dual Modality Imaging. ACS Nano 2012, 6, 5648– 5658.
  103. Pelaz, B.; del Pino, P.; Maffre, P.; Hartmann, R.; Gallego, M.; Rivera-Fernández, S.; de la Fuente, J. M.; Nienhaus, G. U.; Parak, W. J. Surface Functionalization of Nanoparticles with Polyethylene Glycol: Effects on Protein Adsorption and Cellular Uptake. ACS Nano 2015, 9, 6996– 7008.
  104. He X, Nie H, Wang K, Tan W, Wu X, Zhang P. Anal Chem. 2008;80:9597–9603
  105. Huang X, Li L, Liu T, Hao N, Liu H, Chen D, Tang F. ACS Nano. 2011;5:5390–5399
  106. Kumar R, Roy I, Ohulchanskky TY, Vathy LA, Bergey EJ, Sajjad M, Prasad PN. ACS Nano. 2010;4:699–708
  107. Liu T, Li L, Teng X, Huang X, Liu H, Chen D, Ren J, He J, Tang F. Biomaterials. 2011;32:1657–1668
  108. Liu Y, Hu Y, Huang L. Biomaterials. 2014;35:3027–3034
  109. Ohno K, Akashi T, Tsujii Y, Yamamoto M, Tabata Y. Biomacromolecules. 2012;13:927–936
  110. Owens DE, 3rd, Peppas NA. Int J Pharm. 2006;307:93–102.
  111. Nanoparticle Uptake: The Phagocyte Problem.
  112. Dobrovolskaia MA, Aggarwal P, Hall JB, McNeil SE. Mol Pharm. 2008;5:487–495.
  113. Jenkin CR, Rowley D. J Exp Med. 1961;114:363–374.
  114. Absolom DR. Methods Enzymol. 1986;132:281–318.
  115. Mortimer GM, Butcher NJ, Musumeci AW, Deng ZJ, Martin DJ, Minchin RF. ACS Nano. 2014;8:3357–3366.
  116. Andrade J, Hlady V. Biopolymers/Non-Exclusion HPLC. Springer; 1986. Protein adsorption and materials biocompatibility: a tutorial review and suggested hypotheses; pp. 1–63. 
  117. Andrade JD, Hlady VL, Van Wagenen RA. Pure Appl Chem. 1984;56:1345–1350.
  118. Vertegel AA, Siegel RW, Dordick JS. Langmuir. 2004;20:6800–6807
  119. Davidson WS, Jonas A, Clayton DF, George JM. J Biol Chem. 1998;273:9443–9449.
  120. Lord MS, Foss M, Besenbacher F. Nano Today. 2010;5:66–78
  121. Walkey CD, Chan WC. Chem Soc Rev. 2012;41:2780–2799
  122. Banda NK, Mehta G, Chao Y, Wang G, Inturi S, Fossati-Jimack L, Botto M, Wu L, Moghimi SM, Simberg D. Part Fibre Toxicol. 2014;11:64
  123. Nano based drug delivery systems: recent developments and future prospects (good)
  124. Chapter 31 - Advances in delivery of nanomedicines and theranostics for targeting breast cancer
  125. Role of drug delivery technologies in the success of COVID-19 vaccines: a perspective

              No comments:

              Post a Comment