Anticancer Targeted Drug Delivery Nanotechnology: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Wei Wang.

The construction of nanosized drug delivery systems possesses tremendous potential due to their ability to improve the solubility of poorly soluble drugs and to reduce metabolism by dissolving them in their hydrophobic or hydrophilic compartment. In addition, nanomedicine holds the advantages of passive targeting ability due to an enhanced permeability and retention (EPR) effect, a large surface-to-volume ratio for drug loading, a tunable size for modification, a prolonged plasma half-life and a different biodistribution profile compared to conventional chemotherapy. Typical nano-based delivery vehicles include liposome, micelle, dendrimer, inorganic vector, nanogel and nanoemulsion, while novel nanocarriers also contain biomimetic reconstituted high-density lipoprotein (rHDL), exosome and the hybrid nanoparticle, which come from the mixture of nanomaterials. Each of these nanotools displays its unique physiochemical properties and possesses the ability for further modification of active targeting ligands.

  • nanotechnology
  • encapsulation strategy
  • targeted drug delivery
  • cancer therapy
  • chemotherapeutics
  • biopharmaceutics
Please wait, diff process is still running!

References

  1. Franco, M.S.; Gomes, E.R.; Roque, M.C.; Oliveira, M.C. Triggered Drug Release From Liposomes: Exploiting the Outer and Inner Tumor Environment. Front. Oncol. 2021, 11, 623760.
  2. Lakshmi, B.A.; Reddy, A.S.; Sangubotla, R.; Hong, J.W.; Kim, S. Ruthenium(II)-curcumin liposome nanoparticles: Synthesis, characterization, and their effects against cervical cancer. Colloids Surf. B Biointerfaces 2021, 204, 111773.
  3. Zylberberg, C.; Matosevic, S. Pharmaceutical liposomal drug delivery: A review of new delivery systems and a look at the regulatory landscape. Drug Deliv. 2016, 23, 3319–3329.
  4. Alwattar, J.K.; Mneimneh, A.T.; Abla, K.K.; Mehanna, M.M.; Allam, A.N. Smart Stimuli-Responsive Liposomal Nanohybrid Systems: A Critical Review of Theranostic Behavior in Cancer. Pharmaceutics 2021, 13, 355.
  5. Lu, W.; Yao, J.; Zhu, X.; Qi, Y. Nanomedicines: Redefining traditional medicine. Biomed. Pharm. 2021, 134, 111103.
  6. Han, B.; Yang, Y.; Chen, J.; Tang, H.; Sun, Y.; Zhang, Z.; Wang, Z.; Li, Y.; Li, Y.; Luan, X.; et al. Preparation, Characterization, and Pharmacokinetic Study of a Novel Long-Acting Targeted Paclitaxel Liposome with Antitumor Activity. Int. J. Nanomed. 2020, 15, 553–571.
  7. Elamir, A.; Ajith, S.; Sawaftah, N.A.; Abuwatfa, W.; Mukhopadhyay, D.; Paul, V.; Al-Sayah, M.H.; Awad, N.; Husseini, G.A. Ultrasound-triggered herceptin liposomes for breast cancer therapy. Sci. Rep. 2021, 11, 7545.
  8. Darwitan, A.; Tan, Y.F.; Wong, Y.S.; Nedumaran, A.M.; Czarny, B.; Venkatraman, S. Targeting efficiency of nanoliposomes on atherosclerotic foam cells: Polyethylene glycol-to-ligand ratio effects. Expert Opin. Drug Deliv. 2020, 17, 1165–1176.
  9. Bogner, J.R.; Kronawitter, U.; Rolinski, B.; Truebenbach, K.; Goebel, F.D. Liposomal doxorubicin in the treatment of advanced AIDS-related Kaposi sarcoma. J. Acquir. Immune Defic. Syndr. 1994, 7, 463–468.
  10. Mohamed, M.; Abu Lila, A.S.; Shimizu, T.; Alaaeldin, E.; Hussein, A.; Sarhan, H.A.; Szebeni, J.; Ishida, T. PEGylated liposomes: Immunological responses. Sci. Technol. Adv. Mater. 2019, 20, 710–724.
  11. Ichihara, M.; Shimizu, T.; Imoto, A.; Hashiguchi, Y.; Uehara, Y.; Ishida, T.; Kiwada, H. Anti-PEG IgM Response against PEGylated Liposomes in Mice and Rats. Pharmaceutics 2010, 3, 1–11.
  12. Rommasi, F.; Esfandiari, N. Liposomal Nanomedicine: Applications for Drug Delivery in Cancer Therapy. Nanoscale Res. Lett. 2021, 16, 95.
  13. Mojarad-Jabali, S.; Farshbaf, M.; Walker, P.R.; Hemmati, S.; Fatahi, Y.; Zakeri-Milani, P.; Sarfraz, M.; Valizadeh, H. An update on actively targeted liposomes in advanced drug delivery to glioma. Int. J. Pharm. 2021, 602, 120645.
  14. Lee, Y.; Thompson, D.H. Stimuli-responsive liposomes for drug delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9, 10.
  15. Wang, Y.; Kohane, D.S. External triggering and triggered targeting strategies for drug delivery. Nat. Rev. Mater. 2017, 2, 17020.
  16. Haggag, Y.; Abu Ras, B.; El-Tanani, Y.; Tambuwala, M.M.; McCarron, P.; Isreb, M.; El-Tanani, M. Co-delivery of a RanGTP inhibitory peptide and doxorubicin using dual-loaded liposomal carriers to combat chemotherapeutic resistance in breast cancer cells. Expert Opin. Drug Deliv. 2020, 17, 1655–1669.
  17. Liu, J.; Zhao, Z.; Qiu, N.; Zhou, Q.; Wang, G.; Jiang, H.; Piao, Y.; Zhou, Z.; Tang, J.; Shen, Y. Co-delivery of IOX1 and doxorubicin for antibody-independent cancer chemo-immunotherapy. Nat. Commun. 2021, 12, 2425.
  18. Nandi, I.; Aroeti, L.; Ramachandran, R.P.; Kassa, E.G.; Zlotkin-Rivkin, E.; Aroeti, B. Type III secreted effectors that target mitochondria. Cell. Microbiol. 2021, e13352.
  19. Xia, J.; Qian, M.; Yao, Q.; Meng, Z.; Cui, H.; Zhang, L.; Li, Y.; Wu, S.; Wang, J.; Chen, Q.; et al. Synthetic infrared nano-photosensitizers with hierarchical zoom-in target-delivery functionalities for precision photodynamic therapy. J. Control. Release 2021, 334, 263–274.
  20. Chen, Y.; Ai, W.; Guo, X.; Li, Y.; Ma, Y.; Chen, L.; Zhang, H.; Wang, T.; Zhang, X.; Wang, Z. Mitochondria-Targeted Polydopamine Nanocomposite with AIE Photosensitizer for Image-Guided Photodynamic and Photothermal Tumor Ablation. Small 2019, 15, e1902352.
  21. Jiang, L.; Zhou, S.; Zhang, X.; Li, C.; Ji, S.; Mao, H.; Jiang, X. Mitochondrion-specific dendritic lipopeptide liposomes for targeted sub-cellular delivery. Nat. Commun. 2021, 12, 2390.
  22. Liu, Y.; Qin, Y.; Zhang, Q.; Zou, W.; Jin, L.; Guo, R. Arginine-rich peptide/platinum hybrid colloid nanoparticle cluster: A single nanozyme mimicking multi-enzymatic cascade systems in peroxisome. J. Colloid. Interface Sci. 2021, 600, 37–48.
  23. You, Q.; Zhang, K.; Liu, J.; Liu, C.; Wang, H.; Wang, M.; Ye, S.; Gao, H.; Lv, L.; Wang, C.; et al. Persistent Regulation of Tumor Hypoxia Microenvironment via a Bioinspired Pt-Based Oxygen Nanogenerator for Multimodal Imaging-Guided Synergistic Phototherapy. Adv. Sci. 2020, 7, 1903341.
  24. Liu, X.-L.; Dong, X.; Yang, S.-C.; Lai, X.; Liu, H.-J.; Gao, Y.; Feng, H.-Y.; Zhu, M.-H.; Yuan, Y.; Lu, Q.; et al. Biomimetic Liposomal Nanoplatinum for Targeted Cancer Chemophototherapy. Adv. Sci. 2021, 8, 2003679.
  25. Bazylianska, V.; Kalpage, H.A.; Wan, J.; Vaishnav, A.; Mahapatra, G.; Turner, A.A.; Chowdhury, D.D.; Kim, K.; Morse, P.T.; Lee, I.; et al. Lysine 53 Acetylation of Cytochrome c in Prostate Cancer: Warburg Metabolism and Evasion of Apoptosis. Cells 2021, 10, 802.
  26. Xiao, S.; Wang, X.; Xu, L.; Miao, D.; Li, T.; Su, G.; Zhao, Y. Novel ginsenoside derivatives have shown their effects on PC-3 cells by inducing G1-phase arrest and reactive oxygen species-mediate cell apoptosis. Bioorg. Chem. 2021, 112, 104864.
  27. Chen, X.; Zhu, Q.; Xu, X.; Shen, S.; Zhang, Y.; Mo, R. Sequentially Site-Specific Delivery of Apoptotic Protein and Tumor-Suppressor Gene for Combination Cancer Therapy. Small 2019, 15, e1902998.
  28. Song, X.; Xu, J.; Liang, C.; Chao, Y.; Jin, Q.; Wang, C.; Chen, M.; Liu, Z. Self-Supplied Tumor Oxygenation through Separated Liposomal Delivery of H2O2 and Catalase for Enhanced Radio-Immunotherapy of Cancer. Nano Lett. 2018, 18, 6360–6368.
  29. Pownall, H.J.; Rosales, C.; Gillard, B.K.; Ferrari, M. Native and Reconstituted Plasma Lipoproteins in Nanomedicine: Physicochemical Determinants of Nanoparticle Structure, Stability, and Metabolism. Methodist Debakey Cardiovasc. J. 2016, 12, 146–150.
  30. Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433.
  31. Guerin, M.; Le Goff, W.; Lassel, T.S.; Van Tol, A.; Steiner, G.; Chapman, M.J. Atherogenic role of elevated CE transfer from HDL to VLDL(1) and dense LDL in type 2 diabetes: Impact of the degree of triglyceridemia. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 282–288.
  32. Zhang, P.; Gao, J.; Pu, C.; Feng, G.; Wang, L.; Huang, L.; Zhang, Y. ApoM/HDL-C and apoM/apoA-I ratios are indicators of diabetic nephropathy in healthy controls and type 2 diabetes mellitus. Clin. Chim. Acta 2017, 466, 31–37.
  33. Ertek, S. High-density Lipoprotein (HDL) Dysfunction and the Future of HDL. Curr. Vasc. Pharmacol. 2018, 16, 490–498.
  34. Riggs, K.A.; Rohatgi, A. HDL and Reverse Cholesterol Transport Biomarkers. Methodist Debakey Cardiovasc. J. 2019, 15, 39–46.
  35. Kuai, R.; Li, D.; Chen, Y.E.; Moon, J.J.; Schwendeman, A. High-Density Lipoproteins: Nature’s Multifunctional Nanoparticles. ACS Nano 2016, 10, 3015–3041.
  36. Jiang, C.; Qi, Z.; Tang, Y.; Jia, H.; Li, Z.; Zhang, W.; Liu, J. Rational Design of Lovastatin-Loaded Spherical Reconstituted High Density Lipoprotein for Efficient and Safe Anti-Atherosclerotic Therapy. Mol. Pharm. 2019, 16, 3284–3291.
  37. Twiddy, A.L.; Cox, M.E.; Wasan, K.M. Knockdown of scavenger receptor class B type I reduces prostate specific antigen secretion and viability of prostate cancer cells. Prostate 2012, 72, 955–965.
  38. Babitt, J.; Trigatti, B.; Rigotti, A.; Smart, E.J.; Anderson, R.G.; Xu, S.; Krieger, M. Murine SR-BI, a high density lipoprotein receptor that mediates selective lipid uptake, is N-glycosylated and fatty acylated and colocalizes with plasma membrane caveolae. J. Biol. Chem. 1997, 272, 13242–13249.
  39. Witkowski, A.; Carta, S.; Lu, R.; Yokoyama, S.; Rubartelli, A.; Cavigiolio, G. Oxidation of methionine residues in human apolipoprotein A-I generates a potent pro-inflammatory molecule. J. Biol. Chem. 2019, 294, 3634–3646.
  40. Monroy-Iglesias, M.J.; Russell, B.; Crawley, D.; Allen, N.E.; Travis, R.C.; Perez-Cornago, A.; Van Hemelrijck, M.; Beckmann, K. Metabolic syndrome biomarkers and prostate cancer risk in the UK Biobank. Int. J. Cancer 2021, 148, 825–834.
  41. Revilla, G.; Cedo, L.; Tondo, M.; Moral, A.; Perez, J.I.; Corcoy, R.; Lerma, E.; Fuste, V.; Reddy, S.T.; Blanco-Vaca, F.; et al. LDL, HDL and endocrine-related cancer: From pathogenic mechanisms to therapies. Semin. Cancer Biol. 2020, 73, 134–157.
  42. Pedersbaek, D.; Kraemer, M.K.; Kempen, P.J.; Ashley, J.; Braesch-Andersen, S.; Andresen, T.L.; Simonsen, J.B. The Composition of Reconstituted High-Density Lipoproteins (rHDL) Dictates the Degree of rHDL Cargo- and Size-Remodeling via Direct Interactions with Endogenous Lipoproteins. Bioconjug. Chem. 2019, 30, 2634–2646.
  43. Mei, Y.; Tang, L.; Xiao, Q.; Zhang, Z.; Zhang, Z.; Zang, J.; Zhou, J.; Wang, Y.; Wang, W.; Ren, M. Reconstituted high density lipoprotein (rHDL), a versatile drug delivery nanoplatform for tumor targeted therapy. J. Mater. Chem. B 2021, 9, 612–633.
  44. Yoo, J.W.; Irvine, D.J.; Discher, D.E.; Mitragotri, S. Bio-inspired, bioengineered and biomimetic drug delivery carriers. Nat. Rev. Drug Discov. 2011, 10, 521–535.
  45. Zorko, M.; Langel, U. Cell-penetrating peptides: Mechanism and kinetics of cargo delivery. Adv. Drug Deliv. Rev. 2005, 57, 529–545.
  46. Pandey, M.K.; Sung, B.; Ahn, K.S.; Kunnumakkara, A.B.; Chaturvedi, M.M.; Aggarwal, B.B. Gambogic acid, a novel ligand for transferrin receptor, potentiates TNF-induced apoptosis through modulation of the nuclear factor-kappaB signaling pathway. Blood 2007, 110, 3517–3525.
  47. Ding, Y.; Wang, Y.; Opoku-Damoah, Y.; Wang, C.; Shen, L.; Yin, L.; Zhou, J. Dual-functional bio-derived nanoparticulates for apoptotic antitumor therapy. Biomaterials 2015, 72, 90–103.
  48. Tse, J.; Singla, N.; Ghandour, R.; Lotan, Y.; Margulis, V. Current advances in BCG-unresponsive non-muscle invasive bladder cancer. Expert Opin. Investig. Drugs 2019, 28, 757–770.
  49. Sabnis, S.; Sabnis, N.A.; Raut, S.; Lacko, A.G. Superparamagnetic reconstituted high-density lipoprotein nanocarriers for magnetically guided drug delivery. Int. J. Nanomed. 2017, 12, 1453–1464.
  50. Ryther, R.C.; Flynt, A.S.; Phillips, J.A., 3rd; Patton, J.G. siRNA therapeutics: Big potential from small RNAs. Gene Ther. 2005, 12, 5–11.
  51. Ding, Y.; Wang, Y.; Zhou, J.; Gu, X.; Wang, W.; Liu, C.; Bao, X.; Wang, C.; Li, Y.; Zhang, Q. Direct cytosolic siRNA delivery by reconstituted high density lipoprotein for target-specific therapy of tumor angiogenesis. Biomaterials 2014, 35, 7214–7227.
  52. Li, Y.; Cai, B.; Shen, L.; Dong, Y.; Lu, Q.; Sun, S.; Liu, S.; Ma, S.; Ma, P.X.; Chen, J. MiRNA-29b suppresses tumor growth through simultaneously inhibiting angiogenesis and tumorigenesis by targeting Akt3. Cancer Lett. 2017, 397, 111–119.
  53. Chen, X.; Mangala, L.S.; Mooberry, L.; Bayraktar, E.; Dasari, S.K.; Ma, S.; Ivan, C.; Court, K.A.; Rodriguez-Aguayo, C.; Bayraktar, R.; et al. Identifying and targeting angiogenesis-related microRNAs in ovarian cancer. Oncogene 2019, 38, 6095–6108.
  54. Haftka, J.J.; Scherpenisse, P.; Oetter, G.; Hodges, G.; Eadsforth, C.V.; Kotthoff, M.; Hermens, J.L. Critical micelle concentration values for different surfactants measured with solid-phase microextraction fibers. Environ. Toxicol. Chem. 2016, 35, 2173–2781.
  55. Cheng, F.R.; Su, T.; Cao, J.; Luo, X.L.; Li, L.; Pu, Y.; He, B. Environment-stimulated nanocarriers enabling multi-active sites for high drug encapsulation as an “on demand” drug release system. J. Mater. Chem. B 2018, 6, 2258–2273.
  56. Ghosh, S.; Ray, A.; Pramanik, N. Self-assembly of surfactants: An overview on general aspects of amphiphiles. Biophys. Chem. 2020, 265, 106429.
  57. Croy, S.R.; Kwon, G.S. Polymeric micelles for drug delivery. Curr. Pharm. Des. 2006, 12, 4669–4684.
  58. Irby, D.; Du, C.; Li, F. Lipid-Drug Conjugate for Enhancing Drug Delivery. Mol. Pharm. 2017, 14, 1325–1338.
  59. Danov, K.D.; Kralchevsky, P.A.; Stoyanov, S.D.; Cook, J.L.; Stott, I.P. Analytical modeling of micelle growth. 1. Chain-conformation free energy of binary mixed spherical, wormlike and lamellar micelles. J. Colloid. Interface Sci. 2019, 547, 245–255.
  60. Zhao, J.; Wang, Y.; Luan, L. Star-shaped polycaprolactone-polyethyleneglycol copolymer micelle-like nanoparticles for picropodophyllin delivery. J. Biomed. Nanotechnol. 2014, 10, 1627–1634.
  61. Isoglu, I.A.; Ozsoy, Y.; Isoglu, S.D. Advances in Micelle-based Drug Delivery: Cross-linked Systems. Curr. Top. Med. Chem. 2017, 17, 1469–1489.
  62. Keam, B.; Lee, K.W.; Lee, S.H.; Kim, J.S.; Kim, J.H.; Wu, H.G.; Eom, K.Y.; Kim, S.; Ahn, S.H.; Chung, E.J.; et al. A Phase II Study of Genexol-PM and Cisplatin as Induction Chemotherapy in Locally Advanced Head and Neck Squamous Cell Carcinoma. Oncologist 2019, 24, 751-e231.
  63. Du, J.H.; Zhang, H.D.; Ma, Z.J.; Ji, K.M. Artesunate induces oncosis-like cell death in vitro and has antitumor activity against pancreatic cancer xenografts in vivo. Cancer Chemother. Pharmacol. 2010, 65, 895–902.
  64. Efferth, T.; Kaina, B. Toxicity of the antimalarial artemisinin and its dervatives. Crit. Rev. Toxicol. 2010, 40, 405–421.
  65. Hao, D.L.; Xie, R.; De, G.J.; Yi, H.; Zang, C.; Yang, M.Y.; Liu, L.; Ma, H.; Cai, W.Y.; Zhao, Q.H.; et al. pH-Responsive Artesunate Polymer Prodrugs with Enhanced Ablation Effect on Rodent Xenograft Colon Cancer. Int. J. Nanomed. 2020, 15, 1771–1786.
  66. Clamme, J.P.; Krishnamoorthy, G.; Mely, Y. Intracellular dynamics of the gene delivery vehicle polyethylenimine during transfection: Investigation by two-photon fluorescence correlation spectroscopy. Biochim. Biophys. Acta 2003, 1617, 52–61.
  67. Manda-Handzlik, A.; Bystrzycka, W.; Cieloch, A.; Glodkowska-Mrowka, E.; Jankowska-Steifer, E.; Heropolitanska-Pliszka, E.; Skrobot, A.; Muchowicz, A.; Ciepiela, O.; Wachowska, M.; et al. Nitric oxide and peroxynitrite trigger and enhance release of neutrophil extracellular traps. Cell. Mol. Life Sci. 2020, 77, 3059–3075.
  68. Lin, Y.J.; Chen, C.C.; Nguyen, D.; Su, H.R.; Lin, K.J.; Chen, H.L.; Hu, Y.J.; Lai, P.L.; Sung, H.W. Biomimetic Engineering of a Scavenger-Free Nitric Oxide-Generating/Delivering System to Enhance Radiation Therapy. Small 2020, 16, e2000655.
  69. Napoli, A.; Valentini, M.; Tirelli, N.; Muller, M.; Hubbell, J.A. Oxidation-responsive polymeric vesicles. Nat. Mater. 2004, 3, 183–189.
  70. Wu, W.; Chen, M.; Luo, T.; Fan, Y.; Zhang, J.; Zhang, Y.; Zhang, Q.; Sapin-Minet, A.; Gaucher, C.; Xia, X. ROS and GSH-responsive S-nitrosoglutathione functionalized polymeric nanoparticles to overcome multidrug resistance in cancer. Acta Biomater. 2020, 103, 259–271.
  71. Li, J.; Wang, J.; Zhang, X.; Xia, X.; Zhang, C. Biodegradable reduction-responsive polymeric micelles for enhanced delivery of melphalan to retinoblastoma cells. Int. J. Biol. Macromol. 2019, 141, 997–1003.
  72. Mimeault, M.; Batra, S.K. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J. Cell. Mol. Med. 2013, 17, 30–54.
  73. Xin, X.; Kumar, V.; Lin, F.; Kumar, V.; Bhattarai, R.; Bhatt, V.R.; Tan, C.; Mahato, R.I. Redox-responsive nanoplatform for codelivery of miR-519c and gemcitabine for pancreatic cancer therapy. Sci. Adv. 2020, 6, eabd6764.
  74. Lu, F.; Hou, L.; Wang, S.; Yu, Y.; Zhang, Y.; Sun, L.; Wang, C.; Ma, Z.; Yang, F. Lysosome activable polymeric vorinostat encapsulating PD-L1KD for a combination of HDACi and immunotherapy. Drug Deliv. 2021, 28, 963–972.
  75. Cao, J.; Huang, D.; Peppas, N.A. Advanced engineered nanoparticulate platforms to address key biological barriers for delivering chemotherapeutic agents to target sites. Adv. Drug Deliv. Rev. 2020, 167, 170–188.
  76. Wu, L.P.; Ficker, M.; Christensen, J.B.; Trohopoulos, P.N.; Moghimi, S.M. Dendrimers in Medicine: Therapeutic Concepts and Pharmaceutical Challenges. Bioconjug. Chem. 2015, 26, 1198–1211.
  77. Parat, A.; Bordeianu, C.; Dib, H.; Garofalo, A.; Walter, A.; Begin-Colin, S.; Felder-Flesch, D. Dendrimer-nanoparticle conjugates in nanomedicine. Nanomedicine 2015, 10, 977–992.
  78. Zamboulis, A.; Nakiou, E.A.; Christodoulou, E.; Bikiaris, D.N.; Kontonasaki, E.; Liverani, L.; Boccaccini, A.R. Polyglycerol Hyperbranched Polyesters: Synthesis, Properties and Pharmaceutical and Biomedical Applications. Int. J. Mol. Sci. 2019, 20, 6210.
  79. Singh, P.; Gupta, U.; Asthana, A.; Jain, N.K. Folate and folate-PEG-PAMAM dendrimers: Synthesis, characterization, and targeted anticancer drug delivery potential in tumor bearing mice. Bioconjug. Chem. 2008, 19, 2239–2252.
  80. Wang, F.; Cai, X.; Su, Y.; Hu, J.; Wu, Q.; Zhang, H.; Xiao, J.; Cheng, Y. Reducing cytotoxicity while improving anti-cancer drug loading capacity of polypropylenimine dendrimers by surface acetylation. Acta Biomater. 2012, 8, 4304–4313.
  81. Sinokrot, H.; Smerat, T.; Najjar, A.; Karaman, R. Advanced Prodrug Strategies in Nucleoside and Non-Nucleoside Antiviral Agents: A Review of the Recent Five Years. Molecules 2017, 22, 1736.
  82. Yuan, F.; Leunig, M.; Huang, S.K.; Berk, D.A.; Papahadjopoulos, D.; Jain, R.K. Microvascular permeability and interstitial penetration of sterically stabilized (stealth) liposomes in a human tumor xenograft. Cancer Res. 1994, 54, 3352–3356.
  83. Sun, Q.; Sun, X.; Ma, X.; Zhou, Z.; Jin, E.; Zhang, B.; Shen, Y.; Van Kirk, E.A.; Murdoch, W.J.; Lott, J.R.; et al. Integration of nanoassembly functions for an effective delivery cascade for cancer drugs. Adv. Mater. 2014, 26, 7615–7621.
More