Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders
Abstract
:1. Introduction
2. The Immune System in the Brain
3. Challenges in Nanomedicine-Based Immunotherapy in the Brain
3.1. The BBB
3.2. Blood–Cerebrospinal Fluid Barrier
3.3. Multidrug Resistance Proteins
4. Nanomedicines for Targeting CNS Diseases
5. Surface Modification of Nanoparticles
6. Applications of Nanotechnology in CNS Disorders
6.1. Glioblastoma
6.1.1. Cellular Immunology for Malignant Gliomas
6.1.2. Vaccine-Based Immunotherapy of Glioblastomas
6.1.3. Cargo-Loaded NP-Based Immunotherapy of Glioma
6.1.4. Nanomedicine-Based Combination Therapy
6.1.5. Gene Therapy for Glioma
6.1.6. Chemotherapy
6.2. Alzheimer’s Disease (AD)
6.3. Parkinson’s Disease (PD)
7. Huntington’s Disease
8. Ongoing Clinical Trials and Approved Nanomedicines
9. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Lammers, T.; Aime, S.; Hennink, W.E.; Storm, G.; Kiessling, F. Theranostic nanomedicine. Acc. Chem. Res. 2011, 44, 1029–1038. [Google Scholar] [CrossRef] [PubMed]
- Zheng, M.; Tao, W.; Zou, Y.; Farokhzad, O.C.; Shi, B. Nanotechnology-based strategies for siRNA brain delivery for disease therapy. Trends Biotechnol. 2018, 36, 562–575. [Google Scholar] [CrossRef] [PubMed]
- Tayyba, T. Role of extracellular vesicles in human diseases. Biomed. Lett. 2019, 5, 67–68. [Google Scholar]
- Kim, B.Y.; Rutka, J.T.; Chan, W.C. Nanomedicine. N. Engl. J. Med. 2010, 363, 2434–2443. [Google Scholar] [CrossRef] [Green Version]
- Song, S.; Gao, P.; Sun, L.; Kang, D.; Kongsted, J.; Poongavanam, V.; Zhan, P.; Liu, X. Recent developments in the medicinal chemistry of single boron atom-containing compounds. Acta Pharm. Sin. B 2021, 11, 3035–3059. [Google Scholar] [CrossRef]
- Fernandes, G.F.S.; Denny, W.A.; Santos, J.L.D. Boron in drug design: Recent advances in the development of new therapeutic agents. Eur. J. Med. Chem. 2019, 179, 791–804. [Google Scholar] [CrossRef]
- Soriano-Ursúa, M.A.; Farfán-García, E.D.; Geninatti-Crich, S. Turning fear of boron toxicity into boron-containing drug design. Curr. Med. Chem. 2019, 26, 5005–5018. [Google Scholar] [CrossRef] [PubMed]
- Ritacca, A.G.; Ritacco, I.; Dabbish, E.; Russo, N.; Mazzone, G.; Sicilia, E. A Boron-Containing Compound Acting on Multiple Targets Against Alzheimer’s Disease. Insights from Ab Initio and Molecular Dynamics Simulations. J. Chem. Inf. Model. 2021, 61, 3397–3410. [Google Scholar] [CrossRef]
- Jiménez-Aligaga, K.; Bermejo-Bescós, P.; Martín-Aragón, S.; Csákÿ, A.G. Discovery of alkenylboronic acids as neuroprotective agents affecting multiple biological targets involved in Alzheimer’s disease. Bioorganic Med. Chem. Lett. 2013, 23, 426–429. [Google Scholar] [CrossRef]
- Lu, C.-J.; Hu, J.; Wang, Z.; Xie, S.; Pan, T.; Huang, L.; Li, X. Discovery of boron-containing compounds as Aβ aggregation inhibitors and antioxidants for the treatment of Alzheimer’s disease. MedChemComm 2018, 9, 1862–1870. [Google Scholar] [CrossRef]
- Maiti, P.; Manna, J.; Burch, Z.N.; Flaherty, D.B.; Larkin, J.D.; Dunbar, G.L. Ameliorative Properties of Boronic Compounds in In Vitro and In Vivo Models of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 6664. [Google Scholar] [CrossRef] [PubMed]
- Amiji, M.M. Nanotechnology for Cancer Therapy; CRC Press: Boca Raton, FL, USA, 2006. [Google Scholar]
- Athar, M.; Das, A.J. Therapeutic nanoparticles: State-of-the-art of nanomedicine. Adv. Mater. Rev 2014, 1, 25–37. [Google Scholar]
- Rehman, F.U. Nanomedicine: Why it still taking long from “bench to bedside”? Biomed. Lett. 2018, 4, 1373–1378. [Google Scholar]
- Chapman, C.D.; Frey, W.H.; Craft, S.; Danielyan, L.; Hallschmid, M.; Schiöth, H.B.; Benedict, C. Intranasal Treatment of Central Nervous System Dysfunction in Humans. Pharm. Res. 2013, 30, 2475–2484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pridgen, E.M.; Langer, R.; Farokhzad, O.C. Biodegradable, polymeric nanoparticle delivery systems for cancer therapy. Nanomedicine 2007, 2, 669–680. [Google Scholar] [CrossRef]
- Wagner, V.; Dullaart, A.; Bock, A.-K.; Zweck, A. The emerging nanomedicine landscape. Nat. Biotechnol. 2006, 24, 1211. [Google Scholar] [CrossRef]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
- Ozdemir-Kaynak, E.; Qutub, A.A.; Yesil-Celiktas, O. Advances in Glioblastoma Multiforme Treatment: New Models for Nanoparticle Therapy. Front. Physiol. 2018, 9, 170. [Google Scholar] [CrossRef] [Green Version]
- Lie, D.C.; Song, H.; Colamarino, S.A.; Ming, G.-l.; Gage, F.H. Neurogenesis IN THE Adult Brain: New. Annu. Rev. Pharmacol. Toxicol. 2004, 44, 399–421. [Google Scholar] [CrossRef] [Green Version]
- Wen, P.Y.; Kesari, S. Malignant gliomas in adults. New England J. Med. 2008, 359, 492–507. [Google Scholar] [CrossRef] [Green Version]
- Zanganeh, S.; Georgala, P.; Corbo, C.; Arabi, L.; Ho, J.Q.; Javdani, N.; Sepand, M.R.; Cruickshank, K.; Campesato, L.F.; Weng, C.H. Immunoengineering in glioblastoma imaging and therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2019, 11, e1575. [Google Scholar] [CrossRef]
- Perry, J.R.; Laperriere, N.; O’Callaghan, C.J.; Brandes, A.A.; Menten, J.; Phillips, C.; Fay, M.; Nishikawa, R.; Cairncross, J.G.; Roa, W. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N. Engl. J. Med. 2017, 376, 1027–1037. [Google Scholar] [CrossRef] [Green Version]
- Poewe, W.; Seppi, K.; Tanner, C.; Halliday, G.; Brundin, P.; Volkmann, J.; Schrag, A.; Lang, A. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef]
- Hanif, S.; Muhammad, P.; Chesworth, R.; Rehman, F.U.; Qian, R.-j.; Zheng, M.; Shi, B.-y. Nanomedicine-based immunotherapy for central nervous system disorders. Acta Pharmacol. Sin. 2020, 41, 936–953. [Google Scholar] [CrossRef] [PubMed]
- Behrens, S.; Rattinger, G.B.; Schwartz, S.; Matyi, J.; Sanders, C.; DeBerard, M.S.; Lyketsos, C.G.; Tschanz, J.T. Use of FDA approved medications for Alzheimer’s disease in mild dementia is associated with reduced informal costs of care. Int. Psychogeriatr. 2018, 30, 1499–1507. [Google Scholar] [CrossRef]
- Kelly, C.B.; Milligan, J.A.; Tilley, L.J.; Sodano, T.M. Bicyclobutanes: From curiosities to versatile reagents and covalent warheads. Chem. Sci. 2022, 13, 11721–11737. [Google Scholar] [CrossRef] [PubMed]
- Ellis, J.M.; Fell, M.J. Current approaches to the treatment of Parkinson’s Disease. Bioorganic Med. Chem. Lett. 2017, 27, 4247–4255. [Google Scholar] [CrossRef] [PubMed]
- Han, J.; Kiss, L.; Mei, H.; Remete, A.M.; Ponikvar-Svet, M.; Sedgwick, D.M.; Roman, R.; Fustero, S.; Moriwaki, H.; Soloshonok, V.A. Chemical aspects of human and environmental overload with fluorine. Chem. Rev. 2021, 121, 4678–4742. [Google Scholar] [CrossRef]
- Madajewicz, S.; Chowhan, N.; Tfayli, A.; Roque, C.; Meek, A.; Davis, R.; Wolf, W.; Cabahug, C.; Roche, P.; Manzione, J. Therapy for patients with high grade astrocytoma using intraarterial chemotherapy and radiation therapy. Cancer 2000, 88, 2350–2356. [Google Scholar] [CrossRef]
- Buglioni, L.; Raymenants, F.; Slattery, A.; Zondag, S.D.; Noël, T. Technological innovations in photochemistry for organic synthesis: Flow chemistry, high-throughput experimentation, scale-up, and photoelectrochemistry. Chem. Rev. 2021, 122, 2752–2906. [Google Scholar] [CrossRef]
- Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [Green Version]
- Till, S.J.; Francis, J.N.; Nouri-Aria, K.; Durham, S.R. Mechanisms of immunotherapy. J. Allergy Clin. Immunol. 2004, 113, 1025–1034. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Jiang, W.; Nam, J.; Moon, J.J.; Kim, B.Y. Immunomodulating nanomedicine for cancer therapy. Nano Lett. 2018, 18, 6655–6659. [Google Scholar] [CrossRef]
- Cifuentes-Rius, A.; Desai, A.; Yuen, D.; Johnston, A.P.; Voelcker, N.H. Inducing immune tolerance with dendritic cell-targeting nanomedicines. Nat. Nanotechnol. 2021, 16, 37–46. [Google Scholar] [CrossRef]
- Bejarano, L.; Jordāo, M.J.; Joyce, J.A. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
- Brody, D.L.; Holtzman, D.M. Active and passive immunotherapy for neurodegenerative disorders. Annu. Rev. Neurosci. 2008, 31, 175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, W.; Gao, C.; Wang, P.; Huang, J.; Qian, Y.; Guo, L.; Zhang, J.; Jiang, R. Correlation of circulating T lymphocytes and intracranial hypertension in intracerebral hemorrhage. World Neurosurg. 2017, 107, 389–395. [Google Scholar] [CrossRef] [PubMed]
- Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef] [PubMed]
- Rustenhoven, J.; Jansson, D.; Smyth, L.C.; Dragunow, M. Brain pericytes as mediators of neuroinflammation. Trends pharmacolo. Sci. 2017, 38, 291–304. [Google Scholar] [CrossRef]
- Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.H.; Laramy, J.K. Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro-Oncol. 2018, 20, 184–191. [Google Scholar] [CrossRef]
- Nguyen, M.D.; Julien, J.-P.; Rivest, S. Innate immunity: The missing link in neuroprotection and neurodegeneration? Nat. Rev. Neurosci. 2002, 3, 216–227. [Google Scholar] [CrossRef] [PubMed]
- Lowther, D.E.; Hafler, D.A. Regulatory T cells in the central nervous system. Immunol. Rev. 2012, 248, 156–169. [Google Scholar] [CrossRef] [PubMed]
- Reardon, D.A.; Wucherpfennig, K.; Chiocca, E.A. Immunotherapy for glioblastoma: On the sidelines or in the game? Discov. Med. 2017, 24, 201–208. [Google Scholar] [PubMed]
- Woroniecka, K.; Chongsathidkiet, P.; Rhodin, K.; Kemeny, H.; Dechant, C.; Farber, S.H.; Elsamadicy, A.A.; Cui, X.; Koyama, S.; Jackson, C. T-Cell Exhaustion Signatures Vary with Tumor Type and Are Severe in GlioblastomaT-Cell Exhaustion Signatures in Glioblastoma. Clin. Cancer Res. 2018, 24, 4175–4186. [Google Scholar] [CrossRef] [Green Version]
- Sharpe, A.H.; Pauken, K.E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 2018, 18, 153–167. [Google Scholar] [CrossRef]
- Louveau, A.; Plog, B.A.; Antila, S.; Alitalo, K.; Nedergaard, M.; Kipnis, J. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J. Clin. Invest. 2017, 127, 3210–3219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef] [Green Version]
- Jackson, C.M.; Lim, M.; Drake, C.G. Immunotherapy for Brain Cancer: Recent Progress and Future PromiseImmunotherapy for Brain Cancer. Clin. Cancer Res. 2014, 20, 3651–3659. [Google Scholar] [CrossRef] [Green Version]
- Johnson, D.B.; Sullivan, R.J.; Menzies, A.M. Immune checkpoint inhibitors in challenging populations. Cancer 2017, 123, 1904–1911. [Google Scholar] [CrossRef] [Green Version]
- Tomitaka, A.; Kaushik, A.; Kevadiya, B.D.; Mukadam, I.; Gendelman, H.E.; Khalili, K.; Liu, G.; Nair, M. Surface-engineered multimodal magnetic nanoparticles to manage CNS diseases. Drug Discov. Today 2019, 24, 873–882. [Google Scholar] [CrossRef]
- Chakroun, R.W.; Zhang, P.; Lin, R.; Schiapparelli, P.; Quinones-Hinojosa, A.; Cui, H. Nanotherapeutic systems for local treatment of brain tumors. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2018, 10, e1479. [Google Scholar] [CrossRef]
- Rauf, M.A.; Rehman, F.U.; Zheng, M.; Shi, B. The strategies of nanomaterials for traversing blood-brain barrier. In Nanomedicine in Brain Diseases; Springer: Berlin/Heidelberg, Germany, 2019; pp. 29–57. [Google Scholar]
- Lonser, R.R.; Sarntinoranont, M.; Morrison, P.F.; Oldfield, E.H. Convection-enhanced delivery to the central nervous system. J. Neurosurg. 2015, 122, 697–706. [Google Scholar] [CrossRef] [Green Version]
- Zhao, M.; van Straten, D.; Broekman, M.L.; Préat, V.; Schiffelers, R.M. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics 2020, 10, 1355. [Google Scholar] [CrossRef]
- Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar] [CrossRef]
- Banks, W.A. From blood–brain barrier to blood–brain interface: New opportunities for CNS drug delivery. Nat. Rev. Drug Discov. 2016, 15, 275–292. [Google Scholar] [CrossRef]
- Oller-Salvia, B.; Sánchez-Navarro, M.; Giralt, E.; Teixidó, M. Blood–brain barrier shuttle peptides: An emerging paradigm for brain delivery. Chem. Soc. Rev. 2016, 45, 4690–4707. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Liu, L. Modern methods for delivery of drugs across the blood–brain barrier. Adv. Drug Deliv. Rev. 2012, 64, 640–665. [Google Scholar] [CrossRef]
- Ali, I.U.; . Chen, X. Penetrating the blood–brain barrier: Promise of novel nanoplatforms and delivery vehicles. ACS Nano 2015, 9, 9470–9474. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Zou, Y.; Feng, C.; Lee, A.; Yin, J.; Chung, R.; Park, J.B.; Rizos, H.; Tao, W.; Zheng, M. Charge conversional biomimetic nanocomplexes as a multifunctional platform for boosting orthotopic glioblastoma RNAi therapy. Nano Lett. 2020, 20, 1637–1646. [Google Scholar] [CrossRef]
- He, W.; Zou, Y.; Zheng, M.; Shi, B. Cell-derived biomimetic drug delivery systems for cancer therapy. Sci. Sin. Chim. 2019, 49, 1203–1212. [Google Scholar]
- Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 1–33. [Google Scholar] [CrossRef] [Green Version]
- Rubinsztein, D.C. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 2006, 443, 780–786. [Google Scholar] [CrossRef] [PubMed]
- Marsh, J.L.; Lukacsovich, T.; Thompson, L.M. Animal models of polyglutamine diseases and therapeutic approaches. J. Biol. Chem. 2009, 284, 7431–7435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mukherjee, S.; Madamsetty, V.S.; Bhattacharya, D.; Roy Chowdhury, S.; Paul, M.K.; Mukherjee, A. Recent advancements of nanomedicine in neurodegenerative disorders theranostics. Adv. Funct. Mater. 2020, 30, 2003054. [Google Scholar] [CrossRef]
- Caccamo, D.; Currò, M.; Condello, S.; Ferlazzo, N.; Ientile, R. Critical role of transglutaminase and other stress proteins during neurodegenerative processes. Amino Acids 2010, 38, 653–658. [Google Scholar] [CrossRef]
- Aderibigbe, B.; Naki, T.; Owonubi, S. Graphene-Based Materials for Brain Targeting. In Handbook of Graphene Set, I-VIII; Wiley: Hoboken, NJ, USA, 2019; 225p. [Google Scholar]
- Ellermann, C.; Coenen, A.; Niehues, P.; Leitz, P.; Kochhäuser, S.; Dechering, D.G.; Fehr, M.; Eckardt, L.; Frommeyer, G. Proarrhythmic Effect of Acetylcholine-Esterase Inhibitors Used in the Treatment of Alzheimer’s Disease: Benefit of Rivastigmine in an Experimental Whole-Heart Model. Cardiovasc. Toxicol. 2020, 20, 168–175. [Google Scholar] [CrossRef] [PubMed]
- Schenkman, M.; Moore, C.G.; Kohrt, W.M.; Hall, D.A.; Delitto, A.; Comella, C.L.; Josbeno, D.A.; Christiansen, C.L.; Berman, B.D.; Kluger, B.M. Effect of high-intensity treadmill exercise on motor symptoms in patients with de novo Parkinson disease: A phase 2 randomized clinical trial. JAMA Neurol. 2018, 75, 219–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carradori, D.; Balducci, C.; Re, F.; Brambilla, D.; Le Droumaguet, B.; Flores, O.; Gaudin, A.; Mura, S.; Forloni, G.; Ordoñez-Gutierrez, L. Antibody-functionalized polymer nanoparticle leading to memory recovery in Alzheimer’s disease-like transgenic mouse model. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 609–618. [Google Scholar] [CrossRef]
- Sahebjam, S.; Sharabi, A.; Lim, M.; Kesarwani, P.; Chinnaiyan, P. Immunotherapy and radiation in glioblastoma. J. Neuro-Oncol. 2017, 134, 531–539. [Google Scholar] [CrossRef]
- Louveau, A.; Harris, T.H.; Kipnis, J. Revisiting the mechanisms of CNS immune privilege. Trends Immunol. 2015, 36, 569–577. [Google Scholar] [CrossRef] [Green Version]
- Iliff, J.J.; Wang, M.; Liao, Y.; Plogg, B.A.; Peng, W.; Gundersen, G.A.; Benveniste, H.; Vates, G.E.; Deane, R.; Goldman, S.A. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 2012, 4, ra111–ra147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aspelund, A.; Antila, S.; Proulx, S.T.; Karlsen, T.V.; Karaman, S.; Detmar, M.; Wiig, H.; Alitalo, K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 2015, 212, 991–999. [Google Scholar] [CrossRef] [PubMed]
- Ramos, M.; Bechet, N.B.; Battistella, R.; Pavan, C.; Xavier, A.L.; Nedergaard, M.; Lundgaard, I. Cisterna magna injection in rats to study glymphatic function. In Astrocytes; Springer: Berlin/Heidelberg, Germany, 2019; pp. 97–104. [Google Scholar]
- Morris, A.W.; Sharp, M.M.; Albargothy, N.J.; Fernandes, R.; Hawkes, C.A.; Verma, A.; Weller, R.O.; Carare, R.O. Vascular basement membranes as pathways for the passage of fluid into and out of the brain. Acta Neuropathol. 2016, 131, 725–736. [Google Scholar] [CrossRef] [Green Version]
- Thrane, V.R.; Thrane, A.S.; Plog, B.A.; Thiyagarajan, M.; Iliff, J.J.; Deane, R.; Nagelhus, E.A.; Nedergaard, M. Paravascular microcirculation facilitates rapid lipid transport and astrocyte signaling in the brain. Sci. Rep. 2013, 3, 2582. [Google Scholar] [CrossRef]
- Medawar, P. Immunity to homologous grafted skin. II. The relationship between the antigens of blood and skin. Br. J. Exp. Pathol. 1946, 27, 15. [Google Scholar]
- Shechter, R.; London, A.; Schwartz, M. Orchestrated leukocyte recruitment to immune-privileged sites: Absolute barriers versus educational gates. Nat. Rev. Immunol. 2013, 13, 206–218. [Google Scholar] [CrossRef]
- Gutkin, A.; Cohen, Z.R.; Peer, D. Harnessing nanomedicine for therapeutic intervention in glioblastoma. Expert Opin. Drug Deliv. 2016, 13, 1573–1582. [Google Scholar] [CrossRef]
- Birks, J.S.; Evans, J.G. Rivastigmine for Alzheimer’s disease. Cochrane Database Syst. Rev. 2015, 4. [Google Scholar] [CrossRef]
- Furtado, D.; Björnmalm, M.; Ayton, S.; Bush, A.I.; Kempe, K.; Caruso, F. Overcoming the blood–brain barrier: The role of nanomaterials in treating neurological diseases. Adv. Mater. 2018, 30, 1801362. [Google Scholar] [CrossRef] [Green Version]
- Erickson, M.A.; Banks, W.A. Blood–brain barrier dysfunction as a cause and consequence of Alzheimer’s disease. J. Cereb. Blood Flow Metab. 2013, 33, 1500–1513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van Assema, D.M.; Lubberink, M.; Boellaard, R.; Schuit, R.C.; Windhorst, A.D.; Scheltens, P.; Lammertsma, A.A.; van Berckel, B.N. P-glycoprotein function at the blood–brain barrier: Effects of age and gender. Mol. Imaging Biol. 2012, 14, 771–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernández, T.; Martínez-Serrano, A.; Cussó, L.; Desco, M.; Ramos-Gómez, M. Functionalization and characterization of magnetic nanoparticles for the detection of ferritin accumulation in Alzheimer’s disease. ACS Chem. Neurosci. 2018, 9, 912–924. [Google Scholar] [CrossRef] [PubMed]
- Niu, X.; Chen, J.; Gao, J. Nanocarriers as a powerful vehicle to overcome blood-brain barrier in treating neurodegenerative diseases: Focus on recent advances. Asian J. Pharm. Sci. 2019, 14, 480–496. [Google Scholar] [CrossRef]
- Sharma, U.; Badyal, P.N.; Gupta, S. Polymeric nanoparticles drug delivery to brain: A review. Int. J. Pharmacol. 2015, 2, 60–69. [Google Scholar]
- Krol, S.; Macrez, R.; Docagne, F.; Defer, G.; Laurent, S.; Rahman, M.; Hajipour, M.J.; Kehoe, P.G.; Mahmoudi, M. Therapeutic benefits from nanoparticles: The potential significance of nanoscience in diseases with compromise to the blood brain barrier. Chem. Rev. 2012, 113, 1877–1903. [Google Scholar] [CrossRef] [PubMed]
- Carpentier, A.F.; Meng, Y. Recent advances in immunotherapy for human glioma. Curr. Opin. Oncol. 2006, 18, 631–636. [Google Scholar] [CrossRef]
- He, Q.; Liu, J.; Liang, J.; Liu, X.; Li, W.; Liu, Z.; Ding, Z.; Tuo, D. Towards improvements for penetrating the blood–brain barrier—recent progress from a material and pharmaceutical perspective. Cells 2018, 7, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Damkier, H.H.; Brown, P.D.; Praetorius, J. Cerebrospinal fluid secretion by the choroid plexus. Physiolo. Rev. 2013, 93, 1847–1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strazielle, N.; Ghersi-Egea, J.-F. Potential pathways for CNS drug delivery across the blood-cerebrospinal fluid barrier. Curr. Pharm. Des. 2016, 22, 5463–5476. [Google Scholar] [CrossRef] [Green Version]
- Xu, D.-H.; Yan, M.; Fang, P.-F.; Liu, Y.-W. Influence of P-glycoprotein on brucine transport at the in vitro blood–brain barrier. Eur. J. Pharmacol. 2012, 690, 68–76. [Google Scholar] [CrossRef]
- Machtoub, L.; Kasugai, Y. Amyotrophic Lateral Sclerosis: Advances and Perspectives of Neuronanomedicine; CRC Press: Boca Raton, FL, USA, 2016. [Google Scholar]
- Rosenberg, S.A.; Dudley, M.E.; Restifo, N.P. Cancer immunotherapy. N. Engl. J. Med. 2008, 359, 1072. [Google Scholar]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107. [Google Scholar] [CrossRef]
- Lin, Y.; Okada, H. Cellular immunotherapy for malignant gliomas. Expert Opin. Biol. Ther. 2016, 16, 1265–1275. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.-L.; Fan, C.-H.; Ting, C.-Y.; Yeh, C.-K. Combining Microbubbles and Ultrasound for Drug Delivery to Brain Tumors: Current Progress and Overview. Theranostics 2014, 4, 432–444. [Google Scholar] [CrossRef] [PubMed]
- Klafki, H.-W.; Staufenbiel, M.; Kornhuber, J.; Wiltfang, J. Therapeutic approaches to Alzheimer’s disease. Brain 2006, 129, 2840–2855. [Google Scholar] [CrossRef] [PubMed]
- Saraiva, C.; Praça, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. J. Control. Release 2016, 235, 34–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salvati, A.; Pitek, A.S.; Monopoli, M.P.; Prapainop, K.; Bombelli, F.B.; Hristov, D.R.; Kelly, P.M.; Åberg, C.; Mahon, E.; Dawson, K.A. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nature Nanotechnol. 2013, 8, 137–143. [Google Scholar] [CrossRef] [PubMed]
- Hadjipanayis, C.G.; Machaidze, R.; Kaluzova, M.; Wang, L.; Schuette, A.J.; Chen, H.; Wu, X.; Mao, H. EGFRvIII Antibody–Conjugated Iron Oxide Nanoparticles for Magnetic Resonance Imaging–Guided Convection-Enhanced Delivery and Targeted Therapy of GlioblastomaEGFRvIII-Targeted Therapy of GBM by IONPs after CED. Cancer Res. 2010, 70, 6303–6312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- MacDonald, P.A.; MacDonald, A.A.; Seergobin, K.N.; Tamjeedi, R.; Ganjavi, H.; Provost, J.-S.; Monchi, O. The effect of dopamine therapy on ventral and dorsal striatum-mediated cognition in Parkinson’s disease: Support from functional MRI. Brain 2011, 134, 1447–1463. [Google Scholar] [CrossRef] [Green Version]
- Kulkarni, A.D.; Vanjari, Y.H.; Sancheti, K.H.; Belgamwar, V.S.; Surana, S.J.; Pardeshi, C.V. Nanotechnology-mediated nose to brain drug delivery for Parkinson’s disease: A mini review. J. Drug Target. 2015, 23, 775–788. [Google Scholar] [CrossRef] [PubMed]
- Azeem, A.; Talegaonkar, S.; Negi, L.M.; Ahmad, F.J.; Khar, R.K.; Iqbal, Z. Oil based nanocarrier system for transdermal delivery of ropinirole: A mechanistic, pharmacokinetic and biochemical investigation. Int. J. Pharm. 2012, 422, 436–444. [Google Scholar] [CrossRef]
- Md, S.; Khan, R.A.; Mustafa, G.; Chuttani, K.; Baboota, S.; Sahni, J.K.; Ali, J. Bromocriptine loaded chitosan nanoparticles intended for direct nose to brain delivery: Pharmacodynamic, Pharmacokinetic and Scintigraphy study in mice model. Eur. J. Pharm. Sci. 2013, 48, 393–405. [Google Scholar] [CrossRef]
- Gendelman, H.E.; Anantharam, V.; Bronich, T.; Ghaisas, S.; Jin, H.; Kanthasamy, A.G.; Liu, X.; McMillan, J.; Mosley, R.L.; Narasimhan, B.; et al. Nanoneuromedicines for degenerative, inflammatory, and infectious nervous system diseases. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 751–767. [Google Scholar] [CrossRef] [Green Version]
- Huang, R.; Ke, W.; Liu, Y.; Wu, D.; Feng, L.; Jiang, C.; Pei, Y. Gene therapy using lactoferrin-modified nanoparticles in a rotenone-induced chronic Parkinson model. J. Neurol. Sci. 2010, 290, 123–130. [Google Scholar] [CrossRef]
- Konishi, M.; Kawamoto, K.; Izumikawa, M.; Kuriyama, H.; Yamashita, T. Gene transfer into guinea pig cochlea using adeno-associated virus vectors. J. Gene Med. 2008, 10, 610–618. [Google Scholar] [CrossRef]
- Davis, S.S. Biomedical applications of nanotechnology—Implications for drug targeting and gene therapy. Trends Biotechnol. 1997, 15, 217–224. [Google Scholar] [CrossRef] [PubMed]
- Yurek, D.M.; Flectcher, A.M.; Kowalczyk, T.H.; Padegimas, L.; Cooper, M.J. Compacted DNA nanoparticle gene transfer of GDNF to the rat striatum enhances the survival of grafted fetal dopamine neurons. Cell Transplant. 2009, 18, 1183–1196. [Google Scholar] [CrossRef] [PubMed]
- Wong, H.L.; Wu, X.Y.; Bendayan, R. Nanotechnological advances for the delivery of CNS therapeutics. Adv. Drug Deliv. Rev. 2012, 64, 686–700. [Google Scholar] [CrossRef] [PubMed]
- Winer, J.L.; Kim, P.E.; Law, M.; Liu, C.Y.; Apuzzo, M.L. Visualizing the future: Enhancing neuroimaging with nanotechnology. World Neurosurg. 2011, 75, 626–637. [Google Scholar] [CrossRef]
- Kelkar, S.S.; Reineke, T.M. Theranostics: Combining imaging and therapy. Bioconjugate Chem. 2011, 22, 1879–1903. [Google Scholar] [CrossRef] [PubMed]
- Downs, M.E.; Buch, A.; Karakatsani, M.E.; Konofagou, E.E.; Ferrera, V.P. Blood-brain barrier opening in behaving non-human primates via focused ultrasound with systemically administered microbubbles. Sci. Rep. 2015, 5, 15076. [Google Scholar] [CrossRef] [Green Version]
- Yavarpour-Bali, H.; Ghasemi-Kasman, M.; Pirzadeh, M. Curcumin-loaded nanoparticles: A novel therapeutic strategy in treatment of central nervous system disorders. Int. J. Nanomed. 2019, 14, 4449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, F.; Lin, Y.-A.; Kannan, S.; Kannan, R.M. Targeting specific cells in the brain with nanomedicines for CNS therapies. J. Control. Release 2016, 240, 212–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harilal, S.; Jose, J.; Parambi, D.G.T.; Kumar, R.; Mathew, G.E.; Uddin, M.S.; Kim, H.; Mathew, B. Advancements in nanotherapeutics for Alzheimer’s disease: Current perspectives. J. Pharm. Pharmacol. 2019, 71, 1370–1383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, Y.J.; Cutler, E.G.; Cho, H. Therapeutic nanoplatforms and delivery strategies for neurological disorders. Nano Converg. 2018, 5, 1–15. [Google Scholar] [CrossRef]
- Singh, A.P.; Biswas, A.; Shukla, A.; Maiti, P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal Transduct. Target. Ther. 2019, 4, 33. [Google Scholar] [CrossRef] [Green Version]
- Xie, J.; Shen, Z.; Anraku, Y.; Kataoka, K.; Chen, X. Nanomaterial-based blood-brain-barrier (BBB) crossing strategies. Biomaterials 2019, 224, 119491. [Google Scholar] [CrossRef]
- Kreuter, J. Drug delivery to the central nervous system by polymeric nanoparticles: What do we know? Adv. Drug Deliv. Rev. 2014, 71, 2–14. [Google Scholar] [CrossRef]
- Duncan, R.; Vicent, M.J. Polymer therapeutics-prospects for 21st century: The end of the beginning. Adv. Drug Deliv. Rev. 2013, 65, 60–70. [Google Scholar] [CrossRef]
- Kolb-Mäurer, A.; Sunderkötter, C.; Kukowski, B.; Meuth, S.G. An update on Peginterferon beta-1a Management in Multiple Sclerosis: Results from an interdisciplinary Board of German and Austrian Neurologists and dermatologists. BMC Neurol. 2019, 19, 130. [Google Scholar] [CrossRef] [Green Version]
- Robinson, A.P.; Zhang, J.Z.; Titus, H.E.; Karl, M.; Merzliakov, M.; Dorfman, A.R.; Karlik, S.; Stewart, M.G.; Watt, R.K.; Facer, B.D.; et al. Nanocatalytic activity of clean-surfaced, faceted nanocrystalline gold enhances remyelination in animal models of multiple sclerosis. Sci. Rep. 2020, 10, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cummings, J.; Lee, G.; Ritter, A.; Sabbagh, M.; Zhong, K. Alzheimer’s disease drug development pipeline: 2019. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2019, 5, 272–293. [Google Scholar] [CrossRef] [PubMed]
- Dolati, S.; Ahmadi, M.; Aghebti-Maleki, L.; Nikmaram, A.; Marofi, F.; Rikhtegar, R.; Ayromlou, H.; Yousefi, M. Nanocurcumin is a potential novel therapy for multiple sclerosis by influencing inflammatory mediators. Pharmacol. Rep. 2018, 70, 1158–1167. [Google Scholar] [CrossRef]
- Ahmadi, M.; Agah, E.; Nafissi, S.; Jaafari, M.R.; Harirchian, M.H.; Sarraf, P.; Faghihi-Kashani, S.; Hosseini, S.J.; Ghoreishi, A.; Aghamollaii, V. Safety and efficacy of nanocurcumin as add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: A pilot randomized clinical trial. Neurotherapeutics 2018, 15, 430–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Zhai, M.; Chen, Z.; Han, X.; Yu, F.; Li, Z.; Xie, X.; Han, C.; Yu, L.; Yang, Y. Dual-modified liposome codelivery of doxorubicin and vincristine improve targeting and therapeutic efficacy of glioma. Drug Deliv. 2017, 24, 1045–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madhankumar, A.; Slagle-Webb, B.; Mintz, A.; Sheehan, J.M.; Connor, J.R. Interleukin-13 receptor–targeted nanovesicles are a potential therapy for glioblastoma multiforme. Mol. Cancer Ther. 2006, 5, 3162–3169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahmud, H.; Kasai, T.; Khayrani, A.C.; Asakura, M.; Oo, A.K.K.; Du, J.; Vaidyanath, A.; El-Ghlban, S.; Mizutani, A.; Seno, A. Targeting glioblastoma cells expressing CD44 with liposomes encapsulating doxorubicin and displaying chlorotoxin-IgG Fc fusion protein. Int. J. Mol. Sci. 2018, 19, 659. [Google Scholar] [CrossRef]
- Jain, N.K.; Rana, A.; Jain, S.K. Brain drug delivery system bearing dopamine hydrochloride for effective management of parkinsonism. Drug Dev. Ind. Pharm. 1998, 24, 671–675. [Google Scholar] [CrossRef]
- Xiang, Y.; Wu, Q.; Liang, L.; Wang, X.; Wang, J.; Zhang, X.; Pu, X.; Zhang, Q. Chlorotoxin-modified stealth liposomes encapsulating levodopa for the targeting delivery against the Parkinson’s disease in the MPTP-induced mice model. J. Drug Target. 2012, 20, 67–75. [Google Scholar] [CrossRef]
- Sun, D.; Zhuang, X.; Zhang, S.; Deng, Z.-B.; Grizzle, W.; Miller, D.; Zhang, H.-G. Exosomes are endogenous nanoparticles that can deliver biological information between cells. Adv. Drug Deliv. Rev. 2013, 65, 342–347. [Google Scholar] [CrossRef]
- Dixit, S.; Novak, T.; Miller, K.; Zhu, Y.; Kenney, M.E.; Broome, A.-M. Transferrin receptor-targeted theranostic gold nanoparticles for photosensitizer delivery in brain tumors. Nanoscale 2015, 7, 1782–1790. [Google Scholar] [CrossRef]
- Lee, C.; Hwang, H.S.; Lee, S.; Kim, B.; Kim, J.O.; Oh, K.T.; Lee, E.S.; Choi, H.G.; Youn, Y.S. Rabies virus-inspired silica-coated gold nanorods as a photothermal therapeutic platform for treating brain tumors. Adv. Mater. 2017, 29, 1605563. [Google Scholar] [CrossRef] [PubMed]
- Hirschberg, H.; Madsen, S.J. Cell mediated photothermal therapy of brain tumors. J. Neuroimmune Pharmacol. 2017, 12, 99–106. [Google Scholar] [CrossRef]
- Hu, K.; Chen, X.; Chen, W.; Zhang, L.; Li, J.; Ye, J.; Zhang, Y.; Zhang, L.; Li, C.-H.; Yin, L. Neuroprotective effect of gold nanoparticles composites in Parkinson’s disease model. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1123–1136. [Google Scholar] [CrossRef] [PubMed]
- Gao, N.; Sun, H.; Dong, K.; Ren, J.; Qu, X. Gold-Nanoparticle-Based Multifunctional Amyloid-β Inhibitor against Alzheimer’s Disease. Chem.–A Eur. J. 2015, 21, 829–835. [Google Scholar] [CrossRef] [PubMed]
- Gao, H.; Qian, J.; Cao, S.; Yang, Z.; Pang, Z.; Pan, S.; Fan, L.; Xi, Z.; Jiang, X.; Zhang, Q. Precise glioma targeting of and penetration by aptamer and peptide dual-functioned nanoparticles. Biomaterials 2012, 33, 5115–5123. [Google Scholar] [CrossRef] [PubMed]
- Guo, J.; Gao, X.; Su, L.; Xia, H.; Gu, G.; Pang, Z.; Jiang, X.; Yao, L.; Chen, J.; Chen, H. Aptamer-functionalized PEG–PLGA nanoparticles for enhanced anti-glioma drug delivery. Biomaterials 2011, 32, 8010–8020. [Google Scholar] [CrossRef] [PubMed]
- Shah, N.; Chaudhari, K.; Dantuluri, P.; Murthy, R.; Das, S. Paclitaxel-loaded PLGA nanoparticles surface modified with transferrin and Pluronic® P85, an in vitro cell line and in vivo biodistribution studies on rat model. J. Drug Target. 2009, 17, 533–542. [Google Scholar] [CrossRef]
- Wang, B.; Lv, L.; Wang, Z.; Jiang, Y.; Lv, W.; Liu, X.; Wang, Z.; Zhao, Y.; Xin, H.; Xu, Q. Improved anti-glioblastoma efficacy by IL-13Rα2 mediated copolymer nanoparticles loaded with paclitaxel. Sci. Rep. 2015, 5, 16589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xin, H.; Jiang, X.; Gu, J.; Sha, X.; Chen, L.; Law, K.; Chen, Y.; Wang, X.; Jiang, Y.; Fang, X. Angiopep-conjugated poly (ethylene glycol)-co-poly (ε-caprolactone) nanoparticles as dual-targeting drug delivery system for brain glioma. Biomaterials 2011, 32, 4293–4305. [Google Scholar] [CrossRef] [PubMed]
- Jafarieh, O.; Md, S.; Ali, M.; Baboota, S.; Sahni, J.; Kumari, B.; Bhatnagar, A.; Ali, J. Design, characterization, and evaluation of intranasal delivery of ropinirole-loaded mucoadhesive nanoparticles for brain targeting. Drug Dev. Ind. Pharm. 2015, 41, 1674–1681. [Google Scholar] [CrossRef]
- Gambaryan, P.; Kondrasheva, I.; Severin, E.; Guseva, A.; Kamensky, A. Increasing the Efficiency of Parkinson’s Disease Treatment Using a poly (lactic-co-glycolic acid)(PLGA) Based L-DOPA Delivery System. Exp. Neurobiol. 2014, 23, 246148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tiwari, M.N.; Agarwal, S.; Bhatnagar, P.; Singhal, N.K.; Tiwari, S.K.; Kumar, P.; Chauhan, L.K.S.; Patel, D.K.; Chaturvedi, R.K.; Singh, M.P. Nicotine-encapsulated poly (lactic-co-glycolic) acid nanoparticles improve neuroprotective efficacy against MPTP-induced parkinsonism. Free Radic. Biol. Med. 2013, 65, 704–718. [Google Scholar] [CrossRef]
- Hasadsri, L.; Kreuter, J.; Hattori, H.; Iwasaki, T.; George, J.M. Functional protein delivery into neurons using polymeric nanoparticles. J. Biol. Chem. 2009, 284, 6972–6981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mulik, R.S.; Monkkonen, J.; Juvonen, R.O.; Mahadik, K.R.; Paradkar, A.R. ApoE3 mediated poly (butyl) cyanoacrylate nanoparticles containing curcumin: Study of enhanced activity of curcumin against beta amyloid induced cytotoxicity using in vitro cell culture model. Mol. Pharm. 2010, 7, 815–825. [Google Scholar] [CrossRef] [PubMed]
- Kulkarni, P.V.; Roney, C.A.; Antich, P.P.; Bonte, F.J.; Raghu, A.V.; Aminabhavi, T.M. Quinoline-n-butylcyanoacrylate-based nanoparticles for brain targeting for the diagnosis of Alzheimer’s disease. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2010, 2, 35–47. [Google Scholar] [CrossRef]
- Meng, Q.; Wang, A.; Hua, H.; Jiang, Y.; Wang, Y.; Mu, H.; Wu, Z.; Sun, K. Intranasal delivery of Huperzine A to the brain using lactoferrin-conjugated N-trimethylated chitosan surface-modified PLGA nanoparticles for treatment of Alzheimer’s disease. Int. J. Nanomed. 2018, 13, 705. [Google Scholar] [CrossRef]
- Garcion, E.; Lamprecht, A.; Heurtault, B.; Paillard, A.; Aubert-Pouessel, A.; Denizot, B.; Menei, P.; Benoît, J.P. A new generation of anticancer, drug-loaded, colloidal vectors reverses multidrug resistance in glioma and reduces tumor progression in rats. Mol. Cancer Therap. 2006, 5, : 1710–1722. [Google Scholar] [CrossRef] [Green Version]
- Sibov, T.T.; Pavon, L.F.; Miyaki, L.A.; Mamani, J.B.; Nucci, L.P.; Alvarim, L.T.; Silveira, P.H.; Marti, L.C.; Gamarra, L. Umbilical cord mesenchymal stem cells labeled with multimodal iron oxide nanoparticles with fluorescent and magnetic properties: Application for in vivo cell tracking. Int. J. Nanomed. 2014, 9, 337. [Google Scholar]
- Sillerud, L.O.; Solberg, N.O.; Chamberlain, R.; Orlando, R.A.; Heidrich, J.E.; Brown, D.C.; Brady, C.I.; Vander Jagt, T.A.; Garwood, M.; Vander Jagt, D.L. SPION-enhanced magnetic resonance imaging of Alzheimer’s disease plaques in AβPP/PS-1 transgenic mouse brain. J. Alzheimer’s Dis. 2013, 34, 349–365. [Google Scholar] [CrossRef] [Green Version]
- Pardridge, W.M. The blood-brain barrier: Bottleneck in brain drug development. NeuroRx 2005, 2, 3–14. [Google Scholar] [CrossRef] [PubMed]
- Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef] [Green Version]
- Riemenschneider, M.; Wagenpfeil, S.; Vanderstichele, H.; Otto, M.; Wiltfang, J.; Kretzschmar, H.; Vanmechelen, E.; Förstl, H.; Kurz, A. Phospho-tau/total tau ratio in cerebrospinal fluid discriminates Creutzfeldt–Jakob disease from other dementias. Mol. Psychiatry 2003, 8, 343. [Google Scholar] [CrossRef] [PubMed]
- Lopes, M.B.S. The 2017 World Health Organization classification of tumors of the pituitary gland: A summary. Acta Neuropathol. 2017, 134, 521–535. [Google Scholar] [CrossRef]
- Louis, D.N.; Perry, A.; Reifenberger, G.; Von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, P.Y.; Huse, J.T. 2016 World Health Organization classification of central nervous system tumors. Contin. Lifelong Learn. Neurol. 2017, 23, 1531–1547. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Li, C.; Li, Y.; Yi, X.; Wang, R.; Lee, S.M.-Y.; Zheng, Y. Small-sized mPEG–PLGA nanoparticles of Schisantherin A with sustained release for enhanced brain uptake and anti-parkinsonian activity. ACS Appl. Mater. Interfaces 2017, 9, 9516–9527. [Google Scholar] [CrossRef] [PubMed]
- Gao, K.; Jiang, X. Influence of particle size on transport of methotrexate across blood brain barrier by polysorbate 80-coated polybutylcyanoacrylate nanoparticles. Int. J. Pharm. 2006, 310, 213–219. [Google Scholar] [CrossRef]
- Lamprecht, A.; Benoit, J.-P. Etoposide nanocarriers suppress glioma cell growth by intracellular drug delivery and simultaneous P-glycoprotein inhibition. J. Control. Release 2006, 112, 208–213. [Google Scholar] [CrossRef] [PubMed]
- Agrawal, P.; Singh, R.P.; Kumari, L.; Sharma, G.; Koch, B.; Rajesh, C.V.; Mehata, A.K.; Singh, S.; Pandey, B.L.; Muthu, M.S. TPGS-chitosan cross-linked targeted nanoparticles for effective brain cancer therapy. Mater. Sci. Eng. C 2017, 74, 167–176. [Google Scholar] [CrossRef] [PubMed]
- Agrawal, P.; Singh, R.P.; Sharma, G.; Mehata, A.K.; Singh, S.; Rajesh, C.V.; Pandey, B.L.; Koch, B.; Muthu, M.S. Bioadhesive micelles of d-α-tocopherol polyethylene glycol succinate 1000: Synergism of chitosan and transferrin in targeted drug delivery. Colloids Surf. B: Biointerfaces 2017, 152, 277–288. [Google Scholar] [CrossRef] [PubMed]
- Dhanikula, R.S.; Argaw, A.; Bouchard, J.-F.; Hildgen, P. Methotrexate loaded polyether-copolyester dendrimers for the treatment of gliomas: Enhanced efficacy and intratumoral transport capability. Mol. Pharm. 2008, 5, 105–116. [Google Scholar] [CrossRef]
- He, H.; Li, Y.; Jia, X.-R.; Du, J.; Ying, X.; Lu, W.-L.; Lou, J.-N.; Wei, Y. PEGylated Poly (amidoamine) dendrimer-based dual-targeting carrier for treating brain tumors. Biomaterials 2011, 32, 478–487. [Google Scholar] [CrossRef] [PubMed]
- Hernández-Pedro, N.Y.; Rangel-López, E.; Magaña-Maldonado, R.; de la Cruz, V.P.; Santamaría del Angel, A.; Pineda, B.; Sotelo, J. Application of nanoparticles on diagnosis and therapy in gliomas. BioMed Res. Int. 2013, 2013, 351031. [Google Scholar] [CrossRef] [Green Version]
- Brigger, I.; Morizet, J.; Aubert, G.; Chacun, H.; Terrier-Lacombe, M.-J.; Couvreur, P.; Vassal, G. Poly (ethylene glycol)-coated hexadecylcyanoacrylate nanospheres display a combined effect for brain tumor targeting. J. Pharmacol. Exp. Ther. 2002, 303, 928–936. [Google Scholar] [CrossRef] [Green Version]
- Berendsen, S.; Broekman, M.; Seute, T.; Snijders, T.; van Es, C.; de Vos, F.; Regli, L.; Robe, P. Valproic acid for the treatment of malignant gliomas: Review of the preclinical rationale and published clinical results. Expert Opin. Investig. Drugs 2012, 21, 1391–1415. [Google Scholar] [CrossRef] [PubMed]
- Spratt, D.E.; Folkert, M.; Zumsteg, Z.S.; Chan, T.A.; Beal, K.; Gutin, P.H.; Pentsova, E.; Yamada, Y. Temporal relationship of post-operative radiotherapy with temozolomide and oncologic outcome for glioblastoma. J. Neuro-Oncol. 2014, 116, 357–363. [Google Scholar] [CrossRef]
- Prah, M.; Stufflebeam, S.; Paulson, E.; Kalpathy-Cramer, J.; Gerstner, E.; Batchelor, T.; Barboriak, D.; Rosen, B.; Schmainda, K. Repeatability of standardized and normalized relative CBV in patients with newly diagnosed glioblastoma. Am. J. Neuroradiol. 2015, 36, 1654–1661. [Google Scholar] [CrossRef] [PubMed]
- Norden, A.D.; Drappatz, J.; Wen, P.Y. Antiangiogenic therapies for high-grade glioma. Nat. Rev. Neurol. 2009, 5, 610. [Google Scholar] [CrossRef]
- Schweitzer, T.; Vince, G.; Herbold, C.; Roosen, K.; Tonn, J.-C. Extraneural metastases of primary brain tumors. J. Neuro-Oncol. 2001, 53, 107–114. [Google Scholar] [CrossRef] [PubMed]
- Hanssen, A.; Riebensahm, C.; Mohme, M.; Joosse, S.; Velthaus, J.-L.; Berger, L.; Bernreuther, C.; Glatzel, M.; Loges, S.; Lamszus, K. Frequency of Circulating Tumor Cells (CTC) in Patients with Brain Metastases: Implications as a Risk Assessment Marker in Oligo-Metastatic Disease. Cancers 2018, 10, 527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sullivan, J.P.; Nahed, B.V.; Madden, M.W.; Oliveira, S.M.; Springer, S.; Bhere, D.; Chi, A.S.; Wakimoto, H.; Rothenberg, S.M.; Sequist, L.V. Brain tumor cells in circulation are enriched for mesenchymal gene expression. Cancer Discov. 2014, 4, 1299–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker, P.R.; Calzascia, T.; Dietrich, P.Y. All in the head: Obstacles for immune rejection of brain tumours. Immunology 2002, 107, 28–38. [Google Scholar] [CrossRef]
- Engelhardt, B.; Ransohoff, R.M. The ins and outs of T-lymphocyte trafficking to the CNS: Anatomical sites and molecular mechanisms. Trends Immunol. 2005, 26, 485–495. [Google Scholar] [CrossRef] [PubMed]
- Davies, D. Blood–brain barrier breakdown in septic encephalopathy and brain tumours. J. Anat. 2002, 200, 639–646. [Google Scholar] [CrossRef]
- Rascher, G.; Fischmann, A.; Kröger, S.; Duffner, F.; Grote, E.-H.; Wolburg, H. Extracellular matrix and the blood-brain barrier in glioblastoma multiforme: Spatial segregation of tenascin and agrin. Acta Neuropathol. 2002, 104, 85–91. [Google Scholar] [CrossRef]
- Prins, R.M.; Liau, L.M. Cellular immunity and immunotherapy of brain tumors. Front. Biosci. 2004, 9, 3124–3136. [Google Scholar] [CrossRef] [Green Version]
- Fantini, M.C.; Becker, C.; Monteleone, G.; Pallone, F.; Galle, P.R.; Neurath, M.F. Cutting edge: TGF-β induces a regulatory phenotype in CD4+ CD25− T cells through Foxp3 induction and down-regulation of Smad7. J. Immunol. 2004, 172, 5149–5153. [Google Scholar] [CrossRef]
- Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.-K.L.; Flavell, R.A. Transforming growth factor-β regulation of immune responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef]
- Fecci, P.E.; Mitchell, D.A.; Whitesides, J.F.; Xie, W.; Friedman, A.H.; Archer, G.E.; Herndon, J.E.; Bigner, D.D.; Dranoff, G.; Sampson, J.H. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006, 66, 3294–3302. [Google Scholar] [CrossRef] [Green Version]
- Jensen, R.L.; Ragel, B.T.; Whang, K.; Gillespie, D. Inhibition of hypoxia inducible factor-1α (HIF-1α) decreases vascular endothelial growth factor (VEGF) secretion and tumor growth in malignant gliomas. J. Neuro-Oncol. 2006, 78, 233–247. [Google Scholar] [CrossRef] [PubMed]
- Lohr, J.; Ratliff, T.; Huppertz, A.; Ge, Y.; Dictus, C.; Ahmadi, R.; Grau, S.; Hiraoka, N.; Eckstein, V.; Ecker, R.C. Effector T-cell infiltration positively impacts survival of glioblastoma patients and is impaired by tumor-derived TGF-β. Clin. Cancer Res. 2011, 17, 4296–4308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lang, H.; Hu, G.; Chen, Y.; Liu, Y.; Tu, W.; Lu, Y.; Wu, L.; Xu, G. Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur. Rev. Med. Pharm. Sci. 2017, 21, 959–972. [Google Scholar]
- Caffo, M.; Barresi, V.; Caruso, G.; La Fata, G.; Pino, M.A.; Raudino, G.; Alafaci, C.; Tomasello, F. Gliomas Biology: Angiogenesis and Invasion. In Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications; IntechOpen: London, UK, 2013. [Google Scholar]
- Girardi, M.; Oppenheim, D.E.; Steele, C.R.; Lewis, J.M.; Glusac, E.; Filler, R.; Hobby, P.; Sutton, B.; Tigelaar, R.E.; Hayday, A.C. Regulation of cutaneous malignancy by γδ T cells. Science 2001, 294, 605–609. [Google Scholar] [CrossRef]
- Chokshi, C.R.; Brakel, B.A.; Tatari, N.; Savage, N.; Salim, S.K.; Venugopal, C.; Singh, S.K. Advances in immunotherapy for adult glioblastoma. Cancers 2021, 13, 3400. [Google Scholar] [CrossRef]
- Weller, M.; Roth, P.; Preusser, M.; Wick, W.; Reardon, D.A.; Platten, M.; Sampson, J.H. Vaccine-based immunotherapeutic approaches to gliomas and beyond. Nat. Rev. Neurol. 2017, 13, 363–374. [Google Scholar] [CrossRef] [PubMed]
- Chistiakov, D.A.; Chekhonin, I.V.; Chekhonin, V.P. The EGFR variant III mutant as a target for immunotherapy of glioblastoma multiforme. Eur. J. Pharmacol. 2017, 810, 70–82. [Google Scholar] [CrossRef]
- Reardon, D.A.; Schuster, J.; Tran, D.D.; Fink, K.L.; Nabors, L.B.; Li, G.; Bota, D.A.; Lukas, R.V.; Desjardins, A.; Ashby, L.S. ReACT: Overall Survival from a Randomized Phase II Study of Rindopepimut (CDX-110) Plus Bevacizumab in Relapsed Glioblastoma; American Society of Clinical Oncology: Alexandria, VA, USA, 2015. [Google Scholar]
- Kuai, R.; Ochyl, L.J.; Bahjat, K.S.; Schwendeman, A.; Moon, J.J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 2017, 16, 489–496. [Google Scholar] [CrossRef]
- Liu, L.; Wang, Y.; Miao, L.; Liu, Q.; Musetti, S.; Li, J.; Huang, L. Combination immunotherapy of MUC1 mRNA nano-vaccine and CTLA-4 blockade effectively inhibits growth of triple negative breast cancer. Mol. Ther. 2018, 26, 45–55. [Google Scholar] [CrossRef]
- Baratta, M.G. Glioblastoma is ‘hot’for personalized vaccines. Nat. Rev. Cancer 2019, 19, 129. [Google Scholar] [CrossRef]
- Lyon, J.G.; Mokarram, N.; Saxena, T.; Carroll, S.L.; Bellamkonda, R.V. Engineering challenges for brain tumor immunotherapy. Adv. Drug Deliv. Rev. 2017, 114, 19–32. [Google Scholar] [CrossRef] [PubMed]
- Lou, J.; Zhang, L.; Zheng, G. Advancing Cancer Immunotherapies with Nanotechnology. Adv. Ther. 2019, 2, 1800128. [Google Scholar] [CrossRef]
- Mahjub, R.; Jatana, S.; Lee, S.E.; Qin, Z.; Pauli, G.; Soleimani, M.; Madadi, S.; Li, S.-D. Recent advances in applying nanotechnologies for cancer immunotherapy. J. Control. Release 2018, 288, 239–263. [Google Scholar] [CrossRef]
- Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–196. [Google Scholar] [CrossRef]
- Kateb, B.; Van Handel, M.; Zhang, L.; Bronikowski, M.J.; Manohara, H.; Badie, B. Internalization of MWCNTs by microglia: Possible application in immunotherapy of brain tumors. NeuroImage 2007, 37, S9–S17. [Google Scholar] [CrossRef] [PubMed]
- Krishnamachari, Y.; Geary, S.M.; Lemke, C.D.; Salem, A.K. Nanoparticle delivery systems in cancer vaccines. Pharm. Res. 2011, 28, 215–236. [Google Scholar] [CrossRef]
- Qiao, C.; Yang, J.; Shen, Q.; Liu, R.; Li, Y.; Shi, Y.; Chen, J.; Shen, Y.; Xiao, Z.; Weng, J.; et al. Traceable Nanoparticles with Dual Targeting and ROS Response for RNAi-Based Immunochemotherapy of Intracranial Glioblastoma Treatment. Adv. Mater. 2018, 30, 1705054. [Google Scholar] [CrossRef]
- Sun, T.; Patil, R.; Galstyan, A.; Klymyshyn, D.; Ding, H.; Chesnokova, A.; Cavenee, W.K.; Furnari, F.B.; Ljubimov, V.A.; Shatalova, E.S.; et al. Blockade of a Laminin-411–Notch Axis with CRISPR/Cas9 or a Nanobioconjugate Inhibits Glioblastoma Growth through Tumor-Microenvironment Cross-talk. Cancer Res. 2019, 79, 1239–1251. [Google Scholar] [CrossRef] [Green Version]
- Shen, Q.; Yang, J.; Liu, R.; Liu, L.; Zhang, J.; Shen, S.; Zhang, X. Hybrid ‘clusterbombs’ as multifunctional nanoplatforms potentiate brain tumor immunotherapy. Mater. Horiz. 2019, 6, 810–816. [Google Scholar] [CrossRef]
- Liu, H.; Chen, L.; Liu, J.; Meng, H.; Zhang, R.; Ma, L.; Wu, L.; Yu, S.; Shi, F.; Li, Y. Co-delivery of tumor-derived exosomes with alpha-galactosylceramide on dendritic cell-based immunotherapy for glioblastoma. Cancer Lett. 2017, 411, 182–190. [Google Scholar] [CrossRef] [PubMed]
- Zhang, F.; Stephan, S.B.; Ene, C.I.; Smith, T.T.; Holland, E.C.; Stephan, M.T. Nanoparticles that reshape the tumor milieu create a therapeutic window for effective t-cell therapy in solid malignancies. Cancer Res. 2018, 78, 3718–3730. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.; Jiang, Y.; Cao, J.; Yang, W.; Zhang, J.; Meng, F.; Zhong, Z. Boosting RNAi therapy for orthotopic glioblastoma with nontoxic brain-targeting chimaeric polymersomes. J. Control. Release 2018, 292, 163–171. [Google Scholar] [CrossRef]
- Peng, Y.; Huang, J.; Xiao, H.; Wu, T.; Shuai, X. Codelivery of temozolomide and siRNA with polymeric nanocarrier for effective glioma treatment. Int. J. Nanomed. 2018, 13, 3467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galstyan, A.; Markman, J.L.; Shatalova, E.S.; Chiechi, A.; Korman, A.J.; Patil, R.; Klymyshyn, D.; Tourtellotte, W.G.; Israel, L.L.; Braubach, O. Blood–brain barrier permeable nano immunoconjugates induce local immune responses for glioma therapy. Nat. Commun. 2019, 10, 3850. [Google Scholar] [CrossRef] [Green Version]
- Lollo, G.; Vincent, M.; Ullio-Gamboa, G.; Lemaire, L.; Franconi, F.; Couez, D.; Benoit, J.-P. Development of multifunctional lipid nanocapsules for the co-delivery of paclitaxel and CpG-ODN in the treatment of glioblastoma. Int. J. Pharm. 2015, 495, 972–980. [Google Scholar] [CrossRef] [Green Version]
- Badie, B.; Berlin, J.M. The future of CpG immunotherapy in cancer. Immunotherapy 2013, 5, 1–3. [Google Scholar] [CrossRef]
- Bastiancich, C.; Bozzato, E.; Luyten, U.; Danhier, F.; Bastiat, G.; Préat, V. Drug combination using an injectable nanomedicine hydrogel for glioblastoma treatment. Int. J. Pharm. 2019, 559, 220–227. [Google Scholar] [CrossRef] [PubMed]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wainwright, D.A.; Balyasnikova, I.V.; Chang, A.L.; Ahmed, A.U.; Moon, K.S.; Auffinger, B.; Tobias, A.L.; Han, Y.; Lesniak, M.S. IDO Expression in Brain Tumors Increases the Recruitment of Regulatory T Cells and Negatively Impacts SurvivalIDO Regulates Treg Infiltration in Brain Tumors. Clinic. Cancer Res. 2012, 18, 6110–6121. [Google Scholar] [CrossRef] [PubMed]
- Löb, S.; Königsrainer, A.; Rammensee, H.G.; Opelz, G.; Terness, P. Inhibitors of indoleamine-2, 3-dioxygenase for cancer therapy: Can we see the wood for the trees? Nat. Rev. Cancer 2009, 9, 445–452. [Google Scholar] [CrossRef]
- Hou, D.-Y.; Muller, A.J.; Sharma, M.D.; DuHadaway, J.; Banerjee, T.; Johnson, M.; Mellor, A.L.; Prendergast, G.C.; Munn, D.H. Inhibition of indoleamine 2, 3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res. 2007, 67, 792–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muller, A.J.; DuHadaway, J.B.; Donover, P.S.; Sutanto-Ward, E.; Prendergast, G.C. Inhibition of indoleamine 2, 3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat. Med. 2005, 11, 312. [Google Scholar] [CrossRef] [PubMed]
- Kuang, J.; Song, W.; Yin, J.; Zeng, X.; Han, S.; Zhao, Y.P.; Tao, J.; Liu, C.J.; He, X.H.; Zhang, X.Z. iRGD Modified Chemo-immunotherapeutic Nanoparticles for Enhanced Immunotherapy against Glioblastoma. Adv. Funct. Mater. 2018, 28, 1800025. [Google Scholar] [CrossRef]
- Kadiyala, P.; Li, D.; Nuñez, F.M.; Altshuler, D.; Doherty, R.; Kuai, R.; Yu, M.; Kamran, N.; Edwards, M.; Moon, J.J. High-Density Lipoprotein-Mimicking Nanodiscs for Chemo-immunotherapy against Glioblastoma Multiforme. ACS Nano 2019, 13, 1365–1384. [Google Scholar] [CrossRef]
- Van Woensel, M.; Mathivet, T.; Wauthoz, N.; Rosière, R.; Garg, A.D.; Agostinis, P.; Mathieu, V.; Kiss, R.; Lefranc, F.; Boon, L. Sensitization of glioblastoma tumor micro-environment to chemo-and immunotherapy by Galectin-1 intranasal knock-down strategy. Sci. Rep. 2017, 7, 1217. [Google Scholar] [CrossRef] [Green Version]
- Chou, S.-T.; Patil, R.; Galstyan, A.; Gangalum, P.R.; Cavenee, W.K.; Furnari, F.B.; Ljubimov, V.A.; Chesnokova, A.; Kramerov, A.A.; Ding, H. Simultaneous blockade of interacting CK2 and EGFR pathways by tumor-targeting nanobioconjugates increases therapeutic efficacy against glioblastoma multiforme. J. Control. Release 2016, 244, 14–23. [Google Scholar] [CrossRef] [Green Version]
- Kozielski, K.L.; Ruiz-Valls, A.; Tzeng, S.Y.; Guerrero-Cázares, H.; Rui, Y.; Li, Y.; Vaughan, H.J.; Gionet-Gonzales, M.; Vantucci, C.; Kim, J. Cancer-selective nanoparticles for combinatorial siRNA delivery to primary human GBM in vitro and in vivo. Biomaterials 2019, 209, 79–87. [Google Scholar] [CrossRef] [PubMed]
- Wohlfart, S.; Gelperina, S.; Kreuter, J. Transport of drugs across the blood–brain barrier by nanoparticles. J. Control. Release 2012, 161, 264–273. [Google Scholar] [CrossRef]
- Lozada-Delgado, E.L.; Grafals-Ruiz, N.; Vivas-Mejía, P.E. RNA interference for glioblastoma therapy: Innovation ladder from the bench to clinical trials. Life Sci. 2017, 188, 26–36. [Google Scholar] [CrossRef]
- Candolfi, M.; Kroeger, K.M.; Muhammad, A.; Yagiz, K.; Farrokhi, C.; Pechnick, R.N.; Lowenstein, P.R.; Castro, M.G. Gene therapy for brain cancer: Combination therapies provide enhanced efficacy and safety. Curr. Gene Ther. 2009, 9, 409–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.S.; Harford, J.B.; Moghe, M.; Slaughter, T.; Doherty, C.; Chang, E.H. A tumor-targeting nanomedicine carrying the p53 gene crosses the blood–brain barrier and enhances anti-PD-1 immunotherapy in mouse models of glioblastoma. Int. J. Cancer 2019, 145, 2535–2546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Speranza, M.-C.; Passaro, C.; Ricklefs, F.; Kasai, K.; Klein, S.R.; Nakashima, H.; Kaufmann, J.K.; Ahmed, A.-K.; Nowicki, M.O.; Obi, P. Preclinical investigation of combined gene-mediated cytotoxic immunotherapy and immune checkpoint blockade in glioblastoma. Neuro-Oncol. 2018, 20, 225–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhaskaran, V.; Nowicki, M.O.; Idriss, M.; Jimenez, M.A.; Lugli, G.; Hayes, J.L.; Mahmoud, A.B.; Zane, R.E.; Passaro, C.; Ligon, K.L. The functional synergism of microRNA clustering provides therapeutically relevant epigenetic interference in glioblastoma. Nat. Commun. 2019, 10, 442. [Google Scholar] [CrossRef] [Green Version]
- Hajj, K.A.; Whitehead, K.A. Tools for translation: Non-viral materials for therapeutic mRNA delivery. Nat. Rev. Mater. 2017, 2, 17056. [Google Scholar] [CrossRef]
- Zhao, C.-Y.; Cheng, R.; Yang, Z.; Tian, Z.-M. Nanotechnology for cancer therapy based on chemotherapy. Molecules 2018, 23, 826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aparicio-Blanco, J.; Sanz-Arriazu, L.; Lorenzoni, R.; Blanco-Prieto, M.J. Glioblastoma chemotherapeutic agents used in the clinical setting and in clinical trials: Nanomedicine approaches to improve their efficacy. Int. J. Pharm. 2020, 581, 119283. [Google Scholar] [CrossRef] [PubMed]
- Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.; Shi, Z.; Liu, R.; Wu, Y.; Zhang, X. Combined-therapeutic strategies synergistically potentiate glioblastoma multiforme treatment via nanotechnology. Theranostics 2020, 10, 3223–3239. [Google Scholar] [CrossRef]
- Jiang, X.; Wang, C.; Fitch, S.; Yang, F. Targeting tumor hypoxia using nanoparticle-engineered CXCR4-overexpressing adipose-derived stem cells. Theranostics 2018, 8, 1350. [Google Scholar] [CrossRef]
- Khan, A.R.; Yang, X.; Fu, M.; Zhai, G. Recent progress of drug nanoformulations targeting to brain. J. Control. Release 2018, 291, 37–64. [Google Scholar] [CrossRef]
- Ganipineni, L.P.; Danhier, F.; Préat, V. Drug delivery challenges and future of chemotherapeutic nanomedicine for glioblastoma treatment. J. Control. Release 2018, 281, 42–57. [Google Scholar] [CrossRef] [PubMed]
- Masters, C.L.; Bateman, R.; Blennow, K.; Rowe, C.C.; Sperling, R.A.; Cummings, J.L. Alzheimer’s disease. Nat. Rev. Dis. Prim. 2015, 1, 15056. [Google Scholar] [CrossRef] [PubMed]
- Blennow, K.; Hampel, H.; Zetterberg, H. Biomarkers in amyloid-β immunotherapy trials in Alzheimer’s disease. Neuropsychopharmacology 2014, 39, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Buée, L.; Bussière, T.; Buée-Scherrer, V.; Delacourte, A.; Hof, P.R. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res. Rev. 2000, 33, 95–130. [Google Scholar] [CrossRef]
- Foidl, B.M.; Humpel, C. Can mouse models mimic sporadic Alzheimer’s disease? Neural Reg. Res. 2020, 15, 401. [Google Scholar]
- Pai, A.S.; Rubinstein, I.; Önyüksel, H. PEGylated phospholipid nanomicelles interact with β-amyloid (1–42) and mitigate its β-sheet formation, aggregation and neurotoxicity in vitro. Peptides 2006, 27, 2858–2866. [Google Scholar] [CrossRef]
- Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A. Curcumin inhibits formation of amyloid β oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem. 2005, 280, 5892–5901. [Google Scholar] [CrossRef] [Green Version]
- Taylor, M.; Moore, S.; Mourtas, S.; Niarakis, A.; Re, F.; Zona, C.; La Ferla, B.; Nicotra, F.; Masserini, M.; Antimisiaris, S.G. Effect of curcumin-associated and lipid ligand-functionalized nanoliposomes on aggregation of the Alzheimer’s Aβ peptide. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 541–550. [Google Scholar] [CrossRef]
- Lovell, M.; Robertson, J.; Teesdale, W.; Campbell, J.; Markesbery, W. Copper, iron and zinc in Alzheimer’s disease senile plaques. J. Neurol. Sci. 1998, 158, 47–52. [Google Scholar] [CrossRef]
- Cui, Z.; Lockman, P.R.; Atwood, C.S.; Hsu, C.-H.; Gupte, A.; Allen, D.D.; Mumper, R.J. Novel D-penicillamine carrying nanoparticles for metal chelation therapy in Alzheimer’s and other CNS diseases. Eur. J. Pharm. Biopharm. 2005, 59, 263–272. [Google Scholar] [CrossRef]
- Podolski, I.Y.; Podlubnaya, Z.; Kosenko, E.; Mugantseva, E.; Makarova, E.; Marsagishvili, L.; Shpagina, M.; Kaminsky, Y.G.; Andrievsky, G.; Klochkov, V. Effects of hydrated forms of C60 fullerene on amyloid β-peptide fibrillization in vitro and performance of the cognitive task. J. Nanosci. Nanotechnol. 2007, 7, 1479–1485. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Zhang, Y.; Yang, Y.; Sun, L.; Han, D.; Li, H.; Wang, C. Pharmacological and toxicological target organelles and safe use of single-walled carbon nanotubes as drug carriers in treating Alzheimer disease. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 427–441. [Google Scholar] [CrossRef] [PubMed]
- De Strooper, B.; Annaert, W. Proteolytic processing and cell biological functions of the amyloid precursor protein. J Cell Sci 2000, 113, 1857–1870. [Google Scholar] [CrossRef] [PubMed]
- Selkoe, D.J. The cell biology of β-amyloid precursor protein and presenilin in Alzheimer’s disease. Trends Cell Bio. 1998, 8, 447–453. [Google Scholar] [CrossRef] [PubMed]
- Verdile, G.; Fuller, S.; Atwood, C.S.; Laws, S.M.; Gandy, S.E.; Martins, R.N. The role of beta amyloid in Alzheimer’s disease: Still a cause of everything or the only one who got caught? Pharmacol. Res. 2004, 50, 397–409. [Google Scholar] [CrossRef] [PubMed]
- Bachmeier, C.; Shackleton, B.; Ojo, J.; Paris, D.; Mullan, M.; Crawford, F. Apolipoprotein E isoform-specific effects on lipoprotein receptor processing. NeuroMol. Med. 2014, 16, 686–696. [Google Scholar] [CrossRef] [Green Version]
- Zhao, N.; Liu, C.-C.; Qiao, W.; Bu, G. Apolipoprotein E, Receptors, and Modulation of Alzheimer’s Disease. Biol Psychiatry 2018, 83, 347–357. [Google Scholar] [CrossRef]
- Sagare, A.P.; Deane, R.; Zlokovic, B.V. Low-density lipoprotein receptor-related protein 1: A physiological Aβ homeostatic mechanism with multiple therapeutic opportunities. Pharmacol. Ther. 2012, 136, 94–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lahiri, D.K.; Farlow, M.R.; Greig, N.H.; Sambamurti, K. Current drug targets for Alzheimer’s disease treatment. Drug Dev. Res. 2002, 56, 267–281. [Google Scholar] [CrossRef]
- Andrieux, K.; Couvreur, P. Nanomedicine as a promising approach for the treatment and diagnosis of brain diseases: The example of Alzheimer’s disease. Ann. Pharm. Fr. 2013, 71, 225–233. [Google Scholar] [CrossRef]
- McGeer, P.L.; McGeer, E.G. Inflammation, autotoxicity and Alzheimer disease. Neurobio. Aging 2001, 22, 799–809. [Google Scholar] [CrossRef] [PubMed]
- Nicoll, J.A.; Wilkinson, D.; Holmes, C.; Steart, P.; Markham, H.; Weller, R.O. Neuropathology of human Alzheimer disease after immunization with amyloid-β peptide: A case report. Nat. Med. 2003, 9, 448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akiyama, H.; McGeer, P.L. Specificity of mechanisms for plaque removal after Aβ immunotherapy for Alzheimer disease. Nat. Med. 2004, 10, 117. [Google Scholar] [CrossRef]
- Schenk, D.; Barbour, R.; Dunn, W.; Gordon, G.; Grajeda, H.; Guido, T.; Hu, K.; Huang, J.; Johnson-Wood, K.; Khan, K. Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999, 400, 173. [Google Scholar] [CrossRef]
- Wyss-Coray, T.; Lin, C.; Yan, F.; Yu, G.-Q.; Rohde, M.; McConlogue, L.; Masliah, E.; Mucke, L. TGF-β1 promotes microglial amyloid-β clearance and reduces plaque burden in transgenic mice. Nat. Med. 2001, 7, 612. [Google Scholar] [CrossRef] [PubMed]
- Wyss-Coray, T.; Yan, F.; Lin, A.H.-T.; Lambris, J.D.; Alexander, J.J.; Quigg, R.J.; Masliah, E. Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer’s mice. Proc. Natl Acad. Sci. USA 2002, 99, 10837–10842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weiner, H.L.; Frenkel, D. Immunology and immunotherapy of Alzheimer’s disease. Nat. Rev. Immunol. 2006, 6, 404. [Google Scholar] [CrossRef]
- Liu, R.; Yang, J.; Liu, L.; Lu, Z.; Shi, Z.; Ji, W.; Shen, J.; Zhang, X. An “Amyloid-β Cleaner” for the Treatment of Alzheimer’s Disease by Normalizing Microglial Dysfunction. Adv. Sci. 2019, 7, 1901555. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhu, F.; Liu, Y.; Zheng, M.; Wang, Y.; Zhang, D.; Anraku, Y.; Zou, Y.; Li, J.; Wu, H. Blood-brain barrier–penetrating siRNA nanomedicine for Alzheimer’s disease therapy. Sci. Adv. 2020, 6, eabc7031. [Google Scholar] [CrossRef]
- Gu, H.; Dodel, R.; Farlow, M.; Du, Y. Advances in the development of antibody-based immunotherapy against prion disease. Antib. Technol. J. 2014, 4, 45. [Google Scholar]
- Winblad, B.; Graf, A.; Riviere, M.-E.; Andreasen, N.; Ryan, J.M. Active immunotherapy options for Alzheimer’s disease. Alzheimer’s Res. Ther. 2014, 6, 7. [Google Scholar] [CrossRef] [PubMed]
- Jadhav, S.; Avila, J.; Schöll, M.; Kovacs, G.G.; Kövari, E.; Skrabana, R.; Evans, L.D.; Kontsekova, E.; Malawska, B.; de Silva, R. A walk through tau therapeutic strategies. Acta Neuropathol. Commun. 2019, 7, 22. [Google Scholar] [CrossRef] [PubMed]
- Albert, M.; Mairet-Coello, G.; Danis, C.; Lieger, S.; Caillierez, R.; Carrier, S.; Skrobala, E.; Landrieu, I.; Michel, A.; Schmitt, M. Prevention of tau seeding and propagation by immunotherapy with a central tau epitope antibody. Brain 2019, 142, 1736–1750. [Google Scholar] [CrossRef] [PubMed]
- De Genst, E.; Messer, A.; Dobson, C.M. Antibodies and protein misfolding: From structural research tools to therapeutic strategies. Biochim. Biophys. Acta 2014, 1844, 1907–1919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Breijyeh, Z.; Karaman, R. Comprehensive review on Alzheimer’s disease: Causes and treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
- Bellucci, A.; Mercuri, N.B.; Venneri, A.; Faustini, G.; Longhena, F.; Pizzi, M.; Missale, C.; Spano, P. Parkinson’s disease: From synaptic loss to connectome dysfunction. Neuropathol. Appl. Neurobiol. 2016, 42, 77–94. [Google Scholar] [CrossRef]
- Dehay, B.; Bourdenx, M.; Gorry, P.; Przedborski, S.; Vila, M.; Hunot, S.; Singleton, A.; Olanow, C.W.; Merchant, K.M.; Bezard, E. Targeting α-synuclein for treatment of Parkinson’s disease: Mechanistic and therapeutic considerations. Lancet Neurol. 2015, 14, 855–866. [Google Scholar] [CrossRef] [Green Version]
- Spencer, B.; Potkar, R.; Trejo, M.; Rockenstein, E.; Patrick, C.; Gindi, R.; Adame, A.; Wyss-Coray, T.; Masliah, E. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in α-synuclein models of Parkinson’s and Lewy body diseases. J. Neurosci. 2009, 29, 13578–13588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.-J.; Bae, E.-J.; Lee, S.-J. Extracellular α-synuclein—A novel and crucial factor in Lewy body diseases. Nat. Rev. Neurol. 2014, 10, 92. [Google Scholar] [CrossRef] [PubMed]
- Stoddard-Bennett, T.; Reijo Pera, R. Treatment of Parkinson’s Disease through Personalized Medicine and Induced Pluripotent Stem Cells. Cells 2019, 8, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, B.; Ng, H.H.; Takahashi, R.; Tan, E.-K. Induced pluripotent stem cells in Parkinson’s disease: Scientific and clinical challenges. J Neurol Neurosurg Psychiatry 2016, 87, 697–702. [Google Scholar] [CrossRef]
- Witt, J.; Marks, W.J. An update on gene therapy in Parkinson’s disease. Curr. Neurol. Neurosci. Rep. 2011, 11, 362–370. [Google Scholar] [CrossRef] [PubMed]
- Vinogradov, S.V.; Batrakova, E.V.; Kabanov, A.V. Nanogels for oligonucleotide delivery to the brain. Bioconjugate Chem. 2004, 15, 50–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kurakhmaeva, K.B.; Djindjikhashvili, I.A.; Petrov, V.E.; Balabanyan, V.U.; Voronina, T.A.; Trofimov, S.S.; Kreuter, J.; Gelperina, S.; Begley, D.; Alyautdin, R.N. Brain targeting of nerve growth factor using poly (butyl cyanoacrylate) nanoparticles. J. Drug Target. 2009, 17, 564–574. [Google Scholar] [CrossRef]
- Mohanraj, K.; Sethuraman, S.; Krishnan, U.M. Development of poly (butylene succinate) microspheres for delivery of levodopa in the treatment of parkinson’s disease. J. Biomed. Mater. Res. Part B: Appl. Biomater. 2013, 101, 840–847. [Google Scholar] [CrossRef] [PubMed]
- Zhang, G.; Wang, T.; Xia, Y.; Wan, F.; Ma, K.; Guo, X.F.; Kou, L.; Yin, S.; Han, C.; Liu, L. New perspectives on roles of alpha-synuclein in Parkinson’s disease. Front. Aging Neurosci. 2018, 10, 370. [Google Scholar] [CrossRef] [Green Version]
- Olanow, C.W.; Obeso, J.A.; Stocchi, F. Drug insight: Continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Nat. Rev. Neurol. 2006, 2, 382. [Google Scholar] [CrossRef] [PubMed]
- Moloney, K.K. Age and IFN-Gamma Deficiency Modulate the Impact of Repeated Paraquat Exposure in a Mouse Model of Parkinson’s Disease; Carleton University: ottawa, ON, Canada, 2011. [Google Scholar]
- Heinemann, U.; Kaufer, D.; Friedman, A. Blood-brain barrier dysfunction, TGFβ signaling, and astrocyte dysfunction in epilepsy. Glia 2012, 60, 1251–1257. [Google Scholar] [CrossRef] [Green Version]
- Lashuel, H.A.; Overk, C.R.; Oueslati, A.; Masliah, E. The many faces of α-synuclein: From structure and toxicity to therapeutic target. Nat. Rev. Neurol. 2013, 14, 38. [Google Scholar] [CrossRef] [Green Version]
- Valera, E.; Masliah, E. Therapeutic approaches in Parkinson’s disease and related disorders. J. Neurochem. 2016, 139, 346–352. [Google Scholar] [CrossRef] [Green Version]
- Mandler, M.; Valera, E.; Rockenstein, E.; Weninger, H.; Patrick, C.; Adame, A.; Santic, R.; Meindl, S.; Vigl, B.; Smrzka, O. Next-generation active immunization approach for synucleinopathies: Implications for Parkinson’s disease clinical trials. Acta Neuropathol. 2014, 127, 861–879. [Google Scholar] [CrossRef] [PubMed]
- Mandler, M.; Valera, E.; Rockenstein, E.; Mante, M.; Weninger, H.; Patrick, C.; Adame, A.; Schmidhuber, S.; Santic, R.; Schneeberger, A. Active immunization against alpha-synuclein ameliorates the degenerative pathology and prevents demyelination in a model of multiple system atrophy. Mol. Neurodegener. 2015, 10, 10. [Google Scholar] [CrossRef] [Green Version]
- Masliah, E.; Rockenstein, E.; Mante, M.; Crews, L.; Spencer, B.; Adame, A.; Patrick, C.; Trejo, M.; Ubhi, K.; Rohn, T.T. Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease. PLoS ONE 2011, 6, e19338. [Google Scholar] [CrossRef] [Green Version]
- Bae, E.-J.; Lee, H.-J.; Rockenstein, E.; Ho, D.-H.; Park, E.-B.; Yang, N.-Y.; Desplats, P.; Masliah, E.; Lee, S.-J. Antibody-aided clearance of extracellular α-synuclein prevents cell-to-cell aggregate transmission. J. Neurosci. 2012, 32, 13454–13469. [Google Scholar] [CrossRef] [Green Version]
- Roodveldt, C.; Labrador-Garrido, A.; Gonzalez-Rey, E.; Lachaud, C.C.; Guilliams, T.; Fernandez-Montesinos, R.; Benitez-Rondan, A.; Robledo, G.; Hmadcha, A.; Delgado, M. Preconditioning of microglia by α-synuclein strongly affects the response induced by toll-like receptor (TLR) stimulation. PLoS ONE 2013, 8, e79160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, D.; Yoo, J.M.; Hwang, H.; Lee, J.; Lee, S.H.; Yun, S.P.; Park, M.J.; Lee, M.; Choi, S.; Kwon, S.H.; et al. Graphene quantum dots prevent α-synucleinopathy in Parkinson’s disease. Nat. Nanotechnol. 2018, 13, 812–818. [Google Scholar] [CrossRef] [PubMed]
- Bates, G.; Dorsey, R.; Gusella, J. Nat Rev Dis Primers. 2015.
- Bradford, J.; Shin, J.-Y.; Roberts, M.; Wang, C.-E.; Li, X.-J.; Li, S. Expression of mutant huntingtin in mouse brain astrocytes causes age-dependent neurological symptoms. Proc. Natl. Acad. Sci. USA 2009, 106, 22480–22485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative stress in neurodegenerative diseases: From molecular mechanisms to clinical applications. Oxidative Med. Cell. Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef] [PubMed]
- Jin, H.; Chen, W.; Tang, X.; Chiang, L.; Yang, C.; Schloss, J.; Wu, J. Polyhydroxylated C60, fullerenols, as glutamate receptor antagonists and neuroprotective agents. J. Neurosci. Res. 2000, 62, 600–607. [Google Scholar] [CrossRef]
- Godinho, B.M.; Ogier, J.R.; Darcy, R.; O’Driscoll, C.M.; Cryan, J.F. Self-assembling modified β-cyclodextrin nanoparticles as neuronal siRNA delivery vectors: Focus on Huntington’s disease. Mol. Pharm. 2013, 10, 640–649. [Google Scholar] [CrossRef]
- Sandhir, R.; Yadav, A.; Mehrotra, A.; Sunkaria, A.; Singh, A.; Sharma, S. Curcumin nanoparticles attenuate neurochemical and neurobehavioral deficits in experimental model of Huntington’s disease. Neuromolecular Med. 2014, 16, 106–118. [Google Scholar] [CrossRef] [PubMed]
- Bhatt, R.; Singh, D.; Prakash, A.; Mishra, N. Development, characterization and nasal delivery of rosmarinic acid-loaded solid lipid nanoparticles for the effective management of Huntington’s disease. Drug Deliv. 2015, 22, 931–939. [Google Scholar] [CrossRef]
- Cong, W.; Bai, R.; Li, Y.-F.; Wang, L.; Chen, C. Selenium nanoparticles as an efficient nanomedicine for the therapy of Huntington’s disease. ACS Appl. Mater. Interfaces 2019, 11, 34725–34735. [Google Scholar] [CrossRef] [PubMed]
- Ventola, C.L. Progress in nanomedicine: Approved and investigational nanodrugs. Pharm. Ther. 2017, 42, 742. [Google Scholar]
- D’Mello, S.R.; Cruz, C.N.; Chen, M.-L.; Kapoor, M.; Lee, S.L.; Tyner, K.M. The evolving landscape of drug products containing nanomaterials in the United States. Nat. Nanotechnol. 2017, 12, 523–529. [Google Scholar] [CrossRef]
- Maier-Hauff, K.; Ulrich, F.; Nestler, D.; Niehoff, H.; Wust, P.; Thiesen, B.; Orawa, H.; Budach, V.; Jordan, A. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J. Neuro-Oncol. 2011, 103, 317–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Young, J.S.; Bernal, G.; Polster, S.P.; Nunez, L.; Larsen, G.F.; Mansour, N.; Podell, M.; Yamini, B. Convection-enhanced delivery of polymeric nanoparticles encapsulating chemotherapy in canines with spontaneous supratentorial tumors. World Neurosurg. 2018, 117, e698–e704. [Google Scholar] [CrossRef] [PubMed]
- Abdolahi, M.; Jafarieh, A.; Sarraf, P.; Sedighiyan, M.; Yousefi, A.; Tafakhori, A.; Abdollahi, H.; Salehinia, F.; Djalali, M. The neuromodulatory effects of ω-3 fatty acids and nano-curcumin on the COX-2/iNOS network in migraines: A clinical trial study from gene expression to clinical symptoms. Endocr. Metab. Immune Disord. -Drug Targets (Former. Curr. Drug Targets-Immune Endocr. Metab. Disord.) 2019, 19, 874–884. [Google Scholar] [CrossRef]
- Abdolahi, M.; Tafakhori, A.; Togha, M.; Okhovat, A.A.; Siassi, F.; Eshraghian, M.R.; Sedighiyan, M.; Djalali, M.; Mohammadzadeh Honarvar, N.; Djalali, M. The synergistic effects of ω-3 fatty acids and nano-curcumin supplementation on tumor necrosis factor (TNF)-α gene expression and serum level in migraine patients. Immunogenetics 2017, 69, 371–378. [Google Scholar] [CrossRef]
- Soveyd, N.; Abdolahi, M.; Djalali, M.; Hatami, M.; Tafakhori, A.; Sarraf, P.; Honarvar, N.M. The combined effects of ω-3 fatty acids and nano-curcumin supplementation on intercellular adhesion molecule-1 (ICAM-1) gene expression and serum levels in migraine patients. CNS Neurol. Disord. -Drug Targets (Former. Curr. Drug Targets-CNS Neurol. Disord.) 2017, 16, 1120–1126. [Google Scholar] [CrossRef]
- Lackner, P.; Beer, R.; Broessner, G.; Helbok, R.; Galiano, K.; Pleifer, C.; Pfausler, B.; Brenneis, C.; Huck, C.; Engelhardt, K. Efficacy of silver nanoparticles-impregnated external ventricular drain catheters in patients with acute occlusive hydrocephalus. Neurocritical Care 2008, 8, 360–365. [Google Scholar] [CrossRef] [PubMed]
- Grauer, O.; Jaber, M.; Hess, K.; Weckesser, M.; Schwindt, W.; Maring, S.; Wölfer, J.; Stummer, W. Combined intracavitary thermotherapy with iron oxide nanoparticles and radiotherapy as local treatment modality in recurrent glioblastoma patients. J. Neuro-Oncol. 2019, 141, 83–94. [Google Scholar] [CrossRef] [PubMed]
- Saleh, A.; Schroeter, M.; Ringelstein, A.; Hartung, H.-P.; Siebler, M.; Mödder, U.; Jander, S. Iron oxide particle-enhanced MRI suggests variability of brain inflammation at early stages after ischemic stroke. Stroke 2007, 38, 2733–2737. [Google Scholar] [CrossRef] [PubMed]
Name of Nanomedicine | Disease Name | Purpose | Targeting Plan | Outcomes | Reference(s) |
---|---|---|---|---|---|
LBNP | GBM | Anticancer, in vivo image | 17 peptides | Enhanced bioavailability | [130,131,132] |
PD | PD treatment | Lamp-2b Chlorotoxin | Targeted delivery | [133,134] | |
AD | AD treatment | Lamp-2b | Enhances drug delivery, efficiency, and accessibility | [135] | |
Au-NP | GBM | PDT, PTT | Transferrin peptide RVG29 Peptide | Improves the cellular intake/enhances the efficiency of photodynamic therapy | [136,137,138] |
PD | PD Treatment | chitosan | Enhances the efficiency of the | [139] | |
AD | AD treatment | RVG29 Peptide | amyloid-β inhibitor | [140] | |
PNP | GBM | Anticancer | AS14111 aptamer Transferrin Pep-1 Angiopep | Enhance efficiency and anti-glioma | [141,142,143,144,145] |
PD | PD treatment | ApoE | Enhance the neuroprotective efficiency | [146,147,148,149] | |
AD | AD treatment PET | ApoE 125I-clioquinol | Beta amyloid-induced cytotoxicity is enhanced by curcumin | [150,151] | |
IO-NP | GBM | MRI/TEM | Chlorotoxin, chitosan, Anti-EGFRvIII | Targeted therapy and enhanced delivery | [152,153] |
PD | MRI | Anti-ferritin | Detection | [154] | |
AD | MRI | Anti-AβPP, Anti-ferritin | Improved amyloid-β revealing | [86,155] |
NPs | Disease | Mechanisms | Effects | References |
---|---|---|---|---|
Silver NPs | Acute occulsive hydrocephalus | Ventricullitis caused by catheters is prevented | Enhanced the health of patients | [310] |
Magnetic iron oxide NPs + reduced radiotherphy | Glioblastomia multiformie | Increased Caspase-3, heat shock protein, and programmed death ligand 1 levels suppress tumor growth | Enhanced the overall survival rate of patients | [305,311] |
Ultrasmall magnetic iron oxide | Ischemic stroke | Activates macrophages | Targeted inflammatory cytokines more effectively | [312] |
Nano-curcumin + ω-3 fatty acid | Migraine | Intercellular adhesion molecule 1, TNF-a, and cyclooxygenase-2/inducible nitric acid gene expression is suppressed | Relieved headaches | [307,308,309] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Guo, Z.-H.; Khattak, S.; Rauf, M.A.; Ansari, M.A.; Alomary, M.N.; Razak, S.; Yang, C.-Y.; Wu, D.-D.; Ji, X.-Y. Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders. Molecules 2023, 28, 1283. https://doi.org/10.3390/molecules28031283
Guo Z-H, Khattak S, Rauf MA, Ansari MA, Alomary MN, Razak S, Yang C-Y, Wu D-D, Ji X-Y. Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders. Molecules. 2023; 28(3):1283. https://doi.org/10.3390/molecules28031283
Chicago/Turabian StyleGuo, Zi-Hua, Saadullah Khattak, Mohd Ahmar Rauf, Mohammad Azam Ansari, Mohammad N. Alomary, Sufyan Razak, Chang-Yong Yang, Dong-Dong Wu, and Xin-Ying Ji. 2023. "Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders" Molecules 28, no. 3: 1283. https://doi.org/10.3390/molecules28031283
APA StyleGuo, Z. -H., Khattak, S., Rauf, M. A., Ansari, M. A., Alomary, M. N., Razak, S., Yang, C. -Y., Wu, D. -D., & Ji, X. -Y. (2023). Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders. Molecules, 28(3), 1283. https://doi.org/10.3390/molecules28031283