Nano-based Cell therapy for AD and PD

Article Information

Ashok Chakraborty*, Anil Diwan

Department of Biochemistry, AllExcel, Inc., 118, Wood Street, Ste. 201, West Haven, CT 06516; USA

*Corresponding author: Ashok Chakraborty, Department of Biochemistry, AllExcel, Inc., 118, Wood Street, Ste. 201, West Haven, CT 06516; USA.

Received: 03 June 2024; Accepted: 10 June 2024; Published: 21 June 2024

Citation: Ashok Chakraborty, Anil Diwan. Nano-based Cell therapy for AD and PD. Fortune Journal of Health Sciences. 7 (2024): 318-324.

View / Download Pdf Share at Facebook

Abstract

The most prevalent neurodegenerative diseases are Alzheimer’s disease (AD) and Parkinson’s disease (PD). Both AD and PD are classified as proteinopathies where misfolded amyloid-β, and tau proteins in AD and α-synuclein in PD are noticed. The main AD hallmarks are memory loss where the loss of dopaminergic neurons and the development of Lewy-bodies are found in PD. The defects in the motor neuron activities, however, can be noticed after the loss of dopaminergic neurons by 50-70% in the Substantia nigra (SN) region. Emerging Evidences are there to suggest suggests that misfolded protein tangles and/or plaques have prion-like proteins which are the major factor causing the pathogenesis. Additional factors that can affect pathology of theses diseases include oxidative stress, mitochondrial damage, inflammation, and age-related cell death. Chronic inflammation is also universally thought to play a central role in the initiation and progression of PD. At present no such real therapies are yet available for the cure of AD and PD, besides some palliative treatment. However, efforts are in the process to find some effective therapies using transplantable neural cells, gene therapies, and better and some nanomaterials, for better targeting across the blood-brain barrier. Nanomaterials, further can increase the drug half-lives, protect cargo from immune detection, and provide a physical structure that can support cell growth.

Keywords

Alzheimer disease, Parkinson’s disease, Nanoparticles, Macrophages, Cell therapy

Alzheimer disease articles, Parkinson?s disease articles, Nanoparticles articles, Macrophages articles, Cell therapy articles

Article Details

Abbreviations

Angiotensin-converting enzyme 2 (ACE2)

Alzheimer’s disease (AD)

Aminopeptidase N (APN)

Amyloid-β (Aβ)

Blood brain barrier (BBB)

Brain-derived neurotrophic factor (BDNF)

Central nervous system (CNS)

Dopaminergic (DA-ergic)

Glial cell derived neural factor (GDNF)

Human coronavirus 229E (hCoV-229E)

Interleukin-1β (IL-1β)

Inducible nitric oxide synthase (iNOS)

Lewy bodies (LB)

Macrophages (M.s)

Monocyte chemotactic protein-1 (MCP-1)

Neural Stem cells (NSCs)

Parkinson’s disease (PD)

Polyethylene glycol (PEG)

Poly (lactic-co-glycolic acid) (PLGA)

Arg-Gly-Asp (RGD)

Reactive oxygen species (ROS)

Substantia nigra (SN)

α-Synuclein (α-syn)

Severe Acute Respiratory Syndrome (SARS)

Tumor necrosis factor-α (TNF- α)

1. Introduction

Protein aggregation is a typical phenomenon in both Parkinson’s disease (PD) and Alzheimer’s disease (AD). In particular, PD is characterized by the deposition of aggregated misfolded α-synuclein (α-syn) protein, which is known as Lewy bodies (LB) in dopaminergic neurons, and thus causes severe motor dysfunction [1]. On the other hand, AD where the cognitive process is typically lost is diagnosed with the abnormal accumulation of amyloid-β (Aβ) plaque and tau neurofibrillary tangles in the brain [2].  Supporting evidences are there to clarify the proteinopathy due to the accumulation of pathological α-syn, Aβ, and tau that spreads from cell-to-cell [3-12]. The α-syn aggregates localize in the mitochondria and induces mitochondrial fragmentation and decreased membrane potential [13-15].  In AD, the aggregation of Aβ peptide results from the oxidative stress from dysfunctional mitochondrial reactive oxygen species (ROS) [16-18]. Further, abnormal hyper-phosphorylation of microtubule-associated tau protein leads to the formation of tau tangles with prion-like activity. In both the diseases, AD and PD, expression of inflammatory cytokines have been noticed which may cause the protein aggregation and cell death. However, inhibition of amyloidosis in both AD or PD with small molecules and antibodies exhibit only a little success [19-21].

Traditional therapeutic drugs usually have off-target effects. Nanomaterial formulations can ensure targeted delivery and also can overcome the blood brain barrier (BBB) during delivery of the actives [22-25]. Additionally, composite nanomaterials can be developed to ensure the new cell growth in PD and AD [26]. In this review, we will explore the current field of nanomaterials for therapeutic application of cell-based therapies in PD and AD.

2. Present Treatment Strategy

Except some palliative treatment, like using Dopa/Dopamin, currently, there is no curative therapy for PD or AD. However, efforts are in progress to regenerate the neuronal growth, axonal extension and to repair the damaged cell in both the diseases using some neurotropic factors like BDNF, GDNF.  Nanomaterials like scaffolds, PLGA are thoughts to be used not only for targeted delivery of the neurotropic factors but also to increase their half-life and to improve the motor function and dopaminergic neuron restoration without showing any toxic effects in vivo [27-29].

Further, maturation of neurons can be done, in vitro, by using scaffolds pretreated with RGD and heparin before administration to an in vivo system [30].  In this regards, various scaffolds are being made using alternating amino acid sequences, which support neuronal growth and differentiation [31, 32].  Humanin peptide, which is known to inhibit Aβ-related cell death in AD can be delivered successfully with polymersomes (PEG-PLGA) [33].

Cell Therapy: Cell therapy of PD by transplantation of stem cells has been proposed earlier. However, there are enough demerits in that approach, like availability of enough cells for transplantation, probability of future development of carcinoma, and so many other logistic challenges including cost of the treatment. 

Neural Stem cells (NSCs) have been considered as the preferred cells for transplantation as those can produce dopamine, the active principle for PD/AD treatment [34-36].  Besides the production od Dopamine, NSCs posses the ability to control the level dopamine in the synaptic cleft as it contains the Dopamine catabolizing enzymes, and as a result offers a less probability of having future neural tube defects that can result from the excess unused floating dopamine in the Substantia Nigra (SN region). Further, NSCs can also be used as a vehicle for delivering other neuron co-stimulatory drugs, and/or chemotherapeutic agents.  In fact, NSCs have been found to deliver neurotrophic factors to the CNS and promote neuron integrity and regeneration [37-40].

Macrophages, another possible therapeutic cells against PD/AD: PD is believed to be the result of chronic inflammation [41, 42]. MFs, the main regulatory immune cells that acts in the periphery have the ability to polarize either to its M1 type or to M2 type under the influence of environmental factors [43].  M1 type of MFs while is pro-inflammatory, and releases several chemokines, such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF- α), and monocyte chemotactic protein-1 (MCP-1), and iNOS, the M2- MFs produce anti-inflammatory cytokines, like IL- 10, IL-4, IL-13, and promote tissue repair [44-47]. During the inflammatory disease progression, M2-MFs are gradually replaced by M1-type [48]. With the similar note it is believed that somehow in the brain MFs are polarizes to M1-type during the pathogenesis of PD. In fact, MFs and neutrophils are able to cross the blood brain barrier, and secrete proinflammatory cytokines (e.g, ILs, TNFa, IFN-g), which can cause the development of PD development [49].

Use of Nanotechnology for Targeted Tissue Delivery:

As an extension of the targeted cell therapy, the use of nanovehicles to carry NSCs along with the neurotropic factors across the Blood-Brian Barrier (BBB) have been proposed [50-53].

  1. Liposomes: Liposomes are capable to encapsulate both the hydrophobic and hydrophilic drugs into its hollow core and deliver them to the disease sites [54-56]. Long-circulating liposomes, like PEGylated liposomes, can also be prepared by coating with polyethylenglycol [58].  In clinical trials with the treatment of histoplasmosis, meningitis, and neutropenia, PEGylated liposomes showed better efficacy with less side effects [59].
  2. Polymeric Micelles: Amphilic polymeric micelles are made up of PEG-.-PCL), poly(styrene) or PLGA [60]. This type of block copolymers are, indeed, approved by FDA for the targeted drug delivery [60]. In that hydrophilic shell, hydrophobic drugs can stay protected from the surrounding environment. By designing the cell-specific ligands and attached to the polymer can make the polymer to be directed towards the specific cell types [61].
  3. Nanoviricide®: is comprised of a polyethylene glycol (PEG) and alkyl pendants. The alkyl chains while make a flexible core the PEG forms the hydrophilic shell and imparts non-immunogenicity. The resulting polymeric materials form stable micelles with chemical groups that are uniformly distributed along the polymer chain. This polymeric chain attaches with the virus-specific ligands like chemical moieties, peptides, antibody fragments or other proteins (Fig. 1). Recently, NV-CoV-2, a biopolymer designed and made by Nanoviricide® (Shelton, CT) are potentially active against many viruses including Corona virus [62].  In the antiviral therapy of corona virus, NV-CoV-2 is covalently bonded with antiviral small chemical ligands similar to S-protein which binds to the cognate cellular receptor, ACE2. NV-CoV-2 also can bind the other SARS receptor, Aminopeptidase N (APN), and showed inhibitory activities against the the human coronavirus 229E (hCoV-229E) which uses APN receptor for binding to the cell.[62].
fortune-biomass-feedstock

Figure 1A: Key Players for AD Pathogenesis

fortune-biomass-feedstock

Figure 1B: Key players of PD

Conclusions and Future Perspectives:

The treatment of PD is still controversial as there is no defined diagnostic tool for the early detection of the disease and also their pathological pathway. Pharmacological therapy, in fact actually starts when the patient notices the difficult motor activities, and at that point 50-70% of dopaminergic neurons actually have already been lost which makes it practically impossible to cure [62]. Current therapies mainly focusing palliative treatment and/or slowing down and reversing the abnormal motor symptoms.

fortune-biomass-feedstock

Figure 2: Schematic Design of Nanoviricide Biopolymer

fortune-biomass-feedstock

Figure 3: Reprogrammimg Macrophages to its M2 type for AD/PD Theraphy

fortune-biomass-feedstock

Figure 4: Step# 1: Macrophage Repolarisation to it M2-type in cell culture dishes in Presence of reprogramming cytokines. Grow the cell until enough amount of M2-Mφs are obtained.

Step#2: Cell-Cell interaction between M2 type Mφs an hNSCs to get an unique cell which should express anti inflammatory cytokines, as well as need-based dopamine.

Several studies have demonstrated that nanosystems could potentiate sustained release of the repurposed PD drugs with no side-effects, and increase the effectiveness of the therapeutic agents.  Moreover, they can be used for effective delivery to the target across the BBB.  Great efforts are still in the process to develop new effective PD therapeutics, like salfinamide and opicapone which may decrease motor fluctuations [63-66], a.syn target therapies [67], and neural stem cell transplantation therapies, and gene therapy [68]. Our Plan of Action is to create a Novel Therapeutic Module for PD and AD. Accumulation of M1 type of macrophages in the brain is the key player of PD onset [69].  Repolarization to its M2-type by cytokines, nanoparticles or both could be the effective the strategy that could be deployed for PD therapy (Fig. 2). The DA-ergic NSC cells while will replenish the loss of neural cells the anti-inflammatory cytokines can also be the used as a vehicle of the M2 polarizing cytokines (IFNg-GMCSF, IL-10, IL-4, TGF-b, IL-12, etc.) to re-educate M1 type to M2 type.  Thus, in combination, therapeutic benefits could be achieved for the PD victims.

Acknowledgements:

We acknowledge all our colleagues for their help during the preparation of the manuscript by providing all the relevant information.

Funding:

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Consent for Publications: Both the authors have agreed to submit this paper for publication.

Ethical Approval: Not applicable

Conflict of Interest Statement: The authors declare no conflict of interests.

References

  1. Srinivasan E, Chandrasekhar G, Chandrasekar P, Anbarasu K, Vickram AS, Karunakaran R, et. al. Alpha-Synuclein Aggregation in Parkinson's Disease. Front Med (Lausanne) 8 (2021): 736978.
  2. Knopman DS, Amieva H, Petersen RC. et al.Alzheimer disease. Nat Rev Dis Primers 7 (2021): 33.
  3. Braak H, del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Aging 24 (2003): 197-211.
  4. Kordower JH, Chu Y, Hauser RA, Freeman TB, and Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Med 14 (2008): 504-506.
  5. Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Med 14 (2008): 501-503.
  6. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 338 (2012): 949-953.
  7. Guo JL, Narasimhan S, Changolkar L, He Z, Stieber A, Zhang B, et al. Unique pathological tau conformers from Alzheimer’s brains transmit tau pathology in nontransgenic mice. Exp. Med 213 (2016): 2635-2654.
  8. Mao X, Ou MT, Karuppagounder SS, Kam TI, Yin X, Xiong Y, et al. Pathological α-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science 353 (2016): aah3374.
  9. Tyson T, Steiner J. A, and Brundin P. Sorting out release, uptake and processing of alpha-synuclein during prion-like spread of pathology. Neurochem. 2016, 139(Suppl. 1), 275-289. doi: 10.1111/jnc.13449.
  10. He Z, Guo JL, McBride JD, Narasimhan S, Kim H, Changolkar L, et al. Amyloid-β plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Med. 24 (2018): 29-38.
  11. Kam TI, Mao X, Park H, Chou SC, Karuppagounder SS, Umanah GE, et al. Poly (ADP-ribose) drives pathologic a-synuclein neurodegeneration in Parkinson’s disease. Science 362 (2018): eaat8407.
  12. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder S. S, Lee S, et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103 (2019): 627-641.
  13. Li WW, Yang R, Guo JC, Ren HM, Zha XL, Cheng JS, et al. Localization of α-synuclein to mitochondria within midbrain of mice. NeuroReport 18 (2007): 1543-1546.
  14. Devi L, Raghavendran V, Prabhu BM, Avadhani NG and Anandatheerthavarada HK. Mitochondrial import and accumulation of α-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. Biol. Chem 283 (2008): 9089-9100.
  15. Ganjam GK, Bolte K, Matschke LA, Neitemeier S, Dolga AM, Höllerhage M, et al. Mitochondrial damage by α-synuclein causes cell death in human dopaminergic neurons. Cell Death Dis 10 (2019): 865.
  16. Tönnies E and Trushina E. Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease. Alzheimer’s Dis 57 (2017): 1105-1121.
  17. Gupta J, Fatima MT, Islam Z, Khan RH, Uversky VN and Salahuddin P. Nanoparticle formulations in the diagnosis and therapy of Alzheimer’s disease. J. Biol. Macromol 130 (2019): 515-526.
  18. Poulson BG, Szczepski K, Lachowicz JI, Jaremko L, Emwas AH, and Jaremko M. Aggregation of biologically important peptides and proteins: inhibition or acceleration depending on protein and metal ion concentrations. RSC Adv 10 (2019): 215-227.
  19. Tufekci KU, Meuwissen R, Genc S, and Genc K. “Inflammation in Parkinson’s disease,” in Advances in Protein Chemistry and Structural Biology, ed. R. Donev (Oxford: Academic Press Inc.) (2012): 69-132.
  20. Lema Tomé CM, Tyson T, Rey NL, Grathwohl S, Britschgi M, and Brundin P. Inflammation and α-synuclein’s prion-like behavior in Parkinson’s disease-is there a link? Neurobiol 47 (2013): 561-574.
  21. Rivest S. TREM2 enables amyloid β clearance by microglia. Cell Res 25 (2015): 535-536.
  22. Wang H, Wan K and Shi X. Recent advances in nanozyme research. Mater 31 (2019): 1805368.
  23. Eleftheriadou D, Kesidou D, Moura F, Felli E, and Song W. Redox-responsive nanobiomaterials-based therapeutics for neurodegenerative diseases. Small 16 (2020): e1907308.
  24. Ulbrich W and Lamprecht A. Targeted drug-delivery approaches by nanoparticulate carriers in the therapy of inflammatory diseases. R. Soc. Interface 7 (2010): S55-S66.
  25. Leyva-Gómez G, Cortés H, Magaña JJ, Leyva-García N, Quintanar-Guerrero D and Florán B. Nanoparticle technology for treatment of Parkinson’s disease: the role of surface phenomena in reaching the brain. Drug Discov. Today 20 (2015): 824-837.
  26. Bordoni M, Scarian E, Rey F, Gagliardi S, Carelli S, Pansarasa O, et al. Biomaterials in neurodegenerative disorders: a promising therapeutic approach. J. Mol. Sci 21 (2020): 3243.
  27. Garbayo E, Ansorena E, Lana H, Carmona-Abellan M, del M, Marcilla I, et al. Brain delivery of microencapsulated GDNF induces functional and structural recovery in Parkinsonian monkeys. Biomaterials 110 (2016): 11-23.
  28. Wang TY, Bruggeman KF, Kauhausen JA, Rodriguez AL, Nisbet DR and Parish CL. Functionalized composite scaffolds improve the engraftment of transplanted dopaminergic progenitors in a mouse model of Parkinson’s disease. Biomaterials 74 (2016): 89-98.
  29. Ucar B and Humpel C. Therapeutic efficacy of glial cell-derived neurotrophic factor loaded collagen scaffolds in ex vivo organotypic brain slice Parkinson’s disease models. Brain Res. Bull 149 (2019): 86-95.
  30. Adil MM, Vazin T, Ananthanarayanan B, Rodrigues GMC, Rao AT, Kulkarni RU, et al. Engineered hydrogels increase the post-transplantation survival of encapsulated hESC-derived midbrain dopaminergic neurons. Biomaterials 136 (2017): 1-11.
  31. Bordoni M, Scarian E, Rey F, Gagliardi S, Carelli S, Pansarasa O, et al. Biomaterials in neurodegenerative disorders: a promising therapeutic approach. J. Mol. Sci 21 (2020): 3243.
  32. Cui GH, Shao SJ, Yang JJ, Liu JR and Guo HD. Designer self-assemble peptides maximize the therapeutic benefits of neural stem cell transplantation for Alzheimer’s disease via enhancing neuron differentiation and paracrine action. Neurobiol 53 (2016): 1108-1123.
  33. Yu Y, Jiang X, Gong S, Feng L, Zhong Y and Pang Z. The proton permeability of self-assembled polymersomes and their neuroprotection by enhancing a neuroprotective peptide across the blood-brain barrier after modification with lactoferrin. Nanoscale 6 (2014): 3250-3258.
  34. Chakraborty A, Diwan A. Selection of Cells for Parkinson’s Disease Cell-Therapy. Int J Stem Cell Res Ther 6 (2019): 063.
  35. Chakraborty A and Diwan A. Alzheimer Disease (AD) Cell Therapy Using Human Neural Stem Cells: Validity of the Approach. J Brain Res 3 (2020): 2.
  36. Chakraborty A and Diwan A. Modified neural stem cells: A new regimen for cell therapy of Alzheimer’s and Parkinson’s disease. Current Trends in Neurology 15 (2021): 43-48.
  37. Nie L, Yao D, Chen S, et al.Directional induction of neural stem cells, a new therapy for neurodegenerative diseases and ischemic stroke. Cell Death Discov 9 (2023): 215
  38. Vishwakarma SK, Bardia A, Tiwari SK, Paspala SA, Khan AA. Current concept in neural regeneration research: NSCs isolation, characterization and transplantation in various neurodegenerative diseases and stroke: A review. J Adv Res 5 (2014): 277-94.
  39. Muheremu A, Shu L, Liang J, Aili A, Jiang K. Sustained delivery of neurotrophic factors to treat spinal cord injury. Transl Neurosci 12 (2021): 494-511.
  40. De Gioia R, Biella F, Citterio G, Rizzo F, Abati E, Nizzardo M, et al. Neural Stem Cell Transplantation for Neurodegenerative Diseases. Int J Mol Sci 21 (2020): 3103.
  41. Moehle MS, West AB. M1 and M2 immune activation in Parkinson's disease: foe or ally? Neuroscience. 302 (2015): 59-73.
  42. Bartels AL, Willemsen AT, Doorduin J, de Vries EF, Dierckx RA, Leenders, KL. PK- quantification of neuroinflammation and a monitor of anti-inflammatory treatment in Parkinson's disease? Parkinsonism Disord 16 (2010): 57-59.
  43. Edholm ES, Rhoo KH, and Robert J. Evolutionary aspects of macrophages polarization. Results Probl. Cell Differ 62 (2017): 3-22.
  44. Barrientos S, Stojadinovic O, Golinko MS, Brem H, and Tomic-Canic M. Growth factors and cytokines in wound healing. Wound Repair Regen 16 (2018): 585-601.
  45. Yan A, Zhang T, Yang X, Shao J, Fu N, Shen F, et al. Thromboxane A2 receptor antagonist SQ29548 reduces ischemic stroke-induced microglia/macrophages activation and enrichment and ameliorates brain injury. Rep 6 (2016): 35885.
  46. Locati M, Mantovani A, and Sica A. Macrophage activation and polarization as an adaptive component of innate immunity. Immunol 120 (2013): 163-184.
  47. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et. al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41 (2014): 14-20.
  48. Gordon S, and Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity 32 (2010): 593- 604.
  49. Stranahan AM, Hao S, Dey A, Yu X, Baban B. Blood- brain barrier breakdown promotes macrophage infiltration and cognitive impairment in leptin receptor-deficient mice. J Cereb Blood Flow Metab 36 (2016): 2108-2121.
  50. Ballabh P, Braun A, Nedergaard M. The blood-brain barrier: an overview: structure, regulation, and clinical implications.  Dis16 (2004): 1-13.
  51. Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier.  Dis37 (2010): 13-25.
  52. Villaseñor R, Lampe J, Schwaninger M, et al. Intracellular transport and regulation of transcytosis across the blood-brain barrier. Cell Mol Life Sci76 (2019): 1081-1092.
  53. Mulvihill JJ, Cunnane EM, Ross AM, Duskey JT, Tosi G, Grabrucker AM. Drug delivery across the blood-brain barrier: Recent advances in the use of nanocarriers. Nanomedicine15 (2020): 205-214.
  54. Al-Jipouri A, Almurisi SH, Al-Japairai K, Bakar LM, Doolaanea AA. Liposomes or Extracellular Vesicles: A Comprehensive Comparison of Both Lipid Bilayer Vesicles for Pulmonary Drug Delivery. Polymers (Basel) 15 (2023): 318.
  55. Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 8 (2022): e09394.
  56. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, Samiei M, et. al. Liposome: classification, preparation, and applications. Nanoscale Res Lett 8 (2013): 102.
  57. Sriwidodo S, Umar AK, Wathoni N, Zothantluanga JH, Das S, Luckanagul JA. Liposome-polymer complex for drug delivery system and vaccine stabilization. Heliyon 8 (2022): e08934.
  58. Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine 1 (2006): 297-315.
  59. Jarvis JN, Lawrence DS, Meya DB, Kagimu E, Kasibante J, E. Mpoza E, et. al. Single-Dose Liposomal Amphotericin B Treatment for Cryptococcal Meningitis. N Engl J Med 386 (2022): 1109-20.
  60. Makadia HK, Siegel SJ. Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier. Polymers 3 (2011): 1377-1397.
  61. Dilliard SA, Siegwart DJ. Passive, active and endogenous organ-targeted lipid and polymer nanoparticles for delivery of genetic drugs. Nat Rev Mater8 (2023): 282-300.
  62. Diwan A, Chakraborty A, Tatake J, et. al. Broad Spectrum Antiviral Therapy can Supersede Vaccination Strategy for Combating COVID-19 Pandemic. Med J Clin Trials Case Stud 5 (2021): 298-304.
  63. Fariello RG. Safinamide. Neurotherapeutics4 (2007): 110-116.
  64. Bette S, Shpiner DS, Singer C, Moore H. Safinamide in the management of patients with Parkinson’s disease not stabilized on levodopa: A review of the current clinical evidence.  Clin. Risk Manag14 (2018): 1737-1745.
  65. Ferreira JJ, Lees A, Rocha JF, Poewe W, Rascol O, Soares-da-Silva P. Opicapone as an adjunct to levodopa in patients with Parkinson’s disease and end-of-dose motor fluctuations: A randomized, double-blind, controlled trial. Lancet Neurol15 (2016): 154-165.
  66. Scott LJ. Opicapone: A Review in Parkinson’s Disease. Drugs76 (2016): 1293-1300.
  67. Fields CR, Bengoa-Vergniory N, Wade-Martins R. Targeting Alpha-Synuclein as a Therapy for Parkinson’s Disease.  Mol. Neurosci12 (2019).
  68. Parmar M, Grealish S, Henchcliffe C. The future of stem cell therapies for Parkinson disease.  Rev. Neurosci21 (2020): 103-115.
  69. Chakraborty A and Diwan A. How and Why Macrophages are Connected with the Parkinson’s Disease: A Short Review to Develop a Therapeutic Strategy for PD J Biomedical Research and Clinical Reviews 7 (2022).

© 2016-2024, Copyrights Fortune Journals. All Rights Reserved