top of page

Updated: Jul 14

Technical Review Article | Open Access | Published 11th of July 2025

QbD In Solid Lipid Nanoparticles To Enhance Blood-Brain Barrier Permeation For Alzheimer’s Therapy


Sumanji Bala¹, Dr. Panner Selvam R.² , Department of Pharmaceutical Sciences, PES University, Bengaluru. | EJPPS | 302 (2025) https://doi.org/10.37521/ejpps30210

Abstract 

Alzheimer’s disease (AD) continues to pose a major therapeutic challenge, primarily due to the restricted ability of drugs to traverse the blood-brain barrier (BBB). Solid lipid nanoparticles have gained attention as effective nanocarriers capable of addressing this limitation, thanks to their favourable biocompatibility, capacity to improve drug stability, and potential for sustained drug release. This research investigates the application of the QbD framework in formulating SLNs aimed at enhancing BBB penetration and achieving targeted drug delivery for AD management. The finalized SLN formulation exhibited significantly improved permeability across the BBB, as evidenced by both in-vitro and in-vivo assessments. These findings suggest a promising advancement in AD therapeutics. Moreover, the use of QbD principles in the development process contributes to the creation of consistent, high-quality nanoparticle systems that meet current regulatory standards for nanomedicine.


Keywords: Alzheimer’s disease, Rivastigmine, Central nervous system, Dementia, Solid lipid nanoparticle, Nanotechnology.


Introduction


Dementia is a growing global health challenge, affecting approximately 55.2 million people, as noted in the WHO’s 2022 blueprint¹. Prevalence among those over 60 varies by region—2.9% in Southeast Asia, 6.5% in Europe, and 3.1–5.7% elsewhere. While overall cases are increasing, some high-income countries report a decline in incidence². By 2030, the number of affected individuals is projected to reach 78 million, with the economic burden—encompassing medical expenses, social services, and informal caregiving—expected to surpass US$ 2.8 trillion, significantly impacting patients, families, and healthcare systems. Alzheimer’s disease recognized as the most prevalent type of dementia, poses an increasingly significant global health burden. In the United States, it affects about 10.8% of adults aged 65 and older, with a yearly occurrence rate of approximately 1,275 new cases per 100,000 individuals³⁻⁴. Globally, AD is responsible for 60% to 70% of all dementia diagnoses, with an estimated 27 million people currently living with the condition⁵. The disease places immense emotional and financial strain on both families and society. It is pathologically characterized by two primary hallmarks: the accumulation of β-amyloid (Aβ42) peptides forming extracellular amyloid plaques, and the development of intracellular neurofibrillary tangles composed of abnormally phosphorylated tau proteins⁶⁻⁷. Additionally, β-amyloid deposition in cerebral blood vessels can lead to amyloid angiopathy, contributing to vascular degeneration, impaired blood flow, and an increased risk of intracerebral haemorrhage. These pathological mechanisms serve as targets for therapeutic development, yet drug discovery for AD has faced significant hurdles⁸. Tacrine, one of the early treatments, was withdrawn due to hepatotoxicity. Current pharmacological options including donepezil, rivastigmine, galantamine, memantine, and the combination drug Namzaric—offer symptomatic relief but do not halt disease progression and are associated with various side effects ⁹⁻¹⁴. Recently, novel disease-modifying therapies such as sodium oligomannate (GV-971), aducanumab, lecanemab, and donanemab have been developed to intervene in AD progression, though challenges remain regarding their efficacy and accessibility¹⁵⁻¹⁹. Lipid-based nanocarriers such as liposomes, microemulsions, nanoemulsions, and lipid nanoparticles, that have emerged as promising platforms for drug delivery, owing to their excellent biocompatibility, low toxicity, and capacity to encapsulate both water- and fat-soluble compounds without the need for organic solvents. Among these, solid lipid nanoparticles and nanostructured lipid carriers are particularly notable for enabling controlled drug release and site-specific delivery. These nanosystems are typically formed from a lipid matrix and stabilized with surfactants, with their performance and efficiency largely dependent on the choice and proportion of formulation components ²⁰. Owing to their lipophilic characteristics, lipid nanoparticles are capable of crossing physiological barriers, including the epidermis and the blood–brain barrier, without the necessity for supplementary surface alterations. The flexibility of lipid excipients enables the development of tailored formulations that optimize pharmacokinetic profiles and enhance therapeutic efficacy²¹⁻²². Critical considerations in formulation design include the lipid mixture’s melting point (which should be above body temperature to maintain nanoparticle integrity), drug solubility, and structural characteristics. Additionally, the choice and concentration of surfactants are essential in stabilizing the formulation by preventing aggregation and coalescence. Nanoemulsions and NLCs have emerged as effective carriers for delivering lipophilic drugs via nasal administration, offering protection against enzymatic degradation and mucociliary clearance²³⁻²⁴. Advancements such as the functionalization of surfaces with biological molecules like proteins or antibodies, along with the incorporation of thermosensitive and mucoadhesive polymers into hydrogel systems, significantly improve the therapeutic efficacy of these delivery platforms while reducing potential side effects. To ensure the development of safe, effective, and high-quality lipid-based nanosystems, regulatory authorities emphasize the implementation of a QbD framework, which facilitates systematic quality assurance through comprehensive risk management practices²⁵⁻²⁶. This review explores the application of the QbD framework in the development of solid lipid nanoparticles aimed at improving the delivery of therapeutic agents across the blood-brain barrier for the effective management of Alzheimer's disease.


SOLID LIPID NANOPARTICLES:

Since their introduction in 1991, solid lipid nanoparticles have emerged as a prominent drug and payload delivery system. Developed to overcome the limitations of traditional colloidal carriers like emulsions, liposomes, and polymeric nanoparticles, SLNs provide notable benefits such as enhanced stability, biocompatibility, and controlled drug release. These spherical nanoparticles typically range from 10 to 1,000 nm in size and are composed of natural or synthetic lipids. This composition allows them to effectively encapsulate both hydrophilic and lipophilic drugs, thereby improving bioavailability and minimizing systemic side effects²⁷. A major strength of SLNs lies in their ability to protect encapsulated drugs from degradation during transportation by resisting various biochemical and physicochemical challenges. Additionally, SLNs facilitate targeted and sustained drug release, enabling site-specific action with minimal off-target effects. This makes them a valuable alternative to liposomes, especially for drugs that require deeper tissue penetration and prolonged retention²⁸⁻²⁹. Their ability to bypass anatomical barriers and provide controlled drug delivery has led to their exploration for various administration routes, including intravenous and oral delivery. Despite their advantages, challenges such as lipid crystallization, potential drug expulsion during storage, and scalability constraints remain. Additionally, the limited availability of regulatory-approved natural and synthetic polymers and high production costs have hindered the widespread adoption of polymeric NPs. However, advancements in SLN formulation, including surface modifications, ligand conjugation, and hybrid approaches, have significantly improved their efficiency³⁰⁻³¹. The evolving landscape of SLN-based drug delivery continues to expand, with ongoing research focused on optimizing their physicochemical properties, improving loading capacity, and enhancing site-specific targeting. Their adaptability has also facilitated their application in nutraceuticals and other pharmaceutical fields, broadening their scope beyond conventional drug delivery³².


SLN IN THE CNS DRUG DELIVERY SYSTEM:

Despite significant progress in drug delivery systems for neurological disorders, achieving effective therapeutic outcomes continues to be hindered by challenges such as poor target specificity, limited bioavailability, and potential toxicity. Many existing strategies are still based on trial-and-error approaches and lack comprehensive effectiveness. Emerging research emphasises the promise of receptor-mediated drug delivery, which utilises nutrient transport pathways and ligand-receptor interactions to facilitate improved cellular uptake and targeted delivery³³. Drug molecules conjugated with carriers like nanoparticles and liposomes can exploit receptor-mediated internalization, facilitating targeted drug transport across the blood-brain barrier (BBB). Carrier-mediated drug delivery systems, including engineered pharmaceuticals integrating active drug compounds with nanocarriers, have shown promise in penetrating the BBB. Notably, lipophilic molecules under 400 Da can diffuse across the endothelial barrier, making lipid nanoparticles suitable drug carriers. These nanoparticles interact with the BBB, promoting site-specific drug delivery while overcoming anatomical barriers³⁴⁻³⁵. Their physicochemical properties, including sustained drug release and controlled particle size, further enhance therapeutic efficacy. Among them, biocompatible and biodegradable polymeric nanoparticles have emerged as valuable candidates for targeted drug delivery, though their clinical adoption remains limited due to regulatory constraints and cost-effectiveness concerns. Lipid-based nanoparticles, particularly solid lipid nanoparticles (SLNs), are gaining traction as alternatives to polymeric carriers³⁶⁻³⁷. SLN-based drug delivery has demonstrated improved drug accumulation in brain tissues, overcoming BBB permeability limitations. Effective BBB penetration requires high lipid solubility, a molecular weight below 400 Da, and non-substrate characteristics for active efflux transporters. SLNs facilitate drug transport across endothelial layers via transcytosis and endocytosis while modulating transmembrane efflux and loosening tight junctions to enhance permeability. Surfactant incorporation further aids drug transport by fluidizing endothelial lipid membranes. Advanced SLN-based approaches focus on surface modifications, functionalisation, or ligand coating to enhance cellular uptake³⁸. For instance, Dal Magro et al. successfully modified SLNs with apolipoprotein-E-derived peptides, demonstrating effective BBB penetration in vitro. SLNs contribute to brain drug delivery by stabilizing drugs against degradation, extending systemic circulation time, promoting translocation to the brain, and triggering receptor-mediated endocytosis³⁹. These characteristics allow SLNs to improve drug encapsulation, facilitate transport across the blood–brain barrier, extend drug residence time in the brain, and overcome P-glycoprotein-mediated efflux⁴⁰. Beyond CNS applications, SLNs offer advantages in topical and ocular drug delivery. Their lipid composition facilitates interactions with the stratum corneum, optimizing skin penetration by restructuring lipid layers. Additionally, SLNs enhance drug adhesiveness, surface contact, and permeability⁴¹.


QUALITY BY DESIGN:

Traditional methods of pharmaceutical development that depend heavily on end-product testing are increasingly considered outdated. In contrast, the QbD framework offers a more modern, science-based approach. QbD begins with clearly defined goals and emphasises a thorough understanding of both product and process parameters⁴². By incorporating scientific knowledge and risk-based decision-making, this methodology ensures consistent product quality and promotes ongoing improvement throughout the development lifecycle. Among the various components of QbD, experimental designs play a crucial role in maximising data output with minimal experimentation. Unlike conventional methods where quality is tested in the final product, QbD embeds quality at every stage—from formulation to production—enhancing reliability and regulatory compliance. A key innovation in this study is the integration of two modern methodologies and user-friendly equipment to optimise and design experiments within the QbD framework⁴³⁻⁴⁴. The optimised combination of lipid and surfactant in the formulation significantly improved entrapment efficiency, reduced particle size, and enhanced drug release, demonstrating superior performance over previous solid lipid nanoparticle (SLN) formulations. Additionally, the formulation process involved a reduced quantity of organic solvents, which were effectively eliminated using a rotary evaporator, reinforcing its safety profile⁴⁵. This highlights the advantages of QbD-based methods over traditional production techniques, where quality assurance depends on controlling raw materials and manufacturing conditions. In conventional quality-by-testing models, deviations from regulatory specifications necessitate reworking or discarding batches, leading to increased costs and inconsistencies in product safety. Regulatory bodies like the FDA and EMA have supported the implementation of QbD principles through the International Conference on Harmonization (ICH) guidelines (Q8–Q11). These guidelines focus on clearly outlining process goals, strengthening risk management, and establishing robust control strategies to support the development of consistently high-quality pharmaceutical products. Over past decades, the advancement of nanosystems has transformed pharmaceutical technology, with researchers increasingly leveraging QbD to develop nanocarrier-based drug delivery platforms. By incorporating QbD principles, manufacturers can streamline production, reduce costs, minimize variability, optimize process efficiency, and facilitate regulatory approvals. This strategy improves patient outcomes while upholding the highest standards of pharmaceutical safety, effectiveness, and quality⁴⁶⁻⁴⁷.


CHALLENGES IN DRUG DELIVERY TO CNS:

Current Alzheimer’s disease treatments are significantly constrained by the central nervous system's complex protective barriers. The primary obstacles include the blood-brain barrier (BBB), blood-leptomeningeal barrier (BLMB), and the blood–cerebrospinal fluid (CSF) barrier, all of which restrict drug access to the brain. Of these, the BBB is the most selective, allowing only a small fraction of therapeutic compounds to penetrate due to its tight junctions and active efflux mechanisms. Although strategies like chemical modification, nanocarriers, and invasive delivery methods have been explored to enhance brain drug delivery, their clinical effectiveness remains inconsistent and limited⁴⁸⁻⁴⁹.


THERAPEUTIC POTENTIAL OF NANOEMULSION AND NANOSTRUCTURED LIPID CARRIER-BASED FORMULATIONS IN THE MANAGEMENT OF ALZHEIMER’S DISEASE:

Malaiya et al. (2024) developed and optimized chitosan-coated nanostructured lipid carriers (CS-EST-NLCs) to improve intranasal delivery of 17β-oestradiol (EST) to the brain. Using a solvent-free melt-emulsification method followed by sonication and Box–Behnken design for optimization, the resulting formulation exhibited favourable properties, including a particle size of 115.64 nm, zeta potential of −26.5 mV, and an entrapment efficiency of 81.99%. Chitosan coating improved these parameters, shifting zeta potential to +34.07 mV and increasing EE to 84.39%. Structural studies confirmed amorphous drug dispersion, while in vitro release followed a biphasic pattern fitting the Korsmeyer-Peppas model. Stability was maintained for three months, and ex vivo evaluations confirmed safety. Behavioural studies demonstrated superior cognitive efficacy of CS-EST-NLCs over plain EST. These findings support CS-EST-NLCs as a promising, mucoadhesive, and biodegradable intranasal delivery system for Alzheimer’s disease management⁵⁰. In a 2021 study, Costa C.P. and colleagues aimed to optimize two diazepam-loaded nanostructured lipid carrier (NLC) formulations intended for nose-to-brain delivery, utilizing a QbD framework. The research focused on adjusting critical material attributes such as lipid-to-emulsifier ratios and refining process parameters, including the choice of production technique. Among the methods evaluated, high-pressure homogenization (HPH) emerged as the most effective, leading to the development of the F1C15 formulation, which possessed a negative surface charge. This formulation exhibited a particle size of 69.59 nm, a zeta potential of −23.50 mV, and an encapsulation efficiency of 96.6%. In contrast, the positively charged F2A8 formulation showed a larger particle size and higher cytotoxicity. F1C15 also demonstrated superior physical stability over a three-month period and provided a controlled drug release profile⁵¹.

In a 2022 study, Arora D. et al. formulated intranasal solid lipid nanoparticles of rivastigmine tartrate using the solvent-evaporation diffusion method. A central composite design was employed to assess the influence of lipid and surfactant concentrations on particle size, entrapment efficiency, and drug release. The optimized SLNs, composed of glyceryl monostearate and polysorbate 80, exhibited a particle size of 110.2 nm, 82.56% entrapment efficiency, and 94.86% drug release. Compatibility of formulation components was verified through DSC and FTIR analyses. Histopathological evaluation of sheep nasal mucosa indicated no signs of toxicity, supporting the safety of the formulation. Furthermore, ex vivo permeation studies showed that the SLNs offered enhanced nasal absorption compared to a conventional RT solution. Stability studies indicated good shelf life under different conditions. In vivo pharmacokinetic evaluations confirmed enhanced bioavailability and safety of the formulation. Overall, RT-loaded SLNs present a promising and efficient approach for intranasal drug delivery⁵². Khandale et al. (2025) formulated and optimized XH-loaded nanostructured lipid carriers (XH-NLCs) to enhance XH’s oral bioavailability and therapeutic efficacy in an Alzheimer’s disease rat model. The NLCs, prepared using high-pressure homogenization and probe sonication, exhibited ideal particle size, zeta potential, and drug entrapment. Structural studies confirmed the amorphous molecular dispersion of XH within the formulation. In vitro studies demonstrated a 4.93-fold increase in drug release, while in vivo pharmacokinetic analysis showed a 24.7-fold elevation in maximum plasma concentration (Cmax) and an 11.3-fold enhancement in brain penetration. Pharmacodynamic assessments revealed marked improvements in cognitive function, alongside reductions in neuroinflammation, oxidative stress, acetylcholinesterase (AChE) activity, and amyloid-beta (Aβ) levels. These findings suggest that XH-NLCs offer a promising nanocarrier-based strategy for the effective management of Alzheimer’s disease⁵³.

Chen et al. (2016) developed transferrin-coated nanoliposomes to enhance α-mangostin delivery to the brain for Alzheimer’s therapy. The modified liposomes showed slightly larger size, higher PDI, and more negative zeta potential. In vitro and in vivo studies confirmed improved BBB penetration and increased brain accumulation, with extended half-life and residence time⁵⁴.

Kaur et al. developed nanoemulsion-based intranasal delivery systems for Alzheimer’s drugs to address the drawbacks of oral administration. One formulation for donepezil featured a particle size of 65 nm, a PDI of 0.084, and a zeta potential of −10.7 mV, showing sustained release and good biocompatibility in vitro, with significantly enhanced brain uptake in vivo. In a separate study, a nanoemulsion for intranasal memantine delivery demonstrated a particle size of approximately 11 nm, a PDI of 0.080, and a zeta potential of −19.6 mV. This formulation showed 80% drug release in simulated nasal fluid with first-order kinetics, along with improved antioxidant activity and cell viability compared to standard memantine solutions⁵⁵⁻⁵⁶.

Anand et al. developed nanostructured lipid carriers loaded with rivastigmine hydrogen tartrate to enhance brain targeting and therapeutic outcomes in Alzheimer's-related dementia. Using a Box-Behnken design, they optimized sonication time, solid-to-lipid ratio, and emulsifier concentration, evaluating particle size and drug entrapment efficiency (EE) as key responses. Sonication time significantly reduced particle size (p = 0.001), while EE was notably influenced by emulsifier concentration (p = 0.004) and solid/lipid ratio (p = 0.002). The optimized formulation closely matched predicted values. Ex vivo studies confirmed sustained drug release, and aldicarb assay indicated improved brain uptake. RT-PCR analysis showed notable suppression of acetylcholinesterase 1 and 2 expression with NLCs. In vivo, treated subjects demonstrated better memory performance, highlighting the potential of NLC-based rivastigmine delivery for Alzheimer’s treatment⁵⁷.

Ahmad et al. formulated a naringenin-loaded nanoemulsion and an in-situ hydrogel to improve nose-to-brain delivery. Recognized for its antioxidant and anti-inflammatory effects, naringenin demonstrated promise in Alzheimer's therapy. The nanoemulsion exhibited an average droplet size of approximately 91 nm, while the hydrogel—comprising poloxamer 407 and chitosan—had a size near 98 nm, with chitosan inducing a zeta potential shift. Ex vivo and in vivo studies confirmed improved brain bioavailability and sustained drug release with the hydrogel. Behavioural and antioxidant assessments in rats showed superior outcomes for the hydrogel. No adverse effects were observed, confirming both formulations as safe and effective for brain targeting⁵⁸.

Rajput et al. formulated an in-situ hydrogel incorporating resveratrol-loaded nanostructured lipid carriers for intranasal administration to improve brain delivery. To address resveratrol’s poor stability and first-pass metabolism, the NLCs were combined with acyl gellan gum, enhancing both stability and mucoadhesive properties. Critical formulation and processing factors influencing particle size, drug loading, and encapsulation efficiency were optimized using Plackett–Burman and full factorial design approaches. The final formulation showed favourable characteristics and was non-toxic to nasal tissues. Pharmacokinetic studies in mice revealed significantly enhanced brain delivery compared to oral resveratrol administration⁵⁹.

Quercetin is a flavonoid recognized for its antioxidant, anti-inflammatory, and anticancer activities, has been incorporated into solid lipid nanoparticles composed of a tripalmitin/lecithin core and coated with a chitosan shell. This approach improved its solubility and cellular uptake in Caco-2 cells compared to its unformulated form. To enhance brain delivery, SLNs using compritol as the lipid matrix have also been developed, showing potential for treating neurodegenerative diseases like Alzheimer’s⁶⁰. In Alzheimer’s disease, quercetin has shown the ability to reduce oxidative stress, lipid peroxidation, neuronal loss, and amyloid-beta (Aβ) aggregation. Pinheiro et al. developed transferrin-functionalized SLNs and nanostructured lipid carriers to enhance delivery across the blood–brain barrier (BBB). These formulations exhibited particle sizes below 250 nm, a zeta potential around –30 mV, and high encapsulation efficiency (80–90%), with no observed cytotoxicity in hCMEC/D3 cells. NLCs exhibited superior permeability across the BBB model compared to SLNs. Moreover, in vitro studies indicated that transferrin-functionalized NLCs reduced Aβ fibril formation and aggregation, although in vivo validation is still required⁶¹.

Cunha et al. designed two rivastigmine-loaded nanostructured lipid carriers for intranasal brain delivery using a QbD strategy. The formulation process involved two stages: a central composite design assessed the effects of lipid and emulsifier ratios on critical quality attributes, while a Box-Behnken design refined process variables using ultrasound and high-pressure homogenization. Both models demonstrated high predictive accuracy (R² up to 1.0). The optimised NLCs exhibited favourable characteristics, including suitable particle size, low PDI, stable zeta potential, high drug encapsulation, and physiological pH and osmolarity. Drug release exhibited a non-Fickian mechanism, and the formulations maintained stability for up to 90 days. These outcomes highlight the promise of QbD-optimised NLCs for efficient intranasal delivery of rivastigmine, though in vivo studies are still required⁶². Additionally, natural phenolic compounds like resveratrol have shown potential in preventing β-amyloid aggregation, a major contributor to Alzheimer’s disease. Formulating resveratrol into solid lipid nanoparticles enhanced its stability, bypassed hepatic first-pass metabolism, and enabled effective brain delivery when conjugated with OX26 monoclonal antibody. Similarly, galantamine hydrobromide demonstrated successful blood–brain barrier penetration via SLNs. Additionally, RVG-9R peptide combined with BACE1 siRNA in SLN formulation facilitated efficient nasal delivery, enhancing neuronal uptake and improving Alzheimer’s outcomes. Polysorbate-80-coated piperine SLNs, administered at 2 mg/kg, also showed promise in targeting the brain for Alzheimer’s therapy, supporting the role of SLNs in advanced neurotherapeutic strategies⁶³⁻⁶⁴.


FUTURE RECOMMENDATION:

Future research on applying the QbD approach to solid lipid nanoparticles for Alzheimer’s therapy should focus on optimizing brain-targeted delivery. Special attention should be given to incorporating advanced targeting agents like specific peptides or monoclonal antibodies to enhance drug transport across the blood–brain barrier with greater accuracy and efficacy. Moreover, designing SLNs that can encapsulate and simultaneously deliver multiple therapeutic agents—including neuroprotectants, antioxidants, and gene-silencing constructs—may yield enhanced therapeutic efficacy due to their synergistic effects on the diverse pathological pathways of Alzheimer’s disease. Incorporating personalized medicine principles, such as individual genetic and pharmacogenomic profiles, into the QbD framework could enable the creation of customized formulations tailored to patient-specific needs. For successful clinical application, thorough in vivo assessments and long-term toxicity studies are crucial to validate the pharmacokinetics, safety, and overall performance of these formulations. Additionally, the development of eco-friendly, scalable, and economically viable manufacturing processes will be essential to meet regulatory requirements and ensure commercial feasibility. By addressing these strategic directions, the QbD approach can be further optimized for SLN-based drug delivery, paving the way for more effective and targeted treatments for Alzheimer’s disease.


Conclusion

Implementing a QbD approach in the development of solid lipid nanoparticles offers a strategic and scientific framework to enhance drug delivery across the blood-brain barrier—an essential advancement in Alzheimer’s disease therapy. By identifying and optimizing key formulation and process parameters, QbD ensures the production of stable, high-quality SLNs with consistent performance, reduced variability, and improved safety in treatment outcomes. Encapsulating therapeutic agents such as resveratrol, galantamine, and BACE1 siRNA within SLNs designed using QbD principles presents a promising avenue for targeted, effective, and patient-focused management of Alzheimer’s disease, paving the way for advanced nanotherapeutic interventions


Conflicts Of Interest Declaration

Nil


References


1. David S Knopman , Helene Amieva, Ronald C. Alzheimer disease. Nature Review. Disease Primers.2001; 13;7(1):33.

2. Querfurth, H. W. & LaFerla, F. M. Alzheimer’s disease. New England Journal of medicine. 2010 ;362, 329–344.

3. Alzheimer’s disease facts and figures. Alzheimers Dement. 2023; 19: 1598–1695.

4. Abdelnour C, Agosta F, Bozzali M, Fougère B, Iwata A, Nilforooshan R, Takada LT, Viñuela F, Traber M. Perspectives and challenges in patient stratification in Alzheimer’s disease. Alzheimer's research & therapy. 2022; 14, 112.

5. de Abreu ID, Forlenza OV, DE BARROS HL. Alzheimer disease: correlation between memory and autonomy. Archives of Clinical Psychiatry. 2005 ;32(3):131–6.

6. Shinohara M, Sato N, Shimamura M, Kurinami H, Hamasaki T, Chatterjee A, Rakugi H, Morishita R. Possible modification of Alzheimer's disease by statins in midlife: interactions with genetic and non-genetic risk factors. Frontiers in aging neuroscience. 2014; 6:71.

7. Cortes-Canteli M, Paul J, Norris EH, Bronstein R, Ahn HJ, Zamolodchikov D, Bhuvanendran S, Fenz KM, Strickland S. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer's disease. Neuron. 2010;66(5):695–709.

8. Davis, K. L. & Powchik, P. Tacrine. Lancet.1995; 345, 625–630.

9. de Los Ríos, C. & Marco-Contelles, J. Tacrines for Alzheimer’s disease therapy. III. The PyridoTacrines. European journal of medicinal chemistry.2019; 166, 381–389.

10. Birks, J. S. & Harvey, R. J. Donepezil for dementia due to Alzheimer’s disease. Cochrane Database of systematic reviews.2018; 6, CD001190.

11. Feldman, H. H. & Lane, R. Rivastigmine: a placebo-controlled trial of twice daily and three times daily regimens in patients with Alzheimer’s disease. Journal of Neurology, Neurosurgery & Psychiatry.2007; 78, 1056–1063.

12. Coyle, J. & Kershaw, P. Galantamine, a cholinesterase inhibitor that allosterically modulates nicotinic receptors: effects on the course of Alzheimer’s disease. Biological psychiatry.2001; 49, 289–299.

13. Robinson, D. M. & Keating, G. M. Memantine: a review of its use in Alzheimer’s disease. Drugs. 2006; 66, 1515–1534.

14. Greig, S. L. Memantine ER/Donepezil: A Review in Alzheimer’s Disease. CNS Drugs 29, 963–970 (2015). Benek, O., Korabecny, J. & Soukup, O. A Perspective on Multi-target Drugs for Alzheimer’s Disease. Trends in pharmacological sciences. 2020;41, 434–445.

15. Xiao, S. et al. A 36-week multicentre, randomized, double-blind, placebo-controlled, parallel-group, phase 3 clinical trial of sodium oligomannate for mild-to-moderate Alzheimer’s dementia. Alzheimers Res.2021; Ther. 13, 62.

16. Cummings, J. & Salloway, S. Aducanumab: Appropriate use recommendations. Alzheimers Dement .2022;18, 531–533.

17. Van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, Kanekiyo M, Li D, Reyderman L, Cohen S, Froelich L. Lecanemab in early Alzheimer’s disease. New England Journal of Medicine.2023; 388 (1), 9–21.

18. Sims JR, Zimmer JA, Evans CD, Lu M, Ardayfio P, Sparks J, Wessels AM, Shcherbinin S, Wang H, Nery ES, Collins EC. Donanemab in Early Symptomatic Alzheimer Disease: The TRAILBLAZER-ALZ 2 Randomized Clinical Trial. Jama.2023;330 (6), 512–527.

19. Cheng, Q.; Wei, T.; Farbiak, L.; Johnson, L.T.; Dilliard, S.A.; Siegwart, D.J. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nature nanotechnology. 2020; 15(4), 313–320.

20. Di Filippo, L.D.; Duarte, J.L.; Azambuja, J.H.; Mancuso, R.I.; Luiz, M.T.; Araújo, V.H.S.; Figueiredo, I.D.; Barretto-De-Souza, L.; Sábio, R.M.; Sasso-Cerri, E. Baviera AM. Glioblastoma multiforme targeted delivery of docetaxel using bevacizumab-modified nanostructured lipid carriers impair in vitro cell growth and in vivo tumor progression. International journal of pharmaceutics. 2022; 618, 121682.

21. Akbari, J.; Saeedi, M.; Ahmadi, F.; Hashemi, S.M.H.; Babaei, A.; Yaddollahi, S.; Rostamkalaei, S.S.; Asare-Addo, K.; Nokhodchi, A. Solid lipid nanoparticles and nanostructured lipid carriers: A review of the methods of manufacture and routes of administration. Pharmaceutical development and technology. 2022; 27(5), 525–544.

22. Hsu, S.-H.; Wen, C.-J.; Al-Suwayeh, S.A.; Yen, T.-C.; Fang, J.-Y.; Kim, J.-H. Physicochemical characterization and in vivo bioluminescence imaging of nanostructured lipid carriers for targeting the brain: Apomorphine as a model drug. Nanotechnology ,2010; 21, 499802.

23. Galvão, J.G.; Santos, R.L.; Silva, A.R.; Santos, J.S.; Costa, A.M.; Chandasana, H.; Andrade-Neto, V.V.; Torres-Santos, E.C.; Lira, A.A.M.; Dolabella, S.; Scher R. Carvacrol loaded nanostructured lipid carriers as a promising parenteral formulation for leishmaniasis treatment. European Journal of Pharmaceutical Sciences. 2020;150, 105335.

24. Theochari I, Xenakis A, Papadimitriou V. Nanocarriers for effective drug delivery. In: Nguyen-Tri P, Do TO, Nguyen TA, editors. In smart Nanocontainers. Elsevier:2020;315–341.

25. Singh AK, Yadav TP, Pandey B, Gupta V, Singh SP. Engineering Nanomaterials for Smart Drug Release: recent Advances and Challenges. In: Mohapatra M, Ranjan S, Dasgupta N, Mishra R, Thomas S, editors. Applications of targeted nano drugs and delivery systems. Elsevier: 2019;411–449.

26. Mukherjee S, Ray S, Thakur R S. Solid lipid nanoparticles: A modern formulation approach in the drug delivery system. Indian journal of pharmaceutical sciences. 2009;71(4):349–58.

27. Choonara YE, Pillay V, Ndesendo VM, du Toit LC, Kumar P, Khan RA, Murphy CS, Jarvis DL. Polymeric emulsion and crosslink-mediated synthesis of super-stable nanoparticles as sustained-release anti-tuberculosis drug carriers. Colloids and Surface B Biointerfaces. 2011;87(2):243–54.

28. Akhtar, N., Mohammed, S.A., Khan, R.A., Yusuf, M., Singh, V., Mohammed, H.A., Al-Omar, M.S., Abdellatif, A.A., Naz, M. and Khadri, H. Self-generating nano-emulsification techniques for alternatively routed, bioavailability enhanced delivery, especially for anti-cancers, anti-diabetics, and miscellaneous drugs of natural, and synthetic origins. Journal of Drug Delivery Science and Technology. 2020; 58:101808.

29. Yusuf M, Khan M, Khan RA, Maghrabi IA, Ahmed B. Polysorbate-80-coated, polymeric curcumin nanoparticles for in vivo anti-depressant activity across BBB and envisaged biomolecular mechanism of action through a proposed pharmacophore model. Journal of microencapsulation. 2016;33(7):646–55.

30. Khateef R, Khadri H, Almatroudi A, Alsuhaibani SA, Mobeen SA, Khan RA. Potential in-vitro anti-breast cancer activity of green-synthesized silver nanoparticles preparation against human MCF-7 cell-lines. Advances in Natural Sciences: Nanoscience and Nanotechnology. 2019; 10(4):045012.

31. Akhtar N, Mohammed HA, Yusuf M, Al-Subaiyel A, Sulaiman GM, Khan RA. SPIONs Conjugate Supported Anticancer Drug Doxorubicin’s Delivery: Current Status, Challenges, and Prospects. Nanomaterials. 2020;12(20):3686.

32. Pardridge W.M. Drug transport across the blood–brain barrier. Journal of cerebral blood flow & metabolism. 2012; 32(11):1959–1972.

33. Anthony D.P., Hegde M.M., Shetty S.S., Rafic T., Mutalik S., Rao B.S. Targeting receptor-ligand chemistry for drug delivery across blood-brain barrier in brain diseases. Life sciences. 2021; 274:119326.

34. Jumaa M., Müller B.W. Lipid emulsions as a novel system to reduce the hemolytic activity of lytic agents: Mechanism of the protective effect. European journal of pharmaceutical sciences. 2000; 9:285–290.

35. Alavijeh M.S., Chishty M., Qaiser M.Z., Palmer A.M. Drug metabolism and pharmacokinetics, the blood-brain barrier, and central nervous system drug discovery. NeuroRx. 2005; 2:554–571.

36. Rakotoarisoa M., Angelova A. Amphiphilic nanocarrier systems for curcumin delivery in neurodegenerative disorders. Medicines. 2018; 5:126.

37. Gastaldi L, Battaglia L, Peira E, Chirio D, Muntoni E, Solazzi I, Gallarate M, Dosio F. Solid lipid nanoparticles as vehicles of drugs to the brain: current state of the art. European Journal of Pharmaceutics and Biopharmaceutics. 2014;87(3):433–44.

38. Singh I, Swami R, Pooja D, Jeengar MK, Khan W, Sistla R. Lactoferrin bioconjugated solid lipid nanoparticles: a new drug delivery system for potential brain targeting. Journal of drug targeting. 2016;24(3):212–23.

39. Dal Magro R, Ornaghi F, Cambianica I, Beretta S, Re F, Musicanti C, Rigolio R, Donzelli E, Canta A, Ballarini E, Cavaletti G. ApoE-modified solid lipid nanoparticles: A feasible strategy to cross the blood-brain barrier. Journal of Controlled Release. 2017; 249:103–10.

40. Garcês A, Amaral MH, Lobo JS, Silva AC. Formulations based on solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for cutaneous use: A review. European Journal of Pharmaceutical Sciences. 2018; 112:159–67.

41. Lawrence XY. Pharmaceutical quality by design: product and process development, understanding, and control. Pharmaceutical research.2008;25(4):781-91.

42. Pallagi E, Ambrus R, Szabó-Révész P, Csóka I. Adaptation of the quality by design concept in early pharmaceutical development of an intranasal nanosized formulation. International journal of pharmaceutics. 2015;491(1):384-92.

43. Zhang L, Mao S. Application of quality by design in the current drug development. Asian Journal of Pharmaceutical Sciences. 2017;12(1):1-8.

44. Martin‐Moe S, Lim FJ, Wong RL, Sreedhara A, Sundaram J, Sane SU. A new roadmap for biopharmaceutical drug product development: integrating development, validation, and quality by design. Journal of pharmaceutical sciences. 2011;100(8):3031-43.

45. Guideline, I.H.T., Pharmaceutical development. Q8 (2R).2009; As revised in August.

46. Guideline, I.H.T., Quality risk management. Q9, Current step, 2005; 4: p. 408.

47. Guideline, I.H.T., Pharmaceutical quality management. Q10, Current step, 2005; 4: p. 614.

48. Beccaria K, Canney M, Bouchoux G, Puget S, Grill J, Carpentier A. Blood-brain barrier disruption with low-intensity pulsed ultrasound for the treatment of pediatric brain tumors: a review and perspectives. Neurosurgical Focus. 2020;48(1): E10.

49. Pardridge WM. Blood-Brain Barrier and delivery of protein and gene therapeutics to brain. Frontiers in aging neuroscience. 2020; 11:373.

50. Malaiya A, Kenwat R, Mamgain A, Paliwal SR, Sulakhiya K, Maiti S, Paliwal R. QbD-based optimization and evaluation of chitosan-adorned nanostructured lipid carriers for nose-to-brain delivery of 17β-Estradiol in rat model of Alzheimer's disease. Journal of Drug Delivery Science and Technology. 2024 Jun; 96:105716.

51. Costa CP, Cunha S, Moreira JN, Silva R, Gil-Martins E, Silva V, Azevedo L, Peixoto AF, Lobo JS, Silva AC. QbD optimization of diazepam-loaded nanostructured lipid carriers (NLC) for nose-to-brain delivery: Toxicological effect of surface charge on human neuronal cells. International Journal of Pharmaceutics. 2021 Sep; 607:120933.

52. Arora D, Bhatt S, Kumar M, Verma R, Taneja Y, Kaushal N, Tiwari A, Tiwari V, Alexiou A, Albogami S, Alotaibi SS, Mittal V, Singla RK, Kaushik D, Batiha GE. QbD-based rivastigmine tartrate-loaded solid lipid nanoparticles for enhanced intranasal delivery to the brain for Alzheimer's therapeutics. Frontiers in aging neuroscience. 2022 Aug ;11; 14:960246.

53. Khandale N, Birla D, Alam MS, Bashir B, Vishwas S, Kumar A, et al. QbD endorsed fabrication of xanthohumol loaded solid nanostructured lipid carrier based powder for effective treatment of Alzheimer's disease in rats. Journal of Drug Delivery Science and Technology. 2025 May; 107:106792.

54. Chen ZL, Huang M, Wang XR, Fu J, Han M, Shen YQ, Xia Z, Gao JQ. Transferrin-modified liposome promotes α-mangostin to penetrate the blood-brain barrier. Nanomedicine. 2016 Feb;12(2):421-30.

55. Kaur A, Nigam K, Bhatnagar I, Sukhpal H, Awasthy S, Shankar S, Tyagi A, Dang S.. Treatment of Alzheimer’s diseases using donepezil nanoemulsion: an intranasal approach. In: Alonso MJ, editor. Drug Delivery and Translational Research. 2020;1–14.

56. Kaur A, Nigam K, Srivastava S, Tyagi A, Dang S. Memantine nanoemulsion: a new approach to treat Alzheimer’s disease. Journal of Microencapsulation. 2020;1–11.

57. Anand A, Arya M, Kaithwas G, Singh G, Saraf SA. Sucrose stearate as a biosurfactant for development of rivastigmine containing nanostructured lipid carriers and assessment of its activity against dementia in C. elegans model. Journal of Drug Delivery Science and Technology. 2019; 49:219–226.

58. Ahmad N, Ahmad R, Ahmad FJ, Ahmad W, Alam MA, Amir M, Ali A. Poloxamer-chitosan-based Naringenin nanoformulation used in brain targeting for the treatment of cerebral ischemia. Saudi Journal of Biological Sciences. 2020;27(1):500–517.

59. Rajput AP, Butani SB. Resveratrol anchored nanostructured lipid carrier loaded in situ gel via nasal route: formulation, optimization and in vivo characterization. Journal of Drug Delivery Science and Technology. 2019; 51:214–223.

60. Salehi B, Machin L, Monzote L, Sharifi-Rad J, Ezzat SM, Salem MA, Merghany RM, El Mahdy NM, Kılıç CS, Sytar O, Sharifi-Rad M. Therapeutic potential of quercetin: New insights and perspectives for human health. ACS Omega. 2020; 5:11849–72.

61. Pinheiro RG, Granja A, Loureiro JA, Pereira MC, Pinheiro M, Neves AR, Reis S. Quercetin lipid nanoparticles functionalized with transferrin for Alzheimer’s disease. European Journal of Pharmaceutical Sciences. 2020; 148:105314.

62. Cunha S, Costa CP, Loureiro JA, Alves J, Peixoto AF, Forbes B, Sousa Lobo JM, Silva AC. Double optimization of rivastigmine-loaded Nanostructured Lipid Carriers (NLC) for nose-to-brain delivery using the QbD approach: formulation variables and instrumental parameters. Pharmaceutics. 2020;12(7):599.

63. Rassu G, Soddu E, Posadino AM, Pintus G, Sarmento B, Giunchedi P, Gavini E. Nose-to-brain delivery of BACE1 siRNA loaded with solid lipid nanoparticles for Alzheimer’s therapy. Colloids and Surfaces B: Biointerfaces. 2017; 152:296–301.

64. Yusuf M, Khan M, Khan RA, Ahmed B. Preparation, characterization, in vivo and biochemical evaluation of brain targeted Piperine solid lipid nanoparticles in an experimentally induced Alzheimer’s disease model. Journal of drug targeting. 2013;3(21):300–11.


Author Information


Authors: Sumanji Bala¹, Dr. Panner Selvam R.²


01. Research Scholar, Department of Pharmaceutical Sciences, PES University,

02. Dr. Panner Selvam R. Associate Professor, Department of Pharmaceutical Sciences, PES University, PESU ECC Main Rd, Konappana Agrahara, Electronic City, Bengaluru, Karnataka 560100.


Corresponding Author:

Sumanji Bala, Research Scholar, Department of Pharmaceutical Sciences, PES University

Address: Postal Address: PESU ECC Main Rd, Konappana Agrahara, Electronic City, Bengaluru, Karnataka 560100.

Telephone: 9036968274

 
 
 

Comments


bottom of page