Int J App Pharm, Vol 16, Issue 5, 2024, 38-45Review Article

CURRENT PERSPECTIVES ON USING NANOPARTICLES FOR DIABETES MANAGEMENT

NITESH KUMAR YADAV, RUPA MAZUMDER*, ANJNA RANI, ARVIND KUMAR

Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh-201306, India
*Corresponding author: Rupa Mazumder; Email: rupa_mazumder@rediffmail.com

Received: 08 Apr 2024, Revised and Accepted: 02 Jul 2024


ABSTRACT

If ignored, Diabetes Mellitus (DM), a chronic metabolic disease marked by high levels of blood glucose, can have serious negative effects on one's health. The efficacy, safety, and patient compliance of traditional treatment approaches, like insulin injections and oral medications, are frequently hampered. Nanoparticle-based methods have shown promise in recent years as improved diabetes management techniques. Enhanced bioavailability, prolonged therapeutic effects, and targeted drug delivery are just a few of the special benefits that come with using nanoparticles. An overview of current perspectives on using nanoparticles for diabetes control is given in this review. The properties, production processes, and potential uses of several types of nanoparticles, such as polymeric, lipid-based, and inorganic nanoparticles, in the management of diabetes are covered. These nanoparticles allow for the precise delivery of therapeutic agents, such as insulin or anti-diabetic medications, to specific target tissues, like the liver or pancreas. It discusses how inorganic nanoparticles, Polymeric Nanoparticles (PNPs), and Lipid-Based Nanoparticles (LNPs) contribute to improved drug solubility, targeted delivery, and controlled release. Several methods for synthesizing polymeric nanoparticles are described. It also discusses the potential anti-inflammatory and antioxidant properties of some nanoparticles and how crucial they are to lowering diabetes-related issues. By incorporating the most recent research, this review offers a comprehensive summary of the current developments in the use of nanoparticles for diabetes control, paving the way for enhanced therapeutic outcomes and tailored interventions.

Keywords: Diabetes mellitus, Polymeric nanoparticles, Lipid-based nanoparticles, Inorganic nanoparticles


INTRODUCTION

Diabetes Mellitus (DM) is a complex metabolic disorder marked by persistent high blood glucose levels (hyperglycemia) caused by anomalies in the secretion or action of insulin. It involves chronic and diverse dysfunctions in carbohydrate, fat, and protein metabolism, following a progressive and heterogeneous pattern [1, 2]. There are mainly two types of DM. Although there is a strong hereditary component to type 1 DM (T1DM), environmental factors are also believed to be involved in the etiology of the disease. The autoimmune destruction of β cells may be triggered by these factors, or they may quicken a continuing process. Though conclusive causality is yet unknown, viral illnesses such as enteroviruses, Cytomegalovirus, coxsackie virus B, mumps, and rubella have been linked to T1DM [3]. Type 2 Diabetes (T2D), is characterized by a progressive dysfunction in insulin secretion coupled with insulin resistance [4]. It constitutes about 90% of all cases of diabetes [5]. Aging, obesity, a family history of diabetes, physical inactivity, and embracing contemporary lifestyles have all been linked more frequently to T2D [2]. The United States, India, Brazil, China, the United Kingdom, Russia, Algeria, Saudi Arabia, Nigeria, and Germany have the highest rates of new T1DM diagnoses among children under the age of fifteen (around 96,000 cases yearly), based on information made public by the International Diabetes Federation (IDF) in 2017. This represented about 60% of all new cases [6]. By 2030, the number of individuals with T2D globally is estimated to make it to 7079 per 100,000, highlighting persistent growth in all parts of the world. Concerning indications indicate that prevalence is on the rise in lower-income nations [7]. Diabetes is more than just a condition marked by elevated blood sugar. It usually has a great deal of problems. Hyperosmolar nonketotic coma and hyperglycemic acidosis are two acute effects of uncontrolled diabetes. Many tissue injuries that result in cardiovascular disorders, renal failure, eye damage, seizures, podiatric ulcers, compromised immunity, and reduced eyesight are among the long-term effects. Several other dysfunctions have been recognized, such as imbalances in electrolytes, high weight, organ inflammation, stroke, lipid abnormalities, and peripheral coronary diseases [8, 9]. DM affects millions of people, and its effects go beyond personal health to put a burden on economics and healthcare systems around the world. There is a need for immediate clinical preventative and public health actions [10].

Even with the extensive use of several antidiabetic medications for diabetes control, a full and effective recovery is still unattainable. Problems include the danger of hypoglycemia, difficulty in intelligently altering dose forms depending on glucose fluctuations, and insufficient drug concentrations in target locations owing to chemical instability and vulnerability to degradation. In addition, there are obstacles such as limited bioavailability, short plasma half-life, restricted therapeutic window, short absorption efficiency, and inadequate patient compliance [11]. It is well recognized that taking insulin orally causes limited bioavailability and inadequate therapeutic impact because of the polypeptide's physiological instability, which is caused by the Gastrointestinal Tract’s (GIT) enzymatic and chemical breakdown coupled with its rapid systemic clearance [12]. Improving diabetes treatment requires addressing these problems. Herein lies the pivotal role of nanoparticles in revolutionizing diabetes treatment. Because of their distinctive biological, physical, optical, chemical, and magnetic characteristics, nanoparticles are useful for diagnostic applications, especially in glucose biosensors that detect diabetes early. One common enzyme in biosensors, Glucose Oxidase (GO), has difficulties because of its low electron transfer efficiency. To improve the efficacy of enzyme-based biosensors, nanomaterials with greater conductivity, such as graphene and Gold Nanoparticles (AuNPs), are being investigated. Nanoparticles such as graphene, AuNPs, Cadmium Telluride (CdTe) quantum dots, and carbon nanotubes have been used in several studies, showing enhanced sensitivity and better electron transport characteristics in glucose biosensors. Notably, protein nanoparticles exhibit attomolar sensitivity and function as ultrasensitive probes for the detection of disease indicators, including autoantibodies linked to T1DM. The use of nanoparticles in biosensors can progress in the area of diagnosis allowing early management of diabetes [13]. Drug delivery methods using nanoparticles are essential for increasing the bioavaliability and bioactivity of pharmaceuticals. These multipurpose nano-formulations can be used to treat illnesses ranging from minor ailments to life-threatening conditions, as well as in nanoscale Drug Delivery Systems (DDS), nanomachines, and nanorobots. By addressing drug absorption, distribution, metabolism, and excretion, nanoparticles dramatically improve pharmacokinetic parameters and improve therapeutic results. Enhanced bioavailability and solubility, prolonged drug release, targeted distribution, lower dose needs, and fewer adverse effects are just a few of the notable benefits of nanoparticles [14]. Because of their specific reaction to certain stimuli and ability to release their payloads solely at the region of interest, stimuli-responsive nanoparticles formulations have demonstrated great promise in addressing issues related to premature or off-target drug release [15]. Enhancing the transport of nanoparticles across the layer of epithelium can be achieved by receptor-mediated endocytosis, specifically through transcytosis. Ligand functionalization promotes particular receptor recognition, which increases cellular absorption. Examples of ligands that do this include vitamin B12, Apical Sodium-Dependent Bile Acid Transporter (ASBT), Neonatal Fc Receptor (FcRn), CSKSSDYQC (CSK) peptide, folic acid, transferrin, butyrate, and zwitterions [16] (fig. 1). Combination therapy can be made possible by the ability of nanoparticles to co-deliver multiple medications simultaneously, addressing various areas of diabetic pathology at once [17]. Personalized medicine in the treatment of diabetes is made possible by nanoparticles, which allow for the customization of DDS to best suit the needs of each patient [18]. Nanoparticles may reduce the need for invasive procedures by offering alternative techniques for administering drugs, including buccal or transdermal delivery [19, 20]. Inhaled insulin is administered directly into the bloodstream by inhalers. It is encapsulated in nanoparticles inside dry powder formulations. Insulin breakdown is avoided using this technique. It does, however, require patients to undergo frequent, somewhat expensive pulmonary function testing [21]. Nanoparticles are a promising new tool in regenerative medicine that can help deliver therapeutic molecules for tissue regeneration and repair in issues associated with diabetes [22]. We thoroughly searched the databases in MEDLINE, Scopus, Science Direct, PubMed, and Google Scholar. Diabetes mellitus, polymeric nanoparticles, lipid-based nanoparticles, and inorganic nanoparticles were among the search phrases used. We went through more than 90 published papers from 2001 to 2024.

Fig. 1: Benefits of nanoparticles over conventional drug delivery system [23]

Polymeric nanoparticles

The term "Polymeric Nanoparticles (PNPs)" describes particles made of polymers at the nanoscale. Large molecules known as polymers are composed of monomers, which are repeating components. These polymers can produce nanoparticles with special characteristics and are used when they are organized at the nanoscale [24]. The main ingredient in PNPs is polymer, which is combined with other elements to create the particles. Natural and synthetic polymers are the two main kinds of polymers used in the formation of PNPs. Natural polymers, including gelatin, chitosan, albumin, sodium alginate, and synthetic polymers like polycaprolactone, Poly(Lactide Co-Glycolides) (PLGA), polyglycolides, Poly malic acid, Poly(methyl methacrylate) and polylactides are commonly used [25, 26]. Achieving adaptation goals such as biocompatibility, biodegradability, and guaranteeing non-antigenicity and non-toxicity depend heavily on the choice of polymers [27]. Researchers may modify the stability, targeting capabilities, and drug-release kinetics of PNPs by choosing certain polymers [28]. In the GIT, chitosan nanoparticles have mucoadhesive qualities and the ability to affect molecular processes such as lysosome degradation, claudin-4 redistribution, tight junction weakening, and enhanced paracellular permeability [29, 30]. Alginate is a pH-responsive and mucoadhesive polymer. The polymer's guluronic acid acts like a magnet for divalent ions, forming a gel-like net that captures and stores insulin within the nanoparticles through an ion exchange process [31]. PNPs exist in various forms such as Polymeric Nanospheres, Polymeric Nanocapsules, Polymeric Micelles, Polymeric Nanogels, Polymeric Dendrimers, Polymeric Nanorods, Polymeric Nanofibers [32, 33]. The target location, the kind of medication contained, and the intended drug release profile all influence the choice of PNPs. Researchers consistently investigate novel polymeric substances and compositions to enhance the effectiveness and stability of pharmaceutical delivery mechanisms [34, 35]. Some of the recent work on PNPs in the field of diabetes management is mentioned in table 1.

Synthesis methods for polymeric nanoparticles

Single emulsification method

It is the oldest technique for creating nanoparticles from preformed polymers and is particularly used for encapsulating hydrophobic compounds and drugs that are primarily insoluble in water. During emulsification and subsequent evaporation, the drug and polymer are dissolved together in an organic phase in this process, which helps to produce the interfaces that are essential for drug entrapment. But when it comes to hydrophilic drugs like peptides, proteins, and vaccinations, their effectiveness diminishes. The use of the emulsification technique for hydrophilic drugs is limited because it leads to quick diffusion of these compounds into the outer aqueous phase, which causes poor loading and low encapsulation efficiencies [36].

Double emulsion-solvent evaporation method (W1/O/W2)

With the addition of a crucial third emulsion phase for increased encapsulation efficiency and quick solidification, it is an improved method for the encapsulation of hydrophilic drugs. The process requires the evaporation of the organic solvent to transform the emulsion into a nanoparticle suspension. Because W/O/W emulsion is thermodynamically unstable, a rapid procedure is essential. Double-emulsion droplet rapid solidification greatly increases encapsulation efficiency. During the first water-in-oil emulsion, it is critical to quickly deposit a polymer barrier to maximize efficiency and inhibit drug penetration into the organic phase. This is accomplished by either raising the concentration of the stabilizer in the inner aqueous phase to enhance viscosity or employing a high concentration of a high molecular weight polymer in the oil phase. Recovering, cleaning, and lyophilizing the nanoparticles are steps in the finalization processes shown in fig. 2 [36].

Fig. 2: Preparation of polymeric nanoparticles by double solvent evaporation method [37]

Nanoprecipitation

A nanoprecipitation technique entails dissolving the drug and the polymer in a water-miscible organic solvent. Next, while stirring continuously, this solution is gradually added drop by drop to an aqueous phase. When there is water present, the solvent diffuses quickly, causing nanoparticles to develop on their own as shown in fig. 3 the benefit of this technique is its simplicity, which makes it a rather simple and scalable procedure. Precise control over the mixing process is a difficulty in traditional nanoprecipitation procedures, making it difficult to precisely regulate interactions during nanoparticles production. When compared to more sophisticated technologies like flash nanoprecipitation and microfluidic-based approaches, these techniques could produce bigger particle sizes with wider size distributions [38].

Fig. 3: Preparation of polymeric nanoparticles by nanoprecipitation technique [39]

Table 1: Recent advancements in PNPs in the management of DM

Polymers used Drug encapsulated Synthesis method Size (nm) Encapsulation efficiency % Effect in in vivo and in vitro Reference (s)
Polyethylene glycol (PEG) Insulin Self-assembly method of nanoprecipitation 91.06±2.01 53.94±0.05 Enhanced the stronger hypoglycemic response in diabetic rats, Achieved a remarkable 9.28% relative bioavailability [40]
Eudragit RSPO Alogliptin Nanoprecipitation 290.34±3.24 95.45±2.65 Enhanced oral bioavailability and anti-diabetic effect [41]
Eudragit Empagliflozin Emulsification solvent evaporation 270.03 95.82 Showed significant antidiabetic action in in vivo and in vitro studies. [42]
Chitosan and gum arabic Glycyrrhizin Ionotropic gelation 181.4 nm 99.8 Significant anti-diabetic effect [43]
Eudragit, tween 80 and Polyvinyl Alcohol (PVA) Sitagliptin Combined technique of solvent evaporation and nanoprecipitation 135.29±5.12 nm 82.34±3.27 Produce sustained and prolonged action, enhance the permeation across the intestinal mucosa, and successfully reduce the elevated blood sugar level [44]
Poly (ε-caprolactone), PVA Nateglinide Emulsion solvent evaporation 310.40±11.42 64.09± 4.27% [45]
Chitosan Polydatin Modified ionic gelation method 144.25±3.37 96.74±0.39% Prolonged release pattern, much greater antidiabetic effectiveness in diabetic rats compared to free Polydatin [46]
PLGA Diospyros melanoxylon Roxb. Emulsion solvent evaporation 365.7 nm 60.67%. [47]
Chitosan, Polyethylene Glycol (PEG), PLGA Pioglitazone Single emulsion solvent evaporation 813.25 48.93 Prolonged potency of nanoparticles in vivo than pure drug [48]
Conjugated Linoleic Acid (CLA) linked to Carboxymethyl Chitosan (CMCS forming CLA–CMCS (CC), Grafting Arginine to CC via amide bonding resulting in a novel CLA–CMCS–Arg (CCA) polymer. Insulin A combination of several techniques 203.4±3.42 83.78±3.73 Enhanced transdermal delivery, controlled release [49]
Acryloyl crosslinked dextran dialdehyde Human insulin 75 nm 48.68 Glucose-sensitive insulin release [50]
Waxy maize starch (>98% amylopectin) Insulin Combination of gelatinization and coacervation 100–300 69.73 Glucose-sensitive insulin release [51]

Lipid-based nanoparticles (LNPs)

Lipids and surfactants are the main constituents of LNPs, while occasionally other functional molecules are also present. The structural framework of the nanoparticles is formed by lipids, which act as the building blocks. Surfactants help to stabilize the nanoparticles, keep them from aggregating, and make sure they are distributed uniformly. To impart certain qualities, such as ligand targeting for targeted distribution, additional functional molecules may be added [52]. Active Pharmaceutical Ingredients (API) loading in a lipid system can improve solubility in water, leading to high bioavailability. Additionally, lipid-based systems prevent API oxidation, breakdown, and decomposition while improving delivery and storage [53]. LNPs provide easy surface modification and tailored distribution via many pathways. Pharmaceutical research is becoming more interested in them because they offer stability, less toxicity, and controlled release. The in vivo destiny of LNPs is determined by their size, composition, and surface features [54]. There are various types of lipid-based DDS namely liposomes, niosomes, Solid Lipid Nanoparticles (SLNs), Nanostructured Lipid Carriers (NLCs), cubosomes, exosomes, etc. each of them having specific advantages over others [52, 55-57].

Liposomes

Hydrophilic as well as hydrophobic drugs can be encapsulated in liposomes, which are spherical structures with an aqueous space within and hydrophobic lipid bilayers outside that protect the drugs from enzyme destruction. Similar to cell membranes, their bilayer shape improves absorption by intestinal cells. There are three varieties of liposomes: cationic, anionic, and neutral. Because of their hydrophilic and electroneutral surface, neutral liposomes are better at penetrating mucus [16].

Solid lipid nanoparticles

SLNs are 50–1000 nm-sized colloidal DDS. They consist of a solid biodegradable lipid core made up of fatty acids, complicated glyceride mixes, and mono-, di-, or triglycerides. An exterior aqueous surfactant dispersion of Tween 80, poloxamers, soy lecithin, or sodium dodecyl sulfate stabilizes the lipid core [57, 58]. The use of physiological lipids, the avoidance of organic solvents, a potentially broad application range (intravenous, pulmonary, cutaneous), sustained drug release, better stability, better % entrapment efficiency and Drug Loading percentage (%DL), compatibility with hydrophilic and lipophilic drugs, biocompatibility and biodegradability, cost-effectiveness, and high-pressure homogenization as a tried-and-true manufacturing process are all benefits of SLN [59-61].

Nanostructured lipid carriers

With a distinct lipid matrix nanostructure, a new class of NLCs has surfaced that improves drug loading and maintains stability throughout storage. This technique, which produces lipid particle dispersions with solid concentrations ranging from 30% to 80%, involves high-pressure homogenization. By combining liquid and solid lipids, the novel method produces a matrix that has a lower melting point yet is still solid at body temperature [59]. Three types of NLCs exist Type I has an imperfect crystal core that hinders drug expulsion; Type II has an amorphous, structureless core because the solid lipid is polymorphic; and type III is an oil-in-solid fat-in-water (O/F/W) system that works well for drugs that are more soluble in liquid lipid/oil than in solid lipid. Because of their unique structures and compositions, NLCs minimize problems that are related to SLNs and provide better stability, reduced drug ejection, and greater drug loading [62, 63].

Cubosomes

Cubosomes are nanocarriers with bicontinuous cubic phases formed by dispersing cubic liquid crystalline aggregates in an aqueous medium. Using amphiphilic lipids like glycerol monooleate and phytantriol, cubosomes self-assemble in an aqueous medium to form structures that resemble honeycombs and range in size from 100 to 500 nm. Cubosomes are noteworthy for having a large surface area and a microstructure that is identical to parent cubic aggregates. This makes them promising for several uses, such as drug delivery and nanotechnology [64].

Niosomes

Niosomes are nonionic surfactant-based vesicles. The structure and physical characteristics of niosomes are comparable to those of liposomes. Additionally, they are prepared as single-or multilamellar vesicles using the same techniques and conditions [65]. Despite having similarities niosomes are an alternative to liposomes because of their advantages over the latter. Given that the phospholipids that make up liposomes are chemically fragile, niosomes have superior chemical stability. Niosomes are less expensive than liposomes and don't require the same handling, storage, or purification procedures for phospholipids as liposomes do [66]. Table 2 presents recent advancements in LNPs within the realm of diabetes management, including the type of drug encapsulated, other components utilized, preparation methods, and key advantages.

Table 2: Recent advancements: characteristics and advantages of lipid-based nanoparticles

Lipid-based NP type Drug encapsulated Other component used Preparation method Key advantages Reference (s)
Thiamine/nonmodified liposome Recombinant human insulin Soybean phosphatidylcholine, cholesterol, thiamine, niacin Reversed-phase evaporation 72-81% glucose level reduction [67]
Nano liposome Recombinant Cas9-RNP complex Lecithin, cholesterol, 1,2-dioleoyl-sn-glycerol-3-[(N-(-5-Amino-1-Carboxypentyl) Iminodiacetic Acid) Succinyl} (Nickel salt) DOGS-NTA-Ni Lipid film formation, fluorescent labeling Successful gene editing with low cytotoxicity, stable in vivo delivery [68]
Solid lipid nanoparticles Gliclazide Glyceryl behenate, poloxamer 188 Ultra-sonication technique Biphasic in vitro release consisting of a prolonged release phase after an initial burst effect, a 5-fold increase in GLZ oral BA loaded in slns, better anti-diabetic action [69]
Pegylated slns Metformin hydrochloride Phospholipon®, sorbitol, polyethylene glycol 4000, beeswax, Tween® 80 Fusion method Pegylated slns showed greater diabetic control than the commercial formulation (Glucophage ®) after 24 h. [70]
SLN suspensions Extracts of P. Acaciae and P. Curviflorus Sodium Dodecyl Sulfate (SDS) Emulsion solvent evaporation Better antihyperglycemic and antioxidant activities. [71]
SLNs Murraya koenigii leaves extract (murrayanol) Solvent diffusion method Prolonged release, superior to commercially available synthetic anti-diabetic medications. [72]
SLNs Tetrahydrocurcumin Glyceryl monostearate, soy lecithin, tween 80 Emulsification followed by sonication Rapid burst at first followed by control release, greater bioavailability, and antidiabetic action than ordinary drug dispersion [73]
Cumbersome Gliclazide Glyceryl monooleate, poloxamer 407 Emulsification method A doubled up on the bioavailability relative to plain gliclazide suspension [56]
Niosomes Metformin hcl and glipizide Tween 80, phosphate buffer saline, cholesterol, chloroform, methanol Thin film hydration Sustained release of the drugs [55]

Inorganic nanoparticles

Inorganic nanoparticles are nanoscale particles composed of non-carbon-based elements, including metals, metal oxides, and other inorganic materials [74]. Because of their inherent qualities, which are unavailable and unpossessed in their conventional polymer-based or organic counterparts, such as tunable morphology, desirable physiological stability, simple functionalization, and unique physiochemical properties like optical, acoustic, electrical, and magnetic natures, inorganic nanoparticles among these designs hold great promise [75]. Within the realm of medicine, inorganic nanoparticles are being investigated for targeted drug delivery. This approach involves encasing or attaching drugs to the surface of the nanoparticles, so enabling controlled release and improved therapeutic effectiveness [76]. Several inorganic elements have demonstrated antidiabetic properties in vitro and/or in vivo. These elements include silver, iron, magnesium, zinc, copper, chromium, selenium, vanadium, palladium, platinum, nickel, titanium, gold, molybdenum, cerium, manganese, and tungsten. They do this through a variety of mechanisms, such as increasing levels of antioxidant enzymes, glucose utilization, and insulin sensitivity [77]. Inorganic nanoparticles, including zinc oxide and silver nanoparticles, display antibacterial characteristics that can help in wound healing, particularly significant for diabetes patients prone to delayed wound healing [78]. Some inorganic nanoparticles such as AUNP have antiangiogenic properties, which might affect the development of new blood vessels while plant-mediated synthesis of silver nanoparticles possesses potential antimicrobial applications and helps control diabetic retinopathy [79, 80]. Table 3 provides an overview of Inorganic nanoparticles, detailing their synthesizing method, size in nanometers (nm), surface functionalization, and antidiabetic mechanism.

Table 3: Characteristics of inorganic nanoparticles and their role in antidiabetic mechanisms

Inorganic nanoparticles Synthesizing method Size (nm) Surface functionalization Antidiabetic mechanism Reference (s)
AuNPs Bioreduction of auric chloride using phytoconstituents present in eclipta alba (green synthesis) 26.6 Eclipta alba Prevent β-cell damage induced by streptozotocin in rin-5f cells. [81]
Silver nanoparticles Green synthesis 250–800 Galaxaura Elongata {GE}, Turbinaria Ornata {TO} and Enteromorpha Fexuosa {EF} [82]
Mesoporous silica nanoparticles The Stober method with silane polymerization 50–130 Morin Compared to traditional inhibitors, Morin interacts with alpha-amylase and alpha-glucosidase more well at the molecular level. [83]
Selenium nanoparticles Chemical synthesis 50 Catathelasma ventricosum polysaccharides Improve cholesterol levels, blood sugar, antioxidant enzyme activity, and body weight. [84]
Zinc oxide nanoparticles Solution combustion synthesis 29 Areca catechu leaves extract Inhibit carbohydrate digestive enzymes [85]
Silver nanoparticles Green synthesis 34 Seed extract of N. Sativa Increased inhibition of Dipeptidyl peptidase-IV, α-glucosidase, and α-amylase [86]
Silver nanoparticles Green synthesis >100 Cucumis melo L. Leaf Inhibition of α-amylase and α-glucosidase [87]
Mesoporous silica nanoparticles Chemical synthesis 120 Insulin, camp Glucose triggered the release of insulin and camp [88]

CONCLUSION

The fascinating potential of nanotechnology to transform the treatment of diabetes has been examined in this paper. Conventional treatments frequently include drawbacks such as limited absorption, dosing difficulties, and the possibility of hypoglycemia. Targeted distribution, controlled release, and improved drug solubility are among the special qualities that nanoparticles provide, overcoming these drawbacks and opening the door to more efficient and individualized treatment modalities.

The review demonstrated the noteworthy contributions of polymeric, lipid-based, and inorganic nanoparticles, offering encouraging directions for better therapeutic interventions. Liposomes, SLNs, and NLCs are useful tools in the lipid-based approach, and methods like double emulsion-solvent evaporation and single emulsification show promise for the controlled synthesis of PNPs. Interestingly, in addition to helping transport drugs, inorganic nanoparticles may also have anti-inflammatory and antioxidant qualities that may reduce the complications associated with diabetes.

Even though there has been a lot of development, further study is necessary to improve nanoparticles systems, handle any safety issues, and convert preclinical results into clinical uses. Scalability, long-term safety profiles, and regulatory obstacles must all be addressed if this technology is to reach its full potential. However, the current developments show promise for the use of nanotechnology to provide effective and individualized diabetes management in the future. This could lead to better treatment outcomes, fewer complications from diabetes, and eventually, a higher standard of living for the millions of people who suffer from this chronic illness.

ACKNOWLEDGEMENT

The authors express their gratitude to the faculty members of the Noida Institute of Engineering and Technology (Pharmacy Institute) and other supporting staff members for their invaluable contributions to the preparation of the review article.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

Nitesh Kumar Yadav conceptualized and initially wrote the paper. Rupa Mazumder, the corresponding author, was responsible for the design and final revision. Anjna Rani handled data collection and interpretation, while Arvind Kumar performed the quality check on the manuscript.

CONFLICT OF INTERESTS

There is no conflict of interest among the authors of the review article.

REFERENCES

  1. Deepthi B, Sowjanya K, Lidiya B, Bhargavi R, Babu P. A modern review of diabetes mellitus: an annihilatory metabolic disorder. J In Silico In vitro Pharmacol. 2017;3(1). doi: 10.21767/2469-6692.100014.

  2. Banday MZ, Sameer AS, Nissar S. Pathophysiology of diabetes: an overview. Avicenna J Med. 2020;10(4):174-88. doi: 10.4103/ajm.ajm_53_20, PMID 33437689.

  3. Mekala KC, Bertoni AG. Epidemiology of diabetes mellitus. Transplant Bioeng Regen Endocr Pancreas. 2020:49-58.

  4. Arner P. Insulin resistance in type 2 diabetes: role of fatty acids. Diabetes Metab Res Rev. 2002;18Suppl 2:S5-9. doi: 10.1002/dmrr.254, PMID 11921432.

  5. Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. 2018;14(2):88-98. doi: 10.1038/nrendo.2017.151, PMID 29219149.

  6. Diamond Project Group. Incidence and trends of childhood type 1 diabetes worldwide 1990-1999. Diabet Med. 2006;23(8):857-66. doi: 10.1111/j.1464-5491.2006.01925.x, PMID 16911623.

  7. Khan MA, Hashim MJ, King JK, Govender RD, Mustafa H, Al Kaabi J. Epidemiology of type 2 diabetes-global burden of disease and forecasted trends. J Epidemiol Glob Health. 2020;10(1):107-11. doi: 10.2991/jegh.k.191028.001, PMID 32175717.

  8. Gourgari E, Wilhelm EE, Hassanzadeh H, Aroda VR, Shoulson I. A comprehensive review of the FDA-approved labels of diabetes drugs: indications, safety, and emerging cardiovascular safety data. J Diabetes Complications. 2017;31(12):1719-27. doi: 10.1016/j.jdiacomp.2017.08.005, PMID 28939018.

  9. Lotfy M, Adeghate J, Kalasz H, Singh J, Adeghate E. Chronic complications of diabetes mellitus: a mini-review. Curr Diabetes Rev. 2017;13(1):3-10. doi: 10.2174/1573399812666151016101622, PMID 26472574.

  10. Williams R, Karuranga S, Malanda B, Saeedi P, Basit A, Besançon S. Global and regional estimates and projections of diabetes-related health expenditure: results from the international diabetes federation diabetes atlas, 9th edition. Diabetes Res Clin Pract. 2020;162:108072. doi: 10.1016/j.diabres.2020.108072, PMID 32061820.

  11. Gupta R. Diabetes treatment by nanotechnology. J Biotechnol Biomater. 2017;07(3):268. doi: 10.4172/2155-952X.1000268.

  12. Khafagy ES, Morishita M, Onuki Y, Takayama K. Current challenges in non-invasive insulin delivery systems: a comparative review. Adv Drug Deliv Rev. 2007;59(15):1521-46. doi: 10.1016/j.addr.2007.08.019, PMID 17881081.

  13. Debele TA, Park Y. Application of nanoparticles: diagnosis, therapeutics, and delivery of insulin/anti-diabetic drugs to enhance the therapeutic efficacy of diabetes mellitus. Life (Basel). 2022;12(12):2078. doi: 10.3390/life12122078, PMID 36556443.

  14. Rani R, Dahiya S, Dhingra D, Dilbaghi N, Kaushik A, Kim KH. Antidiabetic activity enhancement in streptozotocin+nicotinamide-induced diabetic rats through combinational polymeric nanoformulation. Int J Nanomedicine. 2019;14:4383-95. doi: 10.2147/IJN.S205319, PMID 31354267.

  15. Yuan W, Shen T, Wang J, Zou H. Formation–dissociation of glucose, pH and redox triply responsive micelles and controlled release of insulin. Polym Chem. 2014;5(13):3968-71. doi: 10.1039/c4py00463a.

  16. Li Y, Zhang W, Zhao R, Zhang X. Advances in oral peptide drug nanoparticles for diabetes mellitus treatment. Bioact Mater. 2022;15:392-408. doi: 10.1016/j.bioactmat.2022.02.025, PMID 35386357.

  17. Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther. 2024;9(1):1. doi: 10.1038/s41392-023-01668-1, PMID 38161204.

  18. Radhakrishnan A, Kuppusamy G, Subramaniam T. Personalized nanotools for the treatment of metabolic disorders. Recent Innov Chem Eng. 2022;15(1):3-13. doi: 10.2174/2405520414666210308154038.

  19. Ahad A, Raish M, Bin Jardan YA, Al-Mohizea AM, Al-Jenoobi FI. Delivery of insulin via skin route for the management of diabetes mellitus: approaches for breaching the obstacles. Pharmaceutics. 2021;13(1):100. doi: 10.3390/pharmaceutics13010100, PMID 33466845.

  20. Pandey M, Choudhury H, Yi CX, Mun CW, Phing GK, Rou GX. Recent updates on novel approaches in insulin drug delivery: a review of challenges and pharmaceutical implications. Curr Drug Targets. 2018;19(15):1782-800. doi: 10.2174/1389450119666180523092100, PMID 29792143.

  21. Woldu MA, Lenjisa J. Nanoparticles and the new era in diabetes management. Int J Basic Clin Pharmacol. 2014;3(2):277-84. doi: 10.5455/2319-2003.ijbcp20140405.

  22. Fathi Achachelouei M, Knopf Marques H, Ribeiro da Silva CE, Barthes J, Bat E, Tezcaner A. Use of nanoparticles in tissue engineering and regenerative medicine. Front Bioeng Biotechnol. 2019;7:113. doi: 10.3389/fbioe.2019.00113, PMID 31179276.

  23. Karuppusamy C, Venkatesan P. Role of nanoparticles in drug delivery system: a comprehensive review. J Pharm Sci Res. 2017;9(3):318.

  24. Nasir A, Kausar A, Younus A. A review on preparation, properties and applications of polymeric nanoparticle-based materials. Polym Plast Technol Eng. 2015;54(4):325-41. doi: 10.1080/03602559.2014.958780.

  25. El-Say KM, El-Sawy HS. Polymeric nanoparticles: promising platform for drug delivery. Int J Pharm. 2017;528(1-2):675-91. doi: 10.1016/j.ijpharm.2017.06.052, PMID 28629982.

  26. Nagavarma B, Yadav HK, Ayaz A, Vasudha L, Shivakumar H. Different techniques for preparation of polymeric nanoparticles-a review. Asian J Pharm Clin Res. 2012;5(3):16-23.

  27. Pillai O, Panchagnula R. Polymers in drug delivery. Curr Opin Chem Biol. 2001;5(4):447-51. doi: 10.1016/s1367-5931(00)00227-1, PMID 11470609.

  28. Banik BL, Fattahi P, Brown JL. Polymeric nanoparticles: the future of nanomedicine. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2016;8(2):271-99. doi: 10.1002/wnan.1364, PMID 26314803.

  29. Yeh TH, Hsu LW, Tseng MT, Lee PL, Sonjae K, Ho YC. Mechanism and consequence of chitosan-mediated reversible epithelial tight junction opening. Biomaterials. 2011;32(26):6164-73. doi: 10.1016/j.biomaterials.2011.03.056, PMID 21641031.

  30. Sonaje K, Chuang EY, Lin KJ, Yen TC, Su FY, Tseng MT. Opening of epithelial tight junctions and enhancement of paracellular permeation by chitosan: microscopic, ultrastructural, and computed-tomographic observations. Mol Pharm. 2012;9(5):1271-9. doi: 10.1021/mp200572t, PMID 22462641.

  31. Wee S, Gombotz WR. Protein release from alginate matrices. Adv Drug Deliv Rev. 1998;31(3):267-85. doi: 10.1016/s0169-409x(97)00124-5, PMID 10837629.

  32. Vasilakes AL, Dziubla TD, Wattamwar PP. Polymeric nanoparticles. Eng Polym Syst Improv Drug Deliv. 2013:117-61.

  33. Tang Z, He C, Tian H, Ding J, Hsiao BS, Chu B. Polymeric nanostructured materials for biomedical applications. Prog Polym Sci. 2016;60:86-128. doi: 10.1016/j.progpolymsci.2016.05.005.

  34. Muhamad II, Selvakumaran S, Lazim NAM. Designing polymeric nanoparticles for targeted drug delivery system. Nanomed. 2014;287:287.

  35. Rani S, Sharma AK, Khan I, Gothwal A, Chaudhary S, Gupta U. Polymeric nanoparticles in targeting and delivery of drugs. Nanotechnol-Based Approaches Target Deliv Drugs Genes. 2017:223-55.

  36. Mendoza Munoz N, Alcala Alcala S, Quintanar Guerrero D. Preparation of polymer nanoparticles by the emulsification-solvent evaporation method: from Vanderhoff’s pioneer approach to recent adaptations. Polym Nanoparticles Nanomedicines; 2016. p. 87-121.

  37. Rauta PR, Das NM, Nayak D, Ashe S, Nayak B. Enhanced efficacy of clindamycin hydrochloride encapsulated in PLA/PLGA based nanoparticle system for oral delivery. IET Nanobiotechnology. 2016;10(4):254-61. doi: 10.1049/iet-nbt.2015.0021, PMID 27463797.

  38. Liu Y, Yang G, Zou D, Hui Y, Nigam K, Middelberg AP. Formulation of nanoparticles using mixing-induced nanoprecipitation for drug delivery. Ind Eng Chem Res. 2020;59(9):4134-49. doi: 10.1021/acs.iecr.9b04747.

  39. Wang Y, Li P, Truong Dinh Tran T, Zhang J, Kong L. Manufacturing techniques and surface engineering of polymer based nanoparticles for targeted drug delivery to cancer. Nanomaterials (Basel). 2016;6(2):26. doi: 10.3390/nano6020026, PMID 28344283.

  40. Wu L, Liu M, Shan W, Zhu X, Li L, Zhang Z. Bioinspired butyrate-functionalized nanovehicles for targeted oral delivery of biomacromolecular drugs. J Control Release. 2017;262:273-83. doi: 10.1016/j.jconrel.2017.07.045, PMID 28774842.

  41. Mohanty D, Gilani SJ, Zafar A, Imam SS, Kumar LA, Ahmed MM. Formulation and optimization of alogliptin-loaded polymeric nanoparticles: in vitro to in vivo assessment. Molecules. 2022;27(14):4470. doi: 10.3390/molecules27144470, PMID 35889343.

  42. Podichety N, Jyothi P, Pradeep K, Maddali RK. Formulation and evaluation of empagliflozin drug-loaded polymeric nanoparticles for the treatment of type 2 diabetes mellitus (T2DM). Curr Trends Biotechnol Pharm. 2022;16(3):308-15.

  43. Rani R, Dahiya S, Dhingra D, Dilbaghi N, Kim KH, Kumar S. Evaluation of anti-diabetic activity of glycyrrhizin-loaded nanoparticles in nicotinamide-streptozotocin-induced diabetic rats. Eur J Pharm Sci. 2017;106:220-30. doi: 10.1016/j.ejps.2017.05.068, PMID 28595874.

  44. Jahangir MA, Khan R, Sarim Imam S. Formulation of sitagliptin-loaded oral polymeric nano scaffold: process parameters evaluation and enhanced antidiabetic performance. Artif Cells Nanomed Biotechnol. 2018;46Suppl 1:66-78. doi: 10.1080/21691401.2017.1411933, PMID 29226729.

  45. Kaleemuddin M, Srinivas P. Lyophilized oral sustained release polymeric nanoparticles of nateglinide. AAPS PharmSciTech. 2013;14(1):78-85. doi: 10.1208/s12249-012-9887-z, PMID 23229379.

  46. Abdel Moneim A, El-Shahawy A, Yousef AI, Abd El-Twab SM, Elden ZE, Taha M. Novel polydatin-loaded chitosan nanoparticles for safe and efficient type 2 diabetes therapy: in silico, in vitro and in vivo approaches. Int J Biol Macromol. 2020;154:1496-504. doi: 10.1016/j.ijbiomac.2019.11.031, PMID 31758992.

  47. Al Rashid H. Preparation and characterization of PLGA loaded nanoparticles obtained from D. Melanoxylon Roxb. leaves for their antiproliferative and antidiabetic activity. Int J Green Pharm. 2017;11(03):438-47.

  48. Sharma DK, Pattnaik G, Behera A. Development and in vitro, in vivo evaluation of pioglitazone-loaded polymeric nanoparticles using central composite design surface response methodology. Open Nano. 2023;11:100141. doi: 10.1016/j.onano.2023.100141.

  49. Zhang P, Zhang Y, Liu CG. Polymeric nanoparticles based on carboxymethyl chitosan in combination with painless microneedle therapy systems for enhancing transdermal insulin delivery. RSC Adv. 2020;10(41):24319-29. doi: 10.1039/d0ra04460a, PMID 35516174.

  50. Jamwal S, Ram B, Ranote S, Dharela R, Chauhan GS. New glucose oxidase-immobilized stimuli-responsive dextran nanoparticles for insulin delivery. Int J Biol Macromol. 2019;123:968-78. doi: 10.1016/j.ijbiomac.2018.11.147, PMID 30448487.

  51. Chang R, Li M, Ge S, Yang J, Sun Q, Xiong L. Glucose-responsive biopolymer nanoparticles prepared by co-assembly of concanavalin a and amylopectin for insulin delivery. Ind Crops Prod. 2018;112:98-104. doi: 10.1016/j.indcrop.2017.11.017.

  52. Samimi S, Maghsoudnia N, Eftekhari RB, Dorkoosh F. Lipid-based nanoparticles for drug delivery systems. Char Biol Nanomater Drug Deliv. 2019:47-76.

  53. Rawat M, Singh D, Saraf S, Saraf S. Nanocarriers: promising vehicle for bioactive drugs. Biol Pharm Bull. 2006;29(9):1790-8. doi: 10.1248/bpb.29.1790, PMID 16946487.

  54. Salunkhe SA, Chitkara D, Mahato RI, Mittal A. Lipid-based nanocarriers for effective drug delivery and treatment of diabetes associated liver fibrosis. Adv Drug Deliv Rev. 2021;173:394-415. doi: 10.1016/j.addr.2021.04.003, PMID 33831474.

  55. Samed N, Sharma V, Sundaramurthy A. Hydrogen bonded niosomes for encapsulation and release of hydrophilic and hydrophobic anti-diabetic drugs: an efficient system for oral anti-diabetic formulation. Appl Surf Sci. 2018;449:567-73. doi: 10.1016/j.apsusc.2017.11.055.

  56. Nasr M, Almawash S, Al Saqr A, Bazeed AY, Saber S, Elagamy HI. Bioavailability and antidiabetic activity of gliclazide-loaded cubosomal nanoparticles. Pharmaceuticals (Basel). 2021;14(8):786. doi: 10.3390/ph14080786, PMID 34451883.

  57. Garcia Pinel B, Porras Alcala C, Ortega Rodriguez A, Sarabia F, Prados J, Melguizo C. Lipid-based nanoparticles: application and recent advances in cancer treatment. Nanomaterials. 2019;9(4):638.

  58. Souto EB, Souto SB, Campos JR, Severino P, Pashirova TN, Zakharova LY. Nanoparticle delivery systems in the treatment of diabetes complications. Molecules. 2019;24(23):4209. doi: 10.3390/molecules24234209, PMID 31756981.

  59. Patidar A, Thakur DS, Kumar P, Verma J. A review on novel lipid-based nanocarriers. Int J Pharm Pharm Sci. 2010;2(4):30-5.

  60. Uner M, Yener GJ. Importance of solid lipid nanoparticles (SLN) in various administration routes and future perspectives. Int J Nanomedicine. 2007;2(3):289-300. PMID 18019829.

  61. Uner M. Preparation, characterization and physico-chemical properties of solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC): their benefits as colloidal drug carrier systems. Pharmazie. 2006;61(5):375-86. PMID 16724531.

  62. Salvi VR, Pawar P. Nanostructured lipid carriers (NLC) system: a novel drug targeting carrier. J Drug Deliv Sci Technol. 2019;51:255-67. doi: 10.1016/j.jddst.2019.02.017.

  63. Shidhaye SS, Vaidya R, Sutar S, Patwardhan A, Kadam VJ. Solid lipid nanoparticles and nanostructured lipid carriers-innovative generations of solid lipid carriers. Curr Drug Deliv. 2008;5(4):324-31. doi: 10.2174/156720108785915087, PMID 18855604.

  64. Gaballa SA, El Garhy OH, Abdelkader H. Cubosomes: composition, preparation, and drug delivery applications. J Adv Biomed Pharm Sci. 2019;3(1):1-9. doi: 10.21608/jabps.2019.16887.1057.

  65. Marianecci C, Di Marzio L, Rinaldi F, Celia C, Paolino D, Alhaique F. Niosomes from 80s to present: the state of the art. Adv Colloid Interface Sci. 2014;205:187-206. doi: 10.1016/j.cis.2013.11.018, PMID 24369107.

  66. Khoee S, Yaghoobian M. Niosomes: a novel approach in modern drug delivery systems. Nanostruct Drug Deliv. 2017:207-37.

  67. He H, Lu Y, Qi J, Zhao W, Dong X, Wu W. Biomimetic thiamine and niacin-decorated liposomes for enhanced oral delivery of insulin. Acta Pharm Sin B. 2018;8(1):97-105. doi: 10.1016/j.apsb.2017.11.007, PMID 29872626.

  68. Cho EY, Ryu JY, Lee HA, Hong SH, Park HS, Hong KS. Lecithin nano-liposomal particle as a CRISPR/Cas9 complex delivery system for treating type 2 diabetes. J Nanobiotechnology. 2019;17(1):19. doi: 10.1186/s12951-019-0452-8, PMID 30696428.

  69. Nazief AM, Hassaan PS, Khalifa HM, Sokar MS, El-Kamel AH. Lipid-based gliclazide nanoparticles for treatment of diabetes: formulation, pharmacokinetics, pharmacodynamics and subacute toxicity study. Int J Nanomedicine. 2020;15:1129-48. doi: 10.2147/IJN.S235290, PMID 32110012.

  70. Kenechukwu FC, Nnamani DO, Duhu JC, Nmesirionye BU, Momoh MA, Akpa PA. Potential enhancement of metformin hydrochloride in solidified reverse micellar solution-based pegylated lipid nanoparticles targeting therapeutic efficacy in diabetes treatment. Heliyon. 2022;8(3):e09099. doi: 10.1016/j.heliyon.2022.e09099, PMID 35309393.

  71. Aldawsari HM, Hanafy A, Labib GS, Badr JM. Antihyperglycemic activities of extracts of the mistletoes plicosepalus acaciae and P. curviflorus in comparison to their solid lipid nanoparticle suspension formulations. Z Naturforsch C J Biosci. 2014;69(9-10):391-8. doi: 10.5560/znc.2014-0047, PMID 25711040.

  72. Salvi N, Choudhary GP. Development and characterization of solid lipid nanoparticles containing Murraya koenigii leaves extract for management of diabetes mellitus. Asian J Pharm Pharmacol. 2019;5(5):1048-55. doi: 10.31024/ajpp.2019.5.5.26.

  73. Bharti Sharma JB, Bhatt S, Tiwari A, Tiwari V, Kumar M, Verma R. Statistical optimization of tetrahydro curcumin loaded solid lipid nanoparticles using box behnken design in the management of streptozotocin-induced diabetes mellitus. Saudi Pharm J. 2023;31(9):101727. doi: 10.1016/j.jsps.2023.101727, PMID 37638219.

  74. Murakami T, Tsuchida K. Recent advances in inorganic nanoparticle-based drug delivery systems. Mini Rev Med Chem. 2008;8(2):175-83. doi: 10.2174/138955708783498078, PMID 18289101.

  75. Huang H, Feng W, Chen Y, Shi J. Inorganic nanoparticles in clinical trials and translations. Nano Today. 2020;35:100972. doi: 10.1016/j.nantod.2020.100972.

  76. Paul W, Sharma CP. Inorganic nanoparticles for targeted drug delivery. Biointegration Med Implant Mater. 2020:333-73.

  77. Kambale EK, Quetin Leclercq J, Memvanga PB, Beloqui A. An overview of herbal-based antidiabetic drug delivery systems: focus on lipid and inorganic based nanoformulations. Pharmaceutics. 2022;14(10):2135. doi: 10.3390/pharmaceutics14102135, PMID 36297570.

  78. Nethi SK, Das S, Patra CR, Mukherjee S. Recent advances in inorganic nanomaterials for wound-healing applications. Biomater Sci. 2019;7(7):2652-74. doi: 10.1039/c9bm00423h, PMID 31094374.

  79. Saeed BA, Lim V, Yusof NA, Khor KZ, Rahman HS, Abdul Samad N. Antiangiogenic properties of nanoparticles: a systematic review. Int J Nanomedicine. 2019;14:5135-46. doi: 10.2147/IJN.S199974, PMID 31371952.

  80. Geoprincy G, Srri BV, Poonguzhali U, Gandhi NN, Renganathan S. A review on green synthesis of silver nanoparticles. Asian J Pharm Clin Res. 2013;6(1):8-12.

  81. Vijayakumar S, Vinayagam R, Anand MA, Venkatachalam K, Saravanakumar K, Wang MH. Green synthesis of gold nanoparticle using Eclipta alba and its antidiabetic activities through regulation of Bcl-2 expression in pancreatic cell line. J Drug Deliv Sci Technol. 2020;58:101786. doi: 10.1016/j.jddst.2020.101786.

  82. Azeem MN, Ahmed OM, Shaban M, Elsayed KN. In vitro antioxidant, anticancer, anti-inflammatory, anti-diabetic and anti-Alzheimer potentials of innovative macroalgae bio-capped silver nanoparticles. Environ Sci Pollut Res Int. 2022;29(39):59930-47. doi: 10.1007/s11356-022-20039-x, PMID 35397021.

  83. Kerry RG, Singh KR, Mahari S, Jena AB, Panigrahi B, Pradhan KC. Bioactive potential of morin loaded mesoporous silica nanoparticles: a nobel and efficient antioxidant, antidiabetic and biocompatible abilities in in-silico, in vitro, and in vivo models. Open Nano. 2023;10:100126. doi: 10.1016/j.onano.2023.100126.

  84. Liu Y, Zeng S, Liu Y, Wu W, Shen Y, Zhang L. Synthesis and antidiabetic activity of selenium nanoparticles in the presence of polysaccharides from Catathelasma ventricosum. Int J Biol Macromol. 2018;114:632-9. doi: 10.1016/j.ijbiomac.2018.03.161, PMID 29601883.

  85. Shwetha UR, Latha MS, Rajith Kumar CR, Kiran MS, Betageri VS. Facile synthesis of zinc oxide nanoparticles using novel Areca catechu leaves extract and their in vitro antidiabetic and anticancer studies. J Inorg Organomet Polym. 2020;30(12):4876-83. doi: 10.1007/s10904-020-01575-w.

  86. Vijayakumar S, Divya M, Vaseeharan B, Chen J, Biruntha M, Silva LP. Biological compound capping of silver nanoparticle with the seed extracts of blackcumin (Nigella sativa): a potential antibacterial, antidiabetic, anti-inflammatory, and antioxidant. J Inorg Organomet Polym. 2021;31(2):624-35. doi: 10.1007/s10904-020-01713-4.

  87. Rani P, Kumar N, Perinmbam K, Devanesan S, AlSalhi MS, Asemi N. Synthesis of silver nanoparticles by leaf extract of cucumis melo L. and their in vitro antidiabetic and anticoccidial activities. Molecules. 2023;28(13):4995. doi: 10.3390/molecules28134995, PMID 37446657.

  88. Zhao Y, Trewyn BG, Slowing II, Lin VS. Mesoporous silica nanoparticle-based double drug delivery system for glucose-responsive controlled release of insulin and cyclic AMP. J Am Chem Soc. 2009;131(24):8398-400. doi: 10.1021/ja901831u, PMID 19476380.