Diabetes murine models for advancing oral insulin delivery: a narrative review

Modelos murinos de diabetes para el avance de la administración oral de insulina: una revisión

Revision Article



Alma Daniela Higuera Flores1 , Airam Atanahari Tapia Torres2 , Carmen Candia Plata1 , José Manuel Galván Moroyoqui1 , Gerardo Álvarez Hernández1 , Luis Fernando López Soto1 , Jesús Adriana Soto Guzman1*

1 Departamento de Medicina y Ciencias de la Salud, Universidad de Sonora. Colosio y Reforma S/N, Col. Centro, C.P. 83000, Hermosillo, Sonora, México.

2 Programa de Posgrado en Ciencias de la Salud, Departamento de Ciencias Químico Biológicas, Univer-sidad de Sonora. Luis Encinas y Rosales S/N, Col. Centro, C.P. 83000, Hermosillo, Sonora, México.

ABSTRACT

Despite the discovery of insulin over a century ago, effective oral administration remains a major research focus due to its potential for a non-invasive delivery. This review explores the pivotal role of murine diabetes models in advancing effective oral insulin formulations. A literature search was conducted in PubMed, ScienceDirect, and Google Scholar to identify experimental studies published in the last five years. The findings reveal significant progress in improving oral insulin bioavailability and glucose control using innovative techniques such as nanoparticles, microparticles, hydrogels, and smart delivery systems. These techniques were tested in experimental animal models, predominantly Wistar and Sprague Dawley rats with streptozotocin-induced diabetes. However, translating these advances into clinical practice in humans remains a challenge. Optimizing experimental mo- dels and developing sophisticated delivery technologies are crucial for achieving personalized and effective oral insulin therapies.

Keywords: Preclinical trials, bioavailability, drug delivery systems, streptozotocin, rat diabetes.


RESUMEN

A pesar del descubrimiento de la insulina hace más de un siglo, su administración oral eficaz sigue siendo de alto in- terés en el campo de la investigación debido a su potencial de administración no invasiva. Esta revisión explora el papel fundamental de los modelos murinos de diabetes en el desarrollo de formulaciones de insulina oral efectivas. Se rea- lizó una búsqueda bibliográfica en PubMed, ScienceDirect y Google Académico para identificar estudios experimentales publicados en los últimos cinco años. Los hallazgos revelan un progreso significativo en la mejora de la biodisponibili- dad de la insulina oral y el control de la glucosa empleando técnicas innovadoras como nanopartículas, micropartículas, hidrogeles y sistemas de administración inteligentes. Estas técnicas se probaron en modelos animales de experimenta- ción, predominantemente en ratas Wistar y Sprague Dawley con diabetes inducida por estreptozotocina. Sin embargo, trasladar estos avances a la práctica clínica en humanos sigue

*Author for correspondence: Jesús Adriana Soto Guzman e-mail: adriana.soto@unison.mx

Received: April 23, 2025

Accepted: July 11, 2025

Published: August 23, 2025

siendo un desafío. Optimizar los modelos experimentales y desarrollar tecnologías de administración sofisticadas es crucial para lograr terapias de insulina oral personalizadas y eficaces.

Palabras clave: Estudios preclínicos, biodisponibilidad, sistemas de liberación de fármacos, estreptozotocina, rata diabética.


INTRODUCTION

Diabetes is a chronic disease characterized by persistent hyperglycemia resulting from impaired insulin action or se- cretion (Antar et al., 2023). Type 2 diabetes (T2D) is the most prevalent form, accounting for over 90 % of cases (Ong et al., 2023). According to the International Diabetes Federation (IDF), 589 million adults are currently living with diabetes, and this figure is projected to rise to 853 million by 2050. In 2024, the disease caused approximately 3.4 million deaths (International Diabetes Federation, 2025).

Chronic hyperglycemia induces vascular damage, lea- ding to complications that affect the kidneys, eyes, nerves, and heart, among other organs, ultimately deteriorating the quality of life and life expectancy of affected individuals (Lu et al., 2024). Glycemic control is essential for the prevention and management of diabetes-related complications. Various hy- poglycemic therapies with different routes of administration (oral, subcutaneous, intranasal, etc.) have been developed to slow disease progression and improve quality of life. The current pharmacological management of diabetes includes the administration of insulin, metformin, sodium-glucose co-transporter-2 (SGLT2) inhibitor, glucagon-like peptide-1 (GLP-1) analogs, dipeptidyl peptidase-4 (DDP4) inhibitor, thiazolidinediones, and/or sulfonylureas (American Diabetes Association, 2024). Despite the wide range of treatments, the prevalence and associated morbidity and mortality of diabetes continue to rise (Zhou, B. et al., 2024), highlighting the urgent need for scientific research to develop therapies that optimize glycemic control, such as novel drug delivery systems.


1

Despite over a century since the discovery of insulin and over half a century since its introduction as a therapy


Volume XXVII

DOI: 10.18633/biotecnia.v27.2647

Journal of biological and health sciences http://biotecnia.unison.mx

Universidad de Sonora

ISSN: 1665-1456

for diabetes in humans, the development of new formula- tions, specifically for oral delivery, remains a critical area of research. This review discusses recent advances in oral insulin formulation and the strategies employed in preclinical trials using rats and mouse models of diabetes.

A literature search was conducted for experimental studies on oral insulin in murine models published in the last five years. Relevant publications were collected using the PubMed, ScienceDirect, and Google Scholar databases, employing a combination of keywords and MeSH terms, including “diabetes mellitus”, “insulin”, “oral insulin”, “oral administration”, “murinae”, “mice”, “rats”, “diabetes mellitus ex- perimental”, and “drug therapy”. Priority was given to studies exploring the most recent and relevant strategies for oral insulin administration in diabetic rats and mice. All included studies declared compliance with ethical guidelines for ani- mal experimentation.


INSULIN

Insulin is a peptide hormone secreted by the β-cells of the pancreatic islets. It has a molecular weight of 5,800 Da and comprises 51 amino acids arranged in two polypeptide chains connected by two disulfide bonds. It is synthesized from the precursor pre-proinsulin and proinsulin, releasing C-peptide. Its receptor is a transmembrane tyrosine kinase protein composed of α and β subunits (Dağaşan and Erbaş, 2021). Insulin is an anabolic hormone that primarily regulates blood glucose levels. It acts on the liver, adipose, and muscle tissue, stimulating glucose storage, lipogenesis, and protein synthesis (Rahman et al., 2021). Insulin secretion is largely mediated by glucose levels but is also influenced by hormo- nal mechanisms (incretins, glucagon), metabolic substrates (ketones, lactate, fatty acids), and sympathetic activity (Bolli et al., 2021).

Insulin is released in cyclic pulses, with a surge in con- centration occurring between 30 and 60 min postprandial, accounting for roughly one-third of the total prandial insulin release. The remaining two-thirds are secreted in response to incretin stimulation. In contrast, during fasting, insulin secre- tion is maintained at low concentrations and rates (Polinsky et al., 1988, cited in Bolli et al., 2021).

The discovery of insulin, a milestone in medical history, resulted from decades of research on pancreatic extracts. In 1921, Banting, Best, and MacLeod obtained a purified canine pancreatic extract and administered it in 1922 to a 14-year- old boy with type 1 diabetes (T1D). Early insulins, derived from porcine and bovine pancreases, had significant limita- tions, including adverse reactions because of their low purity and short duration of action. Therefore, researchers introdu- ced purification methods like electrical precipitation and zinc crystallization to overcome these limitations (Karamanou et al., 2016). In 1950, NPH insulin (Neutral Protamine Hagedorn) was launched, becoming the first long-acting basal insulin (Bolli et al., 2021). In 1955, Sanger sequenced the primary structure of bovine insulin, enabling the synthesis of the first animal insulin in the 1960s. Finally, recombinant human

insulin was effectively synthesized in 1974 (Karamanou et al., 2016). Introducing biosynthetic insulins via recombinant DNA technology in the 1980s marked a breakthrough, allowing for the development of insulin analogs with improved action profiles (Brange et al., 1988, cited in Bolli et al., 2022).

Insulin analogs are synthetic formulations designed with specific modifications in the amino acid sequence to achieve defined pharmacokinetic profiles. These include rapid-acting, short-acting, intermediate-acting, and long- acting analogs, as well as premixed combinations of rapid- and long-acting insulin for more convenient dosing based on individual requirements (Sharma et al., 2019).

Rapid-acting analogs, such as lispro and aspart, enhance absorption and minimize postprandial glucose fluctuations, making them ideal for bolus administration before meals. Long-acting analogs, like detemir and degludec, serve as basal doses, providing a flatter action profile and reducing the risk of hypoglycemia (Kramer et al., 2021). Although these analogs have improved glycemic control and patient quality of life, ongoing research focuses on developing faster and smarter insulin that automatically releases in response to plasma glucose levels (Rodbard and Rodbard, 2020).


Oral insulin

Insulin is traditionally administered subcutaneously (SC) at sites such as the abdomen, upper arms, thighs, and buttocks (American Diabetes Association, 2024). Although attempts to develop oral administration (PO) began shortly after its discovery, no commercial product is available because all tested formulations do not successfully pass clinical trials. This is mainly because gastrointestinal (GI) barriers hinder its bioavailability (Zhang et al., 2024).

Oral insulin research holds great promise in biomedicine, as it aims to provide a non-invasive administration method that mimics the physiological secretion of the pancreas. Un- like the SC route, where insulin is absorbed into the systemic circulation, oral insulin is directed to the liver through the portal vein, thus replicating natural release patterns (Nabi- Afjadi et al., 2024) (Figure 1). This approach could optimize hepatic insulinization, reduce peripheral hyperinsulinemia, and minimize the risk of complications such as hypoglycemia, neuropathy, retinopathy, immunogenicity, and weight gain (Wong et al., 2016). Oral insulin would eliminate the need for injections, a frequent concern among patients due to fear of needles (Spain et al., 2016). Injectable insulin can also cause adverse reactions such as lipohypertrophy, hypersensitivity, infections, and pain (Demir et al., 2022). Other significant limitations of injectable therapy include affordability and the complexity of self-administration in older adults, both of which impact treatment adherence (Daniell et al., 2023).

The oral administration of peptides and proteins, such as insulin, faces multiple barriers related to their physicoche- mical properties and the GI environment, which limit their bioavailability and therapeutic efficacy (Verma et al., 2021). The acidic environment of the stomach compromises insulin integrity, as its peptide bonds are susceptible to enzymatic


Figure 1. Comparison of insulin administration routes. A) Physiological release of insulin in response to food intake. B) Oral insulin administration. C) Subcutaneous insulin administration.

Figura 1. Comparación de las vías de administración de insulina. A) Liberación fisiológica de insulina en respuesta a la ingesta de alimentos. B) Administración oral de insulina. C) Administración subcutánea de insulina.

degradation by pepsin. Degradation continues in the in- testine because of the action of trypsin and chymotrypsin (Peng et al., 2023). In addition, intestinal membranes present another obstacle because of the brush border enzymes and mucin secretion, a physical barrier hindering insulin absorption. Moreover, interactions with the gut microbiome influence the stability and absorption efficiency of peptides and proteins (Wong et al., 2016).

Bioavailability of oral insulin

Strategies to enhance the bioavailability of oral insulin focus on protecting its structure, overcoming physical and biological barriers, and promoting its absorption (Peng et al., 2023). Among the developed techniques, controlled-release systems stand out as they protect the hormone during its GI transit. Other approaches include using protease inhibitors to prevent intestinal degradation and applying absorption enhancers facilitating transport across epithelial cells (Su et al., 2012) (Table 1). Formulation with mucoadhesive poly- mers and micronization has shown potential for improving solubility and structural stability (Mumuni et al., 2020). Other innovations include chemical modification of the insulin molecule (Nabi-Afjadi et al., 2024) and administration via nanoparticles or encapsulation (Daniell et al., 2023).

Protease inhibitors slow down protein degradation by binding to target enzymes, increasing the amount of protein

available for absorption (Chouhan et al., 2017). Some exam- ples that inhibit trypsin and chymotrypsin include chicken and duck ovomucoid, aprotinin, and the Bowman-Birk inhi- bitor (Muheem et al., 2016; Dan et al., 2020). However, these inhibitors may interact with other proteins, cause damage to the intestinal mucosa, and alter metabolic status (Verma et al., 2021).

Conversely, bile salts, surfactants, and certain fatty acids facilitate protein passage through the intestinal epithelium by enhancing absorption. These compounds can change tight junctions, increasing permeability (Alqahtani et al., 2021). Nevertheless, prolonged use may damage the intesti- nal mucosa and promote the translocation of pathogens and toxins (Muheem et al., 2016).

The chemical modification of peptides and proteins (whether through ligand conjugation, cyclization, or amino acid modification) can enhance resistance to enzymatic degradation, increase structural stability and cellular pene- tration, and reduce immunogenicity (Dan et al., 2020; Nicze et al., 2024). This approach offers a significant advantage over traditional absorption enhancers, as it does not compromise the integrity of cell membranes (Wong et al., 2016).

Mucoadhesive polymers encapsulate insulin, facilitating adhesion to the GI mucosa and creating a concentration gradient that enhances absorption. This strategy protects the drug and promotes a longer residence time at the absorption

Table 1. Classification of peptide and protein drug delivery systems used for oral insulin research.

Tabla 1. Clasificación de los sistemas de administración de péptidos y proteínas utilizados en la investigación de insulina oral.

Peptide and protein drug delivery systems


According to release type


  • Controlled release

  • Pulsatile release


According to release mechanism

  • Targeted release

  • Antibodies

  • Receptors

  • Stimuli-sensitive release

  • Glucose-sensitive

  • pH sensitive


According to the delivery system used


  • Nanoparticles

  • Polymeric

  • Solid lipid

  • Inorganic (metal-based, silica-based)

  • Liposomes

  • Mucoadhesive


  • Microparticles

  • Polymeric

  • Lipid-based

  • Inorganic

According to origin:

- Natural

  • Hydrogels  - Synthetic                        In particles:

-Nanogels

-Microgels

  • Self-emulsification systems

  • CPP-based systems

  • Mucoadhesive systems

  • Enzyme inhibitors Other mechanisms to improve bioa- • Permeation enhancers vailability • Absorption enhancers

  • Chemical modifications

CPP: cell penetrating peptide


site (Dan et al., 2020). In contrast, cell-penetrating peptides (CPPs) are short peptides (fewer than 30 amino acids), either amphipathic or cationic, capable of interacting with the cell membrane and internalizing biomolecules (Kristensen and Nielsen, 2016). Endocytosis and direct translocation are the primary mechanisms by which CPP-biomolecule complexes enter cells (Korivi et al., 2021), with the advantage of not inducing significant damage to the cell membranes (Bashyal et al., 2016).

With the advancement of biotechnology, new trans- port systems have emerged, such as nanoparticles and microparticles, designed using natural or synthetic materials. These systems encapsulate insulin, protecting it from de- gradation and improving its absorption. Some examples of material used in this method include lecithin, chitosan, and poly(lactic-co-glycolic acid) (PLGA) (Wong et al., 2018; Cao et al., 2019).

Liposomes, vesicles formed by concentric lipid bilayers, can encapsulate hydrophilic and hydrophobic drugs, thus enhancing their bioavailability (He et al., 2019) (Table 1). Micro and nanoemulsions, thermodynamically stable iso- tropic solutions, comparably incorporate hydrophilic and

hydrophobic molecules, improving drug distribution and penetration (Homayun et al., 2019; Nicze et al., 2024). Further innovative strategies include bilosomes, gas-enhanced deli- very systems, and endogenous cellular transport systems (Xu et al., 2020).


MURINE MODELS FOR DIABETES RESEARCH

Using animals in research has been crucial for understanding the etiology and pathophysiology processes of diabetes, as well as for developing new drugs to treat it and its compli- cations (Kottaisamy et al., 2021). Despite scientific and tech- nological progress in the biomedical field, animal models continue to play an irreplaceable role in preclinical studies (Athmuri and Shiekh, 2023). Since the 17th century, diabetes research has relied on experimental animals, with murine models predominating since the 1940s (Martín-Carro et al., 2023; Pandey et al., 2023).

Laboratory rats and mice share biological similarities with humans, with close to 95 % of their genes in common. Their small size, short gestation period, rapid development, and lower cost compared to other animals are key factors that make them ideal for experimental studies (Bryda, 2013).

Experimental models classify according to the type of diabetes they represent (type 1 or type 2) and the method by which the disease develops (spontaneous, induced, or genetically manipulated) (López-Soto et al., 2024). Chemical induction models are the most used for studying the antidia- betic activity of new drugs and insulin formulations (Janapati and Junapudi, 2024). The induction protocols vary in dosage, age, sex, and weight of the specimens, as well as the route and timing of administration (Pandey et al., 2023). The most common chemical induction agents are streptozotocin (STZ) and alloxan, which progressively destroy pancreatic β-cells, resulting in insulin deficiency and sustained hyperglycemia (Athmuri and Shiekh, 2023) (Figure 2). Depending on the aims, high-fat diets sometimes accompany this process (Fur-

sizes smaller than 100 nm. It is essential to highlight that va- rious strategies can be combined to improve bioavailability in these animal models when creating a single oral insulin formulation, for example, a glucose-sensitive polymeric na- noparticle integrated with an enzyme inhibitor.


Stimuli-responsive release nanoparticles

Sharmah et al. (2024), developed nanoceria nanoparticles functionalized with phenylboronic acid coated with dextran and showed excellent insulin loading capacity and glucose- sensitive release. This formulation showed higher bioa- vailability, more effective glucose control than SC insulin, and antioxidant properties in Wistar rats with STZ-induced diabetes. Li et al. (2024), investigated pH- and H O -sensitive

man, 2021). 2 2

Some rodents spontaneously develop autoimmune diabetes, considering genetic predisposition. These models facilitate the evaluation of the activity of active compounds without the interference of adverse effects induced by agents like STZ (Sharma et al., 2016); for example, the Biobreeding (BB) rats, LEW.1AR1-iddm rats, and the NOD mouse (López- Soto et al., 2024). As shown in Table 2, the most frequently used rat strains in recent years include Sprague-Dawley and Wistar, along with various mouse strains such as ICR, C57BL/6J, KM, Akita, Kunming, and db/db.


RECENT ADVANCES IN THE STUDY OF ORAL INSULIN

Nanoparticles

A predominant approach in the past few years is the use of nanoparticles, a versatile platform for insulin delivery with



Figure 2. Chemical induction of diabetes with streptozotozin (STZ).

Figura 2. Inducción química de diabetes con estreptozotocina (STZ).

nanoparticles to encapsulate insulin and glucose oxidase, achieving significantly higher oral insulin bioavailability than SC administration in an STZ-induced diabetic ICR mouse model.


Extended-release nanoparticles

Shapira‐Furman and Domb (2023), formulated nanoparticles made from a biodegradable copolymer of polylactic acid (D-PLA) and polyethylene glycol (PEG) that achieved sustai- ned insulin release for 14 weeks in Akita mice, resulting in significant glucose regulation. Other studies have focused on nanoparticles with hypoglycemic effects and additional properties. Pang et al. (2024), developed double-layered hy- droxide nanoparticles coated with chitosan and alginate, de- monstrating a slower release than SC insulin in STZ-induced C57BL/6J mice. Analogously, nano-carriers with Fucoidan, concanavalin A, and vitamin B12 exhibited long-lasting hy- poglycemic effects and anti-inflammatory properties, as well as a lower risk of hypoglycemia compared to SC administra- tion in STZ-induced KM mice (Zhou, J. et al., 2024).

Mucosal penetration enhancement nanoparticles

Other plausible nanoparticles include chitosan-functiona- lized PLGA nanoparticles (Asal et al., 2022), pectin-modified folic acid nanoparticles (Zhang, F. et al., 2022), and konjac glu- comannan and Concanavalin nanoparticles (Xu et al., 2022), which have achieved better penetration of the intestinal mucosa and greater bioavailability.


Liposomal and Lipid-Based Systems

Wang et al. (2018), employed cationic liposomes with a bovine serum albumin protein corona to enhance muco- sal penetration and transepithelial transport of insulin in Sprague-Dawley rats with STZ-induced diabetes. Muntoni et al. (2021), used nanostructured lipid systems to deliver insulin glargine, achieving a biphasic release pattern. In their study, only capsules demonstrated efficacy in Wistar rats with STZ-induced diabetes, while tablets and liquid formulations failed to show a significant hypoglycemic effect.

Table 2. Preclinical studies of oral insulin using murine models.

Tabla 2. Ensayos preclínicos de insulina oral utilizando modelos murinos.

Author, year

Diabetic model Strategy for insulin

delivery

Mechanism of action

Formulation

Results


Alfa et al. (2024)

Alloxan-induced model Wistar rats


Polymeric microparticles

Sustained release, protection from enzymatic degradation, improved stability.


Gelatinized PEG microparticles.

Formulations with a higher

proportion of gelatin significantly reduced blood glucose levels—no signs of liver toxicity.


Li et al. (2024)


nanoparticles

pH-responsive release, glucose-responsive release, and protection from enzymatic degradation.

Amphiphilic polymer incorporated with PBE- conjugated PHEMA and PCB to encapsulate glucose oxidase

and insulin.


A sustained hypoglycemic effect, with an oral insulin bioavailability of 20.24 %.


Pang et al. (2024)


STZ-induced model C57BL/6 J mice


Polysaccharide- coated nanoparticles


Sustained release, protection from enzymatic degradation, enhanced cellular uptake.


Layered double hydroxide nanocomposites coated with chitosan and alginate.

Significant reduction in plasma glucose levels. Slower and more sustained release, compared to SC insulin.


Sharmah et al.

(2024)


STZ-induced model Wistar rats


Glucose-responsive nanoparticles


Glucose-responsive release, enhanced cellular uptake and antioxidant activity.


Nanoceria nanoparticles are functionalized with

phenylboronic acid and coated with dextran.

Excellent insulin loading capacity and controlled release, Increased bioavailability, maintenance of normoglycemia, and antioxidant activity.


Zhou et al. (2024)


STZ-induced Polysaccharide- model KM mice based nanoparticles


Sustained release, anti- inflammatory effects, enhanced cellular uptake.


The nanoparticle system comprises Fucoidan, Concanavalin A, and Vitamin B12.

Prolonged and stable reduction in glucose levels with a lower risk of hypoglycemia. Anti- inflammatory benefits activity.


Chamsai et al. (2023)


STZ-induced Polysaccharide- model ICR mice based nanoparticles


Mucoadhesion, sustained release, protection from enzymatic degradation.


Tablets and films were developed with thiolated chitosan-based nanoparticles, encapsulated with Cyclodextrin complexes.

Rapid disintegration, high insulin release, good mucoadhesive properties,

and reduction in blood glucose levels with a prolonged hypoglycemic effect.


Chu et al. (2023)


STZ-induced

model Sprague- Lipid nanoparticles Dawley rats


Sustained release, protection from enzymatic degradation, enhanced cellular uptake.


Reverse micelles of insulin-containing sodium

deoxycholate, sulfobetaine 12, and chitosan.

Significantly higher bioavailability compared to free oral insulin and

a greater hypoglycemic response than SC insulin.


Rehmani et al.

(2023)


STZ-induced model HsdOla:TO mice


CPP


Enhanced cellular uptake, sustained release, and protection from enzymatic degradation.


Glycosaminoglycan-binding enhanced transduction uses a peptide designed to bind to heparan sulfate glycans.

Enhance insulin absorption and facilitate its intracellular transit and progressive release within cells, thus achieving sustained normoglycemia.


Ren et al. (2023)


STZ-induced model C57BL/6J mice


pH-sensitive hydrogel


pH-responsive release, sustained release, improved insulin sensitivity.


Microalgae with Chlorella vulgaris, combined with sodium alginate.

More lasting and effective hypoglycemic effects compared to SC insulin, improved insulin sensitivity and favorably regulated the gut microbiota.


Shapira‐Furman and Domb (2023)


Spontaneous diabetic model Akita mice


Polymeric nanoparticles


Sustained release, protection from enzymatic degradation.


Biodegradable copolymer of polylactic acid (D-PLA) and PEG.


A significant reduction in blood glucose levels and a normal increase in body weight.


Author, year

Diabetic model Strategy for insulin

delivery

Mechanism of action

Formulation

Results


Asal et al., (2022)


STZ-induced model Swiss albino rats


Polymeric and nanoparticle composites


Protection from enzymatic degradation, sustained release, and enhanced cellular uptake.


Chitosan-based nanoparticles, gold nanoparticles coated with chitosan, and gold nanoparticles functionalized with PLGA.

Significant reduction in blood glucose and

decrease in Hb1Ac (greater in ChAuNps/PLGA), with a gradual increase in insulin retention in acidic pH.


Goo et al. (2022)


STZ-induced model Sprague- Dawley rats


Self-micro emulsifying drug delivery system (SMEDDS)


Enhanced absorption, protection from enzymatic degradation, and improved stability.

Insulin complexed with anionic counterions (sodium octadecyl sulfate, sodium oleate, and sodium deoxycholate), surfactants (Tween 20, Brij L4, Cremophor EL), cosurfactants (Labrasol and Tetraglycol), and oils (Capmul MCM and Capryol 90).


Significant improvement in insulin stability against digestive enzymes and a

greater reduction in glucose levels compared to free insulin.


Xu et al. (2022)


STZ-induced model Male BALB/c mice


Polysaccharide- based nanoparticles


Glucose-responsive release, sustained release, enhanced cellular uptake.

A combination of konjac glucomannan and concanavalin A is used as an oral delivery system.

Reduced blood glucose levels for up to 6 h, prolonged insulin release, and biocompatibility.


Zhang, F. et al. (2022)


STZ-induced model Sprague- Dawley rats


Polysaccharide- based nanoparticles


Sustained release, protection from enzymatic degradation, targeted delivery.


Pectin nanoparticles were modified with folic acid, using a double-crosslinking method with calcium and dihydrazide adipate.

High encapsulation efficiency and stability against degradation Significant reduction in glucose levels and

improved bioavailability.


Zhang, H. et al. (2022)


STZ-induced model Wistar rats


Polysaccharide- based nano hydrogel


pH-responsive release, sustained release, and protection from enzymatic degradation.


Insulin-loaded nanohydrogel based on O-carboxymethyl chitosan and sodium alginate.

High insulin loading capacity and a controlled, sustained release of insulin for over 12 h, promoting its release in the small intestine and effective regulation of blood glucose levels.


Ito et al. (2021)


STZ-induced model ICR mice


CPP


Enhanced cellular uptake, sustained release, and protection from enzymatic degradation.


A small intestine-permeable cyclic peptide (cyclic L-DNP peptide) and Zn-insulin.


Enhance insulin absorption and show a rapid onset of blood glucose reduction.


Morales-Burgos

et al. (2021)


STZ-induced model Wistar rats


Polysaccharide- based microspheres


Targeted delivery to the colon, protection from enzymatic degradation.


Arabinoxylan microspheres with a focus on colon-targeted release.


39 % reduction in glucose levels (maximum effect after 18 h) and probably probiotic effect.


Muntoni et al.

(2021)


STZ-induced model Wistar rats


Lipid nanoparticles


Sustained release and protection from enzymatic degradation.


Insulin glargine loaded in nanostructured lipid carriers.

Insulin release exhibited a biphasic pattern. However, the capsules were the only ones that showed efficacy compared to tablets and liquid formulations.


Yang et al. (2021)


microparticles


Sustained release, improvement in insulin sensitivity, protection from enzymatic degradation.


Polysaccharide-based hydrogel microparticles with chitosan and β-cyclodextrin.

Improve the clinical manifestations of diabetes and insulin resistance, increase in insulin sensitivity, reduction in fasting

glucose levels, regulation of lipid metabolism, and antioxidant capacity.


Author, year

Diabetic model Strategy for insulin

delivery

Mechanism of action

Formulation

Results


Raguraman et al.

(2020)


STZ-induced model Wistar rats


Magnetosomes microparticles


Targeted delivery, enhanced cellular uptake, and protection from enzymatic degradation.


Microscopic particles composed of magnetite crystals conjugated with PEG.

Significant reduction in fasting blood glucose levels and a notable improvement in triglyceride, total cholesterol, and liver enzyme levels.


Zhou et al. (2020)


STZ-induced model Wistar rats


Polyeric microparticles and iron-based nanoparticles


Sustained release, targeted delivery, and protection from enzymatic degradation.


mPEG-b-PLLA microspheres incorporated with an iron- based nanoparticle modified with sodium dodecyl sulfate.

Greater intestinal absorption compared to free insulin, reduced blood glucose in a more prolonged manner than SC insulin. Insulin was

efficiently distributed to the liver and kidneys.


Mudassir et al. (2019)


STZ-induced model Sprague Dawley rats


Polyelectrolyte complex nanogels (polymers)


Protection from enzymatic degradation, enhanced cellular uptake, and sustained release.


pH-sensitive polyelectrolyte nanogels, composed of methyl methacrylate and itaconic acid.

Reduced glucose levels by 51.10 % after 6 hours. The nanogels facilitated absorption through the inhibition of proteolytic enzymes and the opening of intestinal tight junctions.


Wang et al. (2019)


STZ-induced model Sprague- Dawley rats


Liposomes


Enhanced mucosal penetration, sustained release, and protection from enzymatic degradation.


Cationic liposomes with a ‘protein corona’ formed by BSA.

Higher ability to penetrate the mucus layer and undergo transepithelial transport, and achieved a slower, sustained insulin release compared to free insulin.

BSA, bovine serum albumin; CPP, cell-penetrating peptide; KM, Kunming; PBE, phenylboronic ester; PCB, poly(carboxybetaine); PEG, polyethylene glycol; PHEMA, (PBE)-conjugated poly(2-hydroxyethyl methacrylate); PLGA, poly(lactic-co-glycolic) acid; PLLA, polylactic acid; SC, subcutaneous; STZ; streptozocin.


Microparticles

Like nanoparticles but larger, the microparticles have also been explored for controlled insulin release. Yang et al. (2021), developed polysaccharide microspheres with chitosan and β-cyclodextrin. These microspheres improved clinical diabetes symptoms and insulin resistance, showing a significant reversal of pancreatic damage in Kunming mice with STZ-induced diabetes. Additionally, arabinoxylan microspheres have been studied for colon-targeted insulin release, exhibiting probiotic effects in Wistar rats with STZ-in- duced diabetes (Morales-Burgos et al., 2021). Biodegradable mPEG-b-PLLA microspheres incorporating iron-based nano- particles revealed higher intestinal absorption and greater glycemic reduction than SC insulin or free oral insulin in vivo in Wistar rats with STZ-induced diabetes (Zhou et al., 2020).

Hydrogels

Hydrogels, polymeric networks capable of retaining a large amount of water and providing a protective environment for insulin, represent a promising system for oral insulin delivery and can be administered as nano- or microparticles. For instance, nanogels of methyl methacrylate and itaconic acid have demonstrated high encapsulation efficiency in Sprague-Dawley rat models with STZ-induced diabetes (Mu- dassir et al., 2019).

Self-Microemulsifying Drug Delivery Systems (SMEDDS) SMEDDS are mixtures of oils, surfactants, and co-surfactants that form emulsions upon dilution in aqueous media, en- hancing drug solubilization. Goo et al. (2022), developed a formulation based on this system that protected against enzymatic degradation and improved glucose absorption in STZ-induced diabetic Sprague-Dawley rats. However, cha- llenges remain in matching the efficacy of SC insulin.

Cell-Penetrating Peptide (CPP)-Based Systems

Various CPP-based strategies have been examined to en- hance cellular uptake and transport of insulin. Rehmani et al. (2023), employed a peptide to bind heparan sulfate glycans on cell membranes, resulting in improved insulin absorption, enhanced intracellular transit, and progressive release within cells in STZ-induced diabetic HsdOla:TO mice. Similarly, Ito et al. (2021), reported that intestinally permeable peptides such as the cyclic L-DNP peptide C-DNPGNET-C, when com- bined with more stable insulin formulations like Zn-insulin, exhibited efficient absorption into the portal vein without toxic effects. These CPP-based systems led to a significant reduction in glucose levels in STZ-induced diabetic ICR mice.


Smart Drug Delivery Systems (SDDS)

SDDS have gained prominence in modern medicine, particu- larly for blood glucose regulation in patients with T2D. These

systems enable controlled drug release in response to spe- cific stimuli, such as pH changes or certain enzymes (Elema et al., 2020). Examples of SDDS for oral insulin formulations include those loaded with glucose oxidase, phenylboronic acid, and lectin (Bordbar-Khiabani and Gasik, 2022).

The primary release mechanism involves glucose diffu- sion, its conversion to glucuronic acid, and subsequent pH reduction, which triggers degradation, swelling, or binding disruption, leading to insulin release (Wang et al., 2020). Key advantages of SDDS include enhanced therapeutic efficacy, resulting in better glycemic control and fewer side effects. Limitations include the complexity of their design and a restricted drug-loading capacity (Boppana et al., 2024).


TRANSLATION TO CLINICAL TRIALS AND LIMITATIONS

Despite promising results in murine models, translating oral insulin to clinical practice remains challenging due to multiple factors hindering success in clinical trials. First, va- rious methodological and biological issues compromise the external validity of preclinical studies. The lack of publication of negative results at this stage represents a bias, overestima- ting the likelihood of treatment success. Moreover, the abs- ence of standardized protocols for animal experimentation, the inability of models to replicate accurately the complexity of human diabetes, and the stress associated with laboratory conditions can substantially influence results (Mak et al., 2014). These factors, along with inadequate experimental design, method flaws, cost, commercial competition, and su- boptimal statistical analysis, contribute to the poor reprodu- cibility of preclinical findings in clinical settings (McGonigle and Ruggeri, 2014; Ioannidis et al., 2018).

Rigorous processes such as randomization, blinding, in- cluding animals of all sexes and different age groups, and the transparent publication of results are essential for enhancing the predictability of preclinical studies (Mak et al., 2014). If a clinical trial fails, reverse translation of clinical findings to animal models can provide valuable insights to refine and guide future research (Denayer et al., 2014).

The difficulty in translating oral insulin formulation is evident in the low proportion that progresses beyond phase 2 clinical trials (Table 3). For example, the formulation I338, a basal insulin analog that demonstrated similar glucose con- trol to SC insulin glargine in its early stages, was eventually discontinued due to the requirement for high doses (Halberg et al., 2019). Similarly, Novo Nordisk® halted the development of NN1952, a modified prandial insulin, due to unacceptable interactions with food intake (Zijlstra et al., 2014). The Eligen® formulation, even with a faster onset of action than regular SC insulin, exhibited high variability in absorption among patients, negatively affecting its clinical efficacy in a phase 2 trial and preventing further development (Kapitza et al., 2010; Dan et al., 2020). These cases highlight the complexity of translating preclinical results into real clinical settings and underscore the need to overcome multiple obstacles before achieving a viable formulation.

However, oral insulin research remains active, focusing on formulations with potential approval. ORMD-0801, an enteric-coated capsule, demonstrated a significant reduction in HbA1c levels in a phase 2 study (Eldor et al., 2023), which justifies initiating a phase 3 trial scheduled to begin in 2025 (Oramed, 2025). Tregopil (IN-105), although yielding mixed results in glucose control compared to SC insulin aspart, has made progress by reaching phase 3 and is still under investigation (Lebovitz et al., 2022). Likewise, the formulation


Table 3. Oral insulin clinical trials.

Tabla 3. Ensayos clínicos de insulina oral.

Trial registration number

Name

Release strategy

Formulation

Status

References


NCT06731075


ORMD-0801


Absorption enhancer / Enzyme inhibitor

Human insulin, soytrypsin inhibitor, disodium ethylenediaminetetraacetic acid, Aerosil 200, Tween 80, and enteric coating.

Phase 2

(phase 3 scheduled for 2025)


Eldor et al. (2023)

CTRI/2018/08/ 015519

Capsulin

Absorption enhancer

Recombinant human insulin, antioxidant bile salt, and enteric coating.

Phase 2

New et al. (2023)

NCT04975022

N11005

Solid self-emulsifying microemulsion

Recombinant human insulin and solid self-emulsification system Oralpas Pro®

Phase 1

Pan et al. (2023)

NCT02470039

I-338

Absorption enhancer / Chemical modification

Long-acting basal insulin analogue and sodium caprate.

Phase 2

Halberg et al.

(2019)


NCT03430856


Tregopil (IN-105)

Absorption enhancer / Chemical modification

Chemically modified human insulin, sodium caprate, and polyethylene glycol (PEG).

Phase 2 /

Phase 3

Khedkar et al.

(2019)

NCT01028404

NN1952

Chemical modification

Chemically modified human insulin and enteric coating.

Phase 1

Novo Nordisk A/S (2017)

NCT00982254

Eligen

Permeation enhancer

Insulin and N-(4-chlorosalicyloil)-4- aminobutyrate monosodium (4-CNAB).

Phase 2

Kapitza et al.

(2010)



9

Volume XXVII

8

N11005, a solid self-emulsifying system, exhibited a faster onset of action in a phase 1 study, consistent with preclinical results in rats (Beijing Hospital, 2021; Pan et al., 2023).


PERSPECTIVES

Advanced technologies and smart systems

Advanced approaches in micro- and nanotechnology, such as metallic nanoparticles, polymers, liposomes, emulsions, nanocapsules, and DNA nanoparticles, offer advantages in drug targeting and toxicity reduction (Sun et al., 2020; Sahu et al., 2021). Besides conventional strategies, stimulus- sensitive systems made from smart biomaterials represent a significant step forward in personalized insulin therapy and optimal glycemic control (Sultana et al., 2022).

Correspondingly, micro-electromechanical systems, including implantable microchips and micropumps, are emerging as key tools for drug-controlled release (Chircov and Grumezescu, 2022). Additionally, shape-memory ma- terials and 4D printing technology, which enable dynamic and responsive drug delivery systems, open new avenues for designing even more sophisticated devices that can adapt to the individual characteristics of each patient (Amukarimi et al., 2021).

Advanced preclinical models

Optimizing existing animal model management through improved experimental design and execution, as well as in- vesting in more sophisticated models that closely resemble human diabetes, is crucial. Innovations such as humanized mouse models (Fujiwara, 2018) and “organs-on-chips” technology (Wysoczański et al., 2024), which offer greater physiological relevance and simulate human physiology, could facilitate the analysis of insulin interactions in different organs (Ma et al., 2021).

Incorporating characteristics from clinical trials into preclinical studies can further improve the predictability of animal models. These characteristics include the evaluation of predictive biomarkers and the reverse translation of clini- cal results to experimental models (Denayer et al., 2014).

Although these technologies have made significant progress, many are still in the early stages of development. Therefore, increased investment in research is essential to translate these technologies into clinical practice (Xiao et al., 2020).


CONCLUSIONS

The development of oral insulin remains an up-and- coming area in medicine, potentially transforming diabetes treatment. Murine models have proven essential in diabetes research and the development of new therapies, leading to significant advancements in the design of drug delivery sys- tems that aim to improve insulin bioavailability and reduce the side effects of SC administration. However, translating these innovations to clinical practice remains challenging, underscoring the need to optimize experimental models and

continue investing in new technologies. In this context, the prospects for more effective, personalized, and less invasive insulin therapy continue to strengthen.


CONFLICTS OF INTEREST

The authors declare no conflicts of interest.


REFERENCES

Ahadian, S., Finbloom, J.A., Mofidfar, M., Diltemiz, S.E., Nasrollahi, F., Davoodi, E., Hosseini, V., Mylonaki, I., Sangabathuni, S., Montazerian, H., Fetah, K., Nasiri, R., Dokmeci, M.R., Stevens, M.M., Desai, T.A. and Khademhosseini, A. 2020. Micro and nanoscale technologies in oral drug delivery. Advanced Drug Delivery Reviews, 157: 37-62. https://doi.org/10.1016/j. addr.2020.07.012.

Alfa, J., Ben, A., Buxaderas, E., Akpa, P., Hanifah, A., Oseni,

O.M.-L., Kenechukwu, F.C., Mumuni, M.A. and Diaz, D.D. 2024. Development and evaluation of PEG-gelatin-based microparticles to enhance the oral delivery of nsulin. Current Pharmaceutical Design, 30(24): 1939-1948. https://doi.org/1 0.2174/0113816128309449240527053640.

Alqahtani, M.S., Kazi, M., Alsenaidy, M.A. and Ahmad, M.Z. 2021. Advances in oral drug delivery. Frontiers in Pharmacology,

12. https://doi.org/10.3389/fphar.2021.618411.

American Diabetes Association 2024. 9. Pharmacologic approaches to glycemic treatment: standards of care in diabetes—2024. Diabetes Care, 47(Supplement_1), S158-S178. https://doi.org/10.2337/dc24-S009.

Amukarimi, S., Ramakrishna, S. and Mozafari, M. 2021. Smart biomaterials—A proposed definition and overview of the field. Current Opinion in Biomedical Engineering, 19: 100311. https://doi.org/10.1016/j.cobme.2021.100311.

Antar, S.A., Ashour, Nada A., Sharaky, M., Khattab, M., Ashour, Naira A., Zaid, R.T., Roh, E.J., Elkamhawy, A. and Al-Karmalawy,

A.A. 2023. Diabetes mellitus: classification, mediators, and complications; A gate to identify potential targets for the development of new effective treatments. Biomedicine & Pharmacotherapy, 168: 115734. https://doi.org/10.1016/j. biopha.2023.115734.

Asal, H.A., Shoueir, K.R., El-Hagrasy, M.A. and Toson, E.A. 2022. Controlled synthesis of in-situ gold nanoparticles onto chitosan functionalized PLGA nanoparticles for oral insulin delivery. International Journal of Biological Macromolecules, 209: 2188-2196. https://doi.org/10.1016/j. ijbiomac.2022.04.200.

Athmuri, D.N. and Shiekh, P.A. 2023. Experimental diabetic animal models to study diabetes and diabetic complications. MethodsX, 11: 102474. https://doi.org/10.1016/j. mex.2023.102474.

Bashyal, S., Noh, G., Keum, T., Choi, Y.W. and Lee, S. 2016. Cell penetrating peptides as an innovative approach for drug delivery; then, present and the future. Journal of Pharmaceutical Investigation, 46(3): 205-220. https://doi. org/10.1007/s40005-016-0253-0.

Beijing Hospital. 2021. A study evaluating the bioavailability of oral insulin (N11005). ClinicalTrials.gov. Available at: https:// clinicaltrials.gov/study/NCT04975022?intr=N11005&rank=1 Bolli, G.B., Porcellati, F., Lucidi, P. and Fanelli, C.G. 2021. The physiological basis of insulin therapy in people with diabetes mellitus. Diabetes Research and Clinical Practice, 175: 108839. https://doi.org/10.1016/j.diabres.2021.108839.

Bolli, G.B., Porcellati, F., Lucidi, P., Fanelli, C.G. and Owens, D.R. 2022. One-hundred year evolution of prandial insulin preparations: from animal pancreas extracts to rapid-acting analogs. Metabolism, 126: 154935. https://doi.org/10.1016/J. METABOL.2021.154935.

Boppana, S.H., Kutikuppala, L.V.S., Sharma, S., C, M., Rangari, G., Misra, A.K., Kandi, V., Mishra, S., Singh, P.K., Rabaan, A.A., Mohapatra, R.K. and Kudrat‐E‐Zahan, Md. 2024. Current approaches in smart nano‐inspired drug delivery: a narrative review. Health Science Reports, 7(4). https://doi. org/10.1002/hsr2.2065.

Bordbar-Khiabani, A. and Gasik, M. 2022. Smart hydrogels for advanced drug delivery systems. International Journal of Molecular Sciences, 23(7): 3665. https://doi.org/10.3390/ ijms23073665.

Bryda, E.C. 2013. The Mighty Mouse: the impact of rodents on advances in biomedical research. Missouri medicine, 110(3): 207-211.

Cao, S.-J., Xu, S., Wang, H.-M., Ling, Y., Dong, J., Xia, R.-D. and Sun, X.-H. 2019. Nanoparticles: oral delivery for protein and peptide drugs. AAPS PharmSciTech, 20(5): 190. https://doi. org/10.1208/s12249-019-1325-z.

Chamsai, B., Opanasopit, P. and Samprasit, W. 2023. Fast disintegrating dosage forms of mucoadhesive-based nanoparticles for oral insulin delivery: optimization to in vivo evaluation. International Journal of Pharmaceutics, 647: 123513. https://doi.org/10.1016/j.ijpharm.2023.123513.

Chircov, C. and Grumezescu, A.M. 2022. Microelectromechanical Systems (MEMS) for biomedical applications. Micromachines, 13(2): 164. https://doi.org/10.3390/mi13020164.

Chouhan, R., Goswami, S. and Bajpai, A.K. 2017. Recent advancements in oral delivery of insulin: from challenges to solutions. Nanostructures for Oral Medicine, 435-465. https://doi.org/10.1016/B978-0-323-47720-8.00016-X.

Chu, C., Deng, Y., Liu, H., Wei, M., Xu, X., Gou, J., He, H., Yin, T., Zhang,

Y. and Tang, X. 2023. In situ rearranged multifunctional lipid nanoparticles via synergistic potentiation for oral insulin delivery. International Journal of Pharmaceutics, 636: 122811. https://doi.org/10.1016/j.ijpharm.2023.122811.

Dağaşan, S. and Erbaş, O. 2021. Insulin structure, function and diabetes models in animals. Journal of Experimental and Basic Medical Sciences, 1(3): 96-101. https://doi.org/10.5606/ jebms.2020.75622.

Dan, N., Samanta, K. and Almoazen, H. 2020. An update on pharmaceutical strategies for oral delivery of therapeutic peptides and proteins in adults and pediatrics. Children, 7(12): 307. https://doi.org/10.3390/children7120307.

Daniell, H., Singh, R., Mangu, V., Nair, S.K., Wakade, G. and Balashova, N. 2023. Affordable oral proinsulin bioencapsulated in plant cells regulates blood sugar levels similar to natural insulin. Biomaterials, 298: 122142. https:// doi.org/10.1016/j.biomaterials.2023.122142.

Demir, G., Er, E., Atik Altınok, Y., Özen, S., Darcan, Ş. and Gökşen, D. 2022. Local complications of insulin administration sites and effect on diabetes management. Journal of Clinical Nursing, 31(17-18): 2530-2538. https://doi.org/10.1111/jocn.16071.

Denayer, T., Stöhr, T. and Roy, M. Van. 2014. Animal models in translational medicine: validation and prediction. European Journal of Molecular & Clinical Medicine, 2(1): 5. https://doi. org/10.1016/j.nhtm.2014.08.001.

Eldor, R., Francis, B.H., Fleming, A., Neutel, J., Homer, K., Kidron,

M. and Rosenstock, J. 2023. Oral insulin (ORMD‐0801) in type 2 diabetes mellitus: a dose‐finding 12‐week randomized placebo‐controlled study. Diabetes, Obesity and Metabolism, 25(4): 943-952. https://doi.org/10.1111/ dom.14901.

Elema, B., Degu, T. and Fesseha, H. 2020. Smart drug delivery system: a review. Archives of Clinical Case Reports, 1(1): 11- 16.

Fujiwara, S. 2018. Humanized mice: a brief overview on their diverse applications in biomedical research. Journal of Cellular Physiology, 233(4): 2889-2901. https://doi. org/10.1002/jcp.26022.

Furman, B.L. 2021. Streptozotocin‐induced diabetic models in mice and rats. Current Protocols, 1(4). https://doi. org/10.1002/cpz1.78.

Goo, Y.T., Lee, S., Choi, J.Y., Kim, M.S., Sin, G.H., Hong, S.H.,

Kim, C.H., Song, S.H. and Choi, Y.W. 2022. Enhanced oral absorption of insulin: hydrophobic ion pairing and a self- microemulsifying drug delivery system using a D-optimal mixture design. Drug Delivery, 29(1): 2831-2845. https://doi. org/10.1080/10717544.2022.2118399.

Halberg, I.B., Lyby, K., Wassermann, K., Heise, T., Zijlstra, E. and Plum-Mörschel, L. 2019. Efficacy and safety of oral basal insulin versus subcutaneous insulin glargine in type 2 diabetes: a randomised, double-blind, phase 2 trial. The Lancet Diabetes & Endocrinology, 7(3): 179-188. https://doi. org/10.1016/S2213-8587(18)30372-3.

He, H., Lu, Y., Qi, J., Zhu, Q., Chen, Z. and Wu, W. 2019. Adapting liposomes for oral drug delivery. Acta Pharmaceutica Sinica B, 9(1): 36-48. https://doi.org/10.1016/j.apsb.2018.06.005.

Heise, T., Plum‐Mörschel, L. and Zijlstra, E. 2023. Oral insulin: a history of ambition, failure and data torturing. Diabetes, Obesity and Metabolism, 25(4): 940-942. https://doi. org/10.1111/dom.14984.

Homayun, B., Lin, X. and Choi, H.-J. 2019. Challenges and recent progress in oral drug delivery systems for biopharmaceuticals. Pharmaceutics, 11(3): 129. https://doi. org/10.3390/pharmaceutics11030129.

International Diabetes Federation. 2025. IDF Diabetes Atlas 2025 11th edition. Available at: https://diabetesatlas.org/ resources/idf-diabetes-atlas-2025/

Ioannidis, J.P.A., Kim, B.Y.S. and Trounson, A. 2018. How to design preclinical studies in nanomedicine and cell therapy to maximize the prospects of clinical translation. Nature Biomedical Engineering, 2(11): 797-809. https://doi. org/10.1038/s41551-018-0314-y.

Ito, S., Torii, Y., Chikamatsu, S., Harada, T., Yamaguchi, S., Ogata, S., Sonoda, K., Wakayama, T., Masuda, T. and Ohtsuki, S. 2021. Oral coadministration of Zn-Insulin with d-form small intestine-permeable cyclic peptide enhances its blood glucose-lowering effect in mice. Molecular Pharmaceutics, 18(4): 1593-1603. https://doi.org/10.1021/ acs.molpharmaceut.0c01010.

Janapati, Y.K. and Junapudi, S. 2024. Progress in experimental models to investigate the in vivo and in vitro antidiabetic activity of drugs. Animal Models and Experimental Medicine, 7(3): 297-309. https://doi.org/10.1002/ame2.12442.

Kapitza, C., Zijlstra, E., Heinemann, L., Castelli, M.C., Riley, G. and Heise, T. 2010. Oral insulin: a comparison with subcutaneous regular human insulin in patients with type 2 diabetes.

Diabetes Care, 33(6): 1288-1290. https://doi.org/10.2337/ dc09-1807.

Karamanou, M., Protogerou, A., Tsoucalas, G., Androutsos, G. and Poulakou-Rebelakou, E. 2016. Milestones in the history of diabetes mellitus: the main contributors. World Journal of Diabetes, 7(1): 1. https://doi.org/10.4239/wjd.v7.i1.1.

Khedkar, A., Lebovitz, H., Fleming, A., Cherrington, A., Jose, V., Athalye, S.N. and Vishweswaramurthy, A. 2019. Impact of insulin Tregopil and its permeation enhancer on pharmacokinetics of metformin in healthy volunteers: randomized, open‐label, placebo‐controlled, crossover study. Clinical and Translational Science, 12(3): 276-282. https://doi.org/10.1111/cts.12609.

Korivi, M., Huang, Y.-W. and Liu, B.R. 2021. Cell-penetrating peptides as a potential drug delivery system for effective treatment of diabetes. Current Pharmaceutical Design, 27(6): 816-825. https://doi.org/10.2174/1381612826666201

019102640.

Kottaisamy, C.P.D., Raj, D.S., Prasanth Kumar, V. and Sankaran,

U. 2021. Experimental animal models for diabetes and its related complications—a review. Laboratory Animal Research, 37(1): 23. https://doi.org/10.1186/s42826-021-

00101-4.

Kramer, C.K., Retnakaran, R. and Zinman, B. 2021. Insulin and insulin analogs as antidiabetic therapy: a perspective from clinical trials. Cell Metabolism, 33(4): 740-747. https://doi. org/10.1016/j.cmet.2021.03.014.

Kristensen, M. and Nielsen, H.M. 2016. Cell‐penetrating peptides as carriers for oral delivery of biopharmaceuticals. Basic & Clinical Pharmacology & Toxicology, 118(2): 99-106. https:// doi.org/10.1111/bcpt.12515.

Lebovitz, H.E., Fleming, A., Cherrington, A.D., Joshi, S., Athalye, S.N., Loganathan, S., Vishweswaramurthy, A., Panda, J. and Marwah, A. 2022. Efficacy and safety of Tregopil, a novel, ultra-rapid acting oral prandial insulin analog, as part of a basal-bolus regimen in type 2 diabetes: a randomized, active-controlled phase 2/3 study- Expert Opinion on Pharmacotherapy, 23(16): 1855-1863. https://doi.org/10.10 80/14656566.2022.2141569.

Li, M., Wang, N., Liu, R., Zhang, X., He, W., Zhang, W., Li, J., Peng,

C. and Li, Y. 2024. pH and H2O2 dual-sensitive nanoparticles enable enhanced and safe glucose-responsive oral insulin delivery for diabetes mellitus treatment. Theranostics, 14(14): 5596-5607. https://doi.org/10.7150/thno.98177.

López Soto, L.F., Candia Plata, C., Reyes Márquez, V., Arredondo Damián, J., Mata Pineda, A.L., Álvarez Hernández, G., Lorenzana Basaldúa, R. and Soto Guzman, A. 2024. Modelos murinos de diabetes para el estudio de compuestos bioactivos. TECNOCIENCIA Chihuahua, 18(1): e1402. https:// doi.org/10.54167/tch.v18i1.1402.

Lu, X., Xie, Q., Pan, X., Zhang, R., Zhang, X., Peng, G., Zhang, Y., Shen, S. and Tong, N. 2024. Type 2 diabetes mellitus in adults: pathogenesis, prevention and therapy. Signal Transduction and Targeted Therapy, 9(1): 262. https://doi.org/10.1038/ s41392-024-01951-9.

Ma, C., Peng, Y., Li, H. and Chen, W. 2021. Organ-on-a-Chip: a new paradigm for drug development. Trends in Pharmacological Sciences, 42(2): 119-133. https://doi.org/10.1016/j. tips.2020.11.009.

Mak, I.W., Evaniew, N. and Ghert, M. 2014. Lost in translation: animal models and clinical trials in cancer treatment.

American journal of translational research, 6(2): 114-118.

Martín-Carro, B., Donate-Correa, J., Fernández-Villabrille, S., Martín-Vírgala, J., Panizo, S., Carrillo-López, N., Martínez- Arias, L., Navarro-González, J.F., Naves-Díaz, M., Fernández- Martín, J.L., Alonso-Montes, C. and Cannata-Andía, J.B. 2023. Experimental models to study diabetes mellitus and its complications: limitations and new opportunities. International Journal of Molecular Sciences, 24(12): 10309. https://doi.org/10.3390/ijms241210309.

McGonigle, P. and Ruggeri, B. 2014. Animal models of human disease: challenges in enabling translation. Biochemical Pharmacology, 87(1): 162-171. https://doi.org/10.1016/j. bcp.2013.08.006.

Morales-Burgos, A.M., Carvajal-Millan, E., Sotelo-Cruz, N., Rascón-Chu, A., Lizardi-Mendoza, J., López-Franco, Y.L., Martínez-Porchas, M. and Canett-Romero, R. 2021. Highly cross-linked arabinoxylans microspheres as a microbiota- activated carrier for colon-specific insulin delivery. European Journal of Pharmaceutics and Biopharmaceutics, 163: 16-22. https://doi.org/10.1016/j.ejpb.2021.02.014.

Mudassir, J., Darwis, Y., Muhamad, S. and Ali Khan, A. 2019. Self- assembled insulin and nanogels polyelectrolyte complex (Ins/NGs-PEC) for oral insulin delivery: characterization, lyophilization and in-vivo evaluation. International Journal of Nanomedicine, 14: 4895-4909. https://doi.org/10.2147/ IJN.S199507.

Muheem, A., Shakeel, F., Jahangir, M.A., Anwar, M., Mallick, N., Jain, G.K., Warsi, M.H. and Ahmad, F.J. 2016. A review on the strategies for oral delivery of proteins and peptides and their clinical perspectives. Saudi Pharmaceutical Journal, 24(4): 413-428. https://doi.org/10.1016/j.jsps.2014.06.004.

Mumuni, M.A., Kenechukwu, F.C., Ofokansi, K.C., Attama, A.A. and Díaz, D.D. 2020. Insulin-loaded mucoadhesive nanoparticles based on mucin-chitosan complexes for oral delivery and diabetes treatment. Carbohydrate Polymers, 229: 115506. https://doi.org/10.1016/J.CARBPOL.2019.115506.

Muntoni, E., Anfossi, L., Milla, P., Marini, E., Ferraris, C., Capucchio, M.T., Colombino, E., Segale, L., Porta, M. and Battaglia,

L. 2021. Glargine insulin loaded lipid nanoparticles: oral delivery of liquid and solid oral dosage forms. Nutrition, Metabolism and Cardiovascular Diseases, 31(2): 691-698. https://doi.org/10.1016/j.numecd.2020.09.020.

Nabi-Afjadi, M., Ostadhadi, S., Liaghat, M., Pasupulla, A.P., Masoumi, S., Aziziyan, F., Zalpoor, H., Abkhooie, L. and Tarhriz,

V. 2024. Revolutionizing type 1 diabetes management: exploring oral insulin and adjunctive treatments. Biomedicine & Pharmacotherapy, 176: 116808. https://doi. org/10.1016/j.biopha.2024.116808.

New, R.R.C., Ramanujam, S., Chaudhari, V., Bogus, M., Travers,

G.N. and Namjoshi, G. 2023. Safety and efficacy of an oral insulin (Capsulin) in patients with early‐stage type 2 diabetes: a dose‐ranging phase 2b study. Diabetes, Obesity and Metabolism, 25(4): 953-960. https://doi.org/10.1111/ dom.14922.

Nicze, M., Borówka, M., Dec, A., Niemiec, A., Bułdak, Ł. and Okopień, B. 2024. The current and promising oral delivery methods for protein- and peptide-based drugs. International Journal of Molecular Sciences, 25(2): 815. https://doi. org/10.3390/ijms25020815.

Novo Nordisk A/S. 2017. A two part trial investigating NN1952 in healthy subjects and subjects with type 1 and 3ype 2

diabetes. ClinicalTrial.gov. Available at: https://clinicaltrials. gov/study/NCT01028404?intr=NN1952%20&rank=1

Ojo, O.A., Ibrahim, H.S., Rotimi, D.E., Ogunlakin, A.D. and Ojo,

A.B. 2023. Diabetes mellitus: from molecular mechanism to pathophysiology and pharmacology. Medicine in Novel Technology and Devices, 19: 100247. https://doi. org/10.1016/j.medntd.2023.100247.

Ong, K.L., Stafford, L.K., McLaughlin, S.A., Boyko, E.J., Vollset, S.E., Smith, A.E et al. 2023. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: a systematic analysis for the Global Burden of Disease Study 2021. The Lancet, 402(10397): 203-

234. https://doi.org/10.1016/S0140-6736(23)01301-6.

Oramed, Ltd. 2025. A phase 3 study to evaluate the efficacy and safety of ORMD-0801 in subjects with type 2 diabetes mellitus. ClinicalTrial.gov. Available at: https://clinicaltrials. gov/study/NCT04754334?intr=ORMD-0801&rank=5

Pan, Q., Wang, X., Li, W., Chen, X., Zhuang, Y., Zhou, Q., Huang,

Y., Zhou, Y., Lan, L., Wang, Z., Wang, W., Hong, J., Hao, W.-H., Yang, Y.-T. and Guo, L. 2023. Pharmacokinetics, pharmacodynamics, and safety of prandial oral insulin (N11005) in healthy subjects. Frontiers in Endocrinology, 14. https://doi.org/10.3389/fendo.2023.1172327.

Pandey, S., Chmelir, T. and Chottova Dvorakova, M. 2023. Animal models in diabetic research—History, presence, and future perspectives. Biomedicines, 11(10): 2852. https://doi. org/10.3390/biomedicines11102852.

Pang, H., Wu, Y., Chen, Y., Chen, C., Nie, X., Li, P., Huang, G., Xu, Z.P. and Han, F.Y. 2024. Development of polysaccharide-coated layered double hydroxide nanocomposites for enhanced oral insulin delivery. Drug Delivery and Translational Research, 14(9): 2345-2355. https://doi.org/10.1007/s13346-

023-01504-7.

Peng, H., Wang, J., Chen, J., Peng, Y., Wang, X., Chen, Y., Kaplan,

D.L. and Wang, Q. 2023. Challenges and opportunities in delivering oral peptides and proteins. Expert Opinion on Drug Delivery, 20(10): 1349-1369. https://doi.org/10.1080/1

7425247.2023.2237408.

Raguraman, V., Jayasri, M.A. and Suthindhiran, K. 2020. Magnetosome mediated oral insulin delivery and its possible use in diabetes management. Journal of Materials Science: Materials in Medicine, 31(8): 75. https://doi.org/10.1007/ s10856-020-06417-2.

Rahman, M.S., Hossain, K.S., Das, S., Kundu, S., Adegoke, E.O., Rahman, Md.A., Hannan, Md.A., Uddin, M.J. and Pang, M.-

G. 2021. Role of insulin in health and disease: an update. International Journal of Molecular Sciences, 22(12): 6403. https://doi.org/10.3390/ijms22126403.

Rehmani, S., McLaughlin, C.M., Eltaher, H.M., Moffett, R.C., Flatt,

P.R. and Dixon, J.E. 2023. Orally-delivered insulin-peptide nanocomplexes enhance transcytosis from cellular depots and improve diabetic blood glucose control. Journal of Controlled Release, 360: 93-109. https://doi.org/10.1016/j. jconrel.2023.06.006.

Ren, C., Zhong, D., Qi, Y., Liu, C., Liu, X., Chen, S., Yan, S. and Zhou,

M. 2023. Bioinspired pH-responsive microalgal hydrogels for oral insulin delivery with both hypoglycemic and insulin sensitizing effects. ACS Nano, 17(14): 14161-14175. https:// doi.org/10.1021/acsnano.3c04897.

Rodbard, H.W. and Rodbard, D. 2020. Biosynthetic human insulin and insulin analogs. American Journal of Therapeutics, 27(1): e42-e51. https://doi.org/10.1097/MJT.0000000000001089.

Sahu, T., Ratre, Y.K., Chauhan, S., Bhaskar, L.V.K.S., Nair, M.P. and Verma, H.K. 2021. Nanotechnology based drug delivery system: current strategies and emerging therapeutic potential for medical science. Journal of Drug Delivery Science and Technology, 63: 102487. https://doi. org/10.1016/j.jddst.2021.102487.

Shapira‐Furman, T. and Domb, A.J. 2023. Insulin extended release from PLA‐PEG stereocomplex nanoparticles. Macromolecular Bioscience, 24(5). https://doi.org/10.1002/ mabi.202300497.

Sharma, A.K., Taneja, G., Kumar, A., Sahu, M., Sharma, G., Kumar, A., Sardana, S. and Deep, A. 2019. Insulin analogs: glimpse on contemporary facts and future prospective. Life Sciences, 219: 90-99. https://doi.org/10.1016/j.lfs.2019.01.011.

Sharma, P., Garg, A., Garg, S. and Singh, V. 2016. Animal model used for experimental study of diabetes mellitus: an overview. Asian Journal of Biomaterial Research, 2(4): 99- 110.

Sharmah, B., Barman, H., Afzal, N.U., Loying, R., Kabir, M.E., Borah, A., Das, J., Kalita, J. and Manna, P. 2024. Surface-functionalized nanoceria: dual action in diabetes management via glucose- responsive insulin delivery and oxidative stress mitigation. ACS Biomaterials Science & Engineering, 10(10): 6397-6414. https://doi.org/10.1021/acsbiomaterials.4c01368.

Spain, C.V., Wright, J.J., Hahn, R.M., Wivel, A. and Martin, A.A. 2016. Self-reported barriers to adherence and persistence to treatment with injectable medications for type 2 diabetes. Clinical Therapeutics, 38(7): 1653-1664.e1. https://doi. org/10.1016/j.clinthera.2016.05.009.

Su, F.Y., Lin, K.J., Sonaje, K., Wey, S.P., Yen, T.C., Ho, Y.C., Panda, N.,

Chuang, E.Y., Maiti, B. and Sung, H.W. 2012. Protease inhibition and absorption enhancement by functional nanoparticles for effective oral insulin delivery. Biomaterials, 33(9): 2801- 2811. https://doi.org/10.1016/J.BIOMATERIALS.2011.12.038.

Sultana, A., Zare, M., Thomas, V., Kumar, T.S.S. and Ramakrishna,

S. 2022. Nano-based drug delivery systems: conventional drug delivery routes, recent developments and future prospects. Medicine in Drug Discovery, 15: 100134. https:// doi.org/10.1016/j.medidd.2022.100134.

Sun, J., Yang, Z. and Teng, L. 2020. Nanotechnology and microtechnology in drug delivery systems. Dose- Response, 18(2): 55932582090781. https://doi. org/10.1177/1559325820907810.

Verma, S., Goand, U.K., Husain, A., Katekar, R.A., Garg, R. and Gayen, J.R. 2021. Challenges of peptide and protein drug delivery by oral route: current strategies to improve the bioavailability. Drug Development Research, 82(7): 927-944. https://doi.org/10.1002/ddr.21832.

Wang, A., Yang, T., Fan, W., Yang, Y., Zhu, Q., Guo, S., Zhu, C., Yuan, Y., Zhang, T. and Gan, Y. 2019. Protein corona liposomes achieve efficient oral insulin delivery by overcoming mucus and epithelial barriers. Advanced Healthcare Materials,

8(12). https://doi.org/10.1002/adhm.201801123.

Wang, J., Wang, Z., Yu, J., Kahkoska, A.R., Buse, J.B. and Gu, Z. 2020. Glucose‐responsive insulin and delivery dystems: innovation and translation. Advanced Materials, 32(13). https://doi.org/10.1002/adma.201902004.

Wong, C.Y., Al-Salami, H. and Dass, C.R. 2018. Microparticles, microcapsules and microspheres: a review of recent developments and prospects for oral delivery of insulin. International Journal of Pharmaceutics, 537(1-2): 223-244. https://doi.org/10.1016/j.ijpharm.2017.12.036.

Wong, C.Y., Martinez, J. and Dass, C.R. 2016. Oral delivery of insulin for treatment of diabetes: status quo, challenges and opportunities. Journal of Pharmacy and Pharmacology, 68(9): 1093-1108. https://doi.org/10.1111/jphp.12607.

Wysoczański, B., Świątek, M. and Wójcik-Gładysz, A. 2024. Organ- on-a-Chip models—New possibilities in experimental science and disease modeling. Biomolecules, 14(12): 1569. https://doi.org/10.3390/biom14121569.

Xiao, Y., Tang, Z., Wang, J., Liu, C., Kong, N., Farokhzad, O.C. and Tao, W. 2020. Oral insulin delivery platforms: strategies to address the biological barriers. Angewandte Chemie International Edition, 59(45): 19787-19795. https://doi. org/10.1002/anie.202008879.

Xu, M., Huang, J., Jiang, S., He, J., Wang, Z., Qin, H. and Guan, Y.-Q. 2022. Glucose sensitive konjac glucomannan/concanavalin A nanoparticles as oral insulin delivery system. International Journal of Biological Macromolecules, 202: 296-308. https:// doi.org/10.1016/j.ijbiomac.2022.01.048.

Xu, Y., Shrestha, N., Préat, V. and Beloqui, A. 2020. Overcoming the intestinal barrier: a look into targeting approaches for improved oral drug delivery systems. Journal of Controlled Release, 322: 486-508. https://doi.org/10.1016/j. jconrel.2020.04.006.

Yang, Y., Chen, S., Liu, Y., Huang, Y., Cheong, K.-L., Teng, B. and Liu, W. 2021. Long-term treatment of polysaccharides- based hydrogel microparticles as oral insulin delivery in streptozotocin-induced type 2 diabetic mice. Biomedicine & Pharmacotherapy, 133: 110941. https://doi.org/10.1016/j. biopha.2020.110941.

Zhang, E., Zhu, H., Song, B., Shi, Y. and Cao, Z. 2024. Recent advances in oral insulin delivery technologies. Journal of Controlled Release, 366: 221-230. https://doi.org/10.1016/j. jconrel.2023.12.045.

Zhang, F., Pei, X., Peng, X., Gou, D., Fan, X., Zheng, X., Song, C., Zhou, Y. and Cui, S. 2022. Dual crosslinking of folic acid- modified pectin nanoparticles for enhanced oral insulin delivery. Biomaterials Advances, 135: 212746. https://doi. org/10.1016/j.bioadv.2022.212746.

Zhang, H., Gu, Z., Li, W., Guo, Lili, Wang, L., Guo, Lan, Ma, S., Han, B. and Chang, J. 2022. pH-sensitive O-carboxymethyl chitosan/sodium alginate nanohydrogel for enhanced oral delivery of insulin. International Journal of Biological Macromolecules, 223: 433-445.: https://doi.org/10.1016/j. ijbiomac.2022.10.274.

Zhou, B., Rayner, A.W., Gregg, E.W., Sheffer, K.E., Carrillo-Larco, R.M., Bennett, J.E. et al. 2024. Worldwide trends in diabetes prevalence and treatment from 1990 to 2022: a pooled analysis of 1108 population-representative studies with 141 million participants. The Lancet, 404(10467): 2077-2093. https://doi.org/10.1016/S0140-6736(24)02317-1

Zhou, J., Ma, H., Guan, M., Feng, J., Dong, X., Wei, Y. and Zhang, T. 2024. Anti-inflammatory Fucoidan-ConA oral insulin nanosystems for smart blood glucose regulation. International Journal of Pharmaceutics, 659: 124250. https:// doi.org/10.1016/j.ijpharm.2024.124250.

Zhou, Y., Liu, L., Cao, Y., Yu, S., He, C. and Chen, X. 2020. A nanocomposite vehicle based on metal–organic framework nanoparticle incorporated biodegradable microspheres for enhanced oral insulin delivery. ACS Applied Materials & Interfaces, 12(20): 22581-22592. https://doi.org/10.1021/ acsami.0c04303.

Zijlstra, E., Heinemann, L. and Plum-Mörschel, L. 2014. Oral insulin reloaded. Journal of Diabetes Science and Technology, 8(3): 458-465. https://doi.org/10.1177/1932296814529988.