HOME JOURNALS CONTACT

Journal of Medical Sciences

Year: 2015 | Volume: 15 | Issue: 5 | Page No.: 209-220
DOI: 10.3923/jms.2015.209.220
Oral Delivery of Insulin for Treatment of Diabetes: Classical Challenges and Current Opportunities
M. J. Ansari

Abstract: Oral dosage form is the most common form of delivery systems due to several benefits such as ease of manufacturing, ease of administration, better formulation stability and patient compliance. Recently many proteins and peptides have been investigated for their usefulness in therapy, but mainly as parenterals due to the associated inherent problems, for instance its rapid degradation, low permeability and absorption in gastrointestinal tract. These problems must be solved or minimized to an extent that would be clinically significant before the oral delivery of proteins and peptides becomes a reality. Among the proteins, oral delivery of insulin has been attempted extensively but without much success so far and yet no formulation with oral delivery of insulin could be marketed. Several approaches have been developed to enhance oral absorption of insulin, such as inhibition of acidic and enzymatic degradation, enhancement of membrane permeability or widening of tight junctions, enhancement of insulin uptake and development of novel insulin carriers. This review article mainly focuses on the classical challenge of oral delivery of insulin and different strategies to overcome the related issues. Moreover, the current drug delivery technologies adopted in an attempt to develop practicable oral insulin have been discussed.

Fulltext PDF Fulltext HTML

How to cite this article
M. J. Ansari , 2015. Oral Delivery of Insulin for Treatment of Diabetes: Classical Challenges and Current Opportunities. Journal of Medical Sciences, 15: 209-220.

Keywords: bioavailability, absorption, permeability, protease inhibitor, oral, insulin and Diabetes

INTRODUCTION

Diabetes is a group of metabolic disorders of multiple etiologies characterized by chronic hyperglycemia with disturbance of carbohydrate, fat and protein metabolism resulting from a defect in insulin secretion or action or both (American Diabetes Association, 2006). It is a global epidemic with devastating humanitarian, social and economic consequences affecting more than 230 million people worldwide and this figure is expected to rise to 366 million by the year 2030 (WHO., 2005). It is a condition primarily defined by the level of hyperglycemia giving rise to risk of microvascular damage (retinopathy, nephropathy and neuropathy) and increased risk of macrovascular complications (ischaemic heart disease, stroke and peripheral vascular disease) and diminished quality of life. It is the fourth leading cause of death in most of the developed countries. At least 50% of all people with diabetes are unaware of their condition. In some countries, this figure may be as high as 80%. By the time diabetes is diagnosed, many have already started to develop the complications of diabetes: visual impairment, kidney failure, heart disease, stroke and nerve damage (American Diabetes Association, 2009).

Type 1 diabetes that accounts for 5-10% of cases is characterized by beta cell destruction leading to absolute insulin deficiency. Autoimmune destruction of beta cell has multiple genetic predisposing factors and directly related to the surrounding environment (Wahl et al., 1998). Type 2 diabetes is characterized by predominant insulin resistance and relative insulin deficiency or vice versa. This is the most common form of diabetes mellitus forming 90-95% of diabetic cases. It is highly associated with a family history of diabetes, older age, obesity and lack of exercise (American Diabetes Association, 2009; Lisa, 2007).

Keeping in view of devastating consequences associated with diabetes, an extensive effort have been made to overcome this health disorder through insulin administration. So, far practical purposes insulin therapy is the most common and acceptable therapy. However, due to ease of administration and several advantages associated with oral dosage forms, oral delivery of insulin is also being attempted as an alternative but has experienced several technical problem and minimal success. This review article focuses on challenges associated with oral insulin delivery and different strategies to overcome. Moreover, current developments and trends have also been discussed.

INSULIN INJECTION THERAPY: ADVANTAGES AND DISADVANTAGES

For the treatment of diabetes, nothing better than insulin has been discovered as insulin therapy is known to improve insulin receptor sensitivity, reduces glucotoxicity and lipotoxicity (Scarlett et al., 1982; Zhao et al., 2009). Insulin injection therapy has advantages such as prompt and precise monitoring of blood glucose level as no absorption and other confounding factors are involved. Moreover, it reduces the risk of long-term diabetes complications and produces sustained tight glycaemic control provided that patients start it early and adhered well to the treatment (Turner et al., 1999; Li et al., 2004; Ryan et al., 2004; Weng et al., 2008). However, using insulin injections as long-term therapy has several disadvantages such as pain at the site of injection, inconvenience of multiple injections, allergic reactions, hyper-insulinemia and insulin lipodystrophy around the injection site (Gowthamarajan and Kulkarni, 2003; Funnell, 2007; Vardar and Kizilci, 2007). Lipodystrophy is known to reduce further insulin absorption as much as 25% thus compromising glycemic control (Johansson et al., 2005). Weight gain and hypoglycemia are other common adverse reactions of insulin therapy (Bott et al., 1997; Allen et al., 2001). This review article reveals that at least one third of the diabetes patients fail to take their insulin as prescribed or intentionally skip their doses which may be related to adverse effects associated with insulin injections. Fear of insulin or fear of injection is another factor which is known to be associated with reduced compliance and adherence to the treatment and hence poor glycemic control, clinical complications, psychological co-morbidities, poor health status and increased risk of mortality for diabetes patients (Pamnani, 2008).

ORAL DELIVERY OF INSULIN: POSSIBLE ADVANTAGES AND DISADVANTAGES

The oral delivery of drugs is considered as the most acceptable and convenient route of drug administration especially for chronic diseased conditions like diabetes. Orally administered insulin would eliminate side effects, compliance problems to treatment adherence and other complications associated with insulin injection therapy such as pain caused by injection, psychological barriers associated with multiple daily injections such as needle anxiety (Korytkowski, 2002) and possible infections (Lin et al., 2007a). Current subcutaneous insulin treatment does not replicate the normal dynamics of endogenous insulin release, resulting in a failure to achieve a lasting glycemic control in patients (Agarwal and Khan, 2001; Morishita et al., 2007). Oral insulin delivery would be advantageous because it would be delivered directly to its target organ, liver, through the portal circulation, a mechanism very similar to endogenous insulin. Likely disadvantages of oral delivery of insulin would be slow onset of action and imprecise glycemic control as oral delivery systems usually have large number of excipients and suffer with dose dumping, release burst, variability in absorption and bioavailability due to several confounding factors such as fasting and fed states, concomitant administration and drug-drug interactions.

CLASSICAL CHALLENGES AND CURRENT OPPORTUNITIES TO THE ORAL DELIVERY OF INSULIN

Proteins and peptides including insulin may not be successfully delivered per-oral due to rapid enzymatic degradation in the stomach, inactivation and digestion by proteolytic enzymes in the intestinal lumen and poor permeability across intestinal epithelium owing to its high molecular weight and lack of lipophilicity (Fix, 1996; Wang, 1996; Saffran et al., 1997). Pepsin, pancreatic proteolytic enzymes such as trypsin and α-chymotrypsin and cytosolic insulin is insulin-degrading enzyme causes degradation of insulin (Patki and Jagasia, 1996; Chang et al., 1997). Overall, insulin degrades very quickly by acid and enzymes and absorbs very slowly resulting in very low bioavailability that is clinically insufficient (Shah et al., 2002; Morishita and Peppas, 2006).

INSULIN PROTECTION IN GASTROINTESTINAL TRACT BY GASTRIC AND PANCREATIC ENZYME INHIBITORS

Removal of enzyme attack or protection from it may be helpful in successful oral delivery of insulin. For this reason several researcher have investigated enzyme inhibitors and fate of oral insulin. Enzyme inhibitors slow the rate of degradation of insulin, which increases the amount of insulin available for absorption. This review article revealed the use of different protease inhibitors resulting in significant hypoglycemic effects which include aprotinin (Ziv et al., 1987; Morishita et al., 1993; Laurenti et al., 1996; Kimura et al., 1996; Morimoto et al., 2000; Radwan and Aboul-Enein, 2002; Katayama et al., 2003; Cilek et al., 2005; Park et al., 2007; Jelvehgari et al., 2011; Boateng et al., 2014) bacitracin (Yamamoto et al., 1994; Kimura et al., 1996; Bernkop-Schnurch, 1998; Park et al., 2007; Su et al., 2012; Balabushevich et al., 2013; Jose et al., 2013) camostat mesilate (Yamamoto et al., 1994; Ogiso et al., 1997; Tozaki et al., 2001; Del Curto et al., 2009), leupeptin (Tasaka et al., 1989; Liu et al., 2003) and diethylene triaminepentaacetic acid (Su et al., 2012).

Concurrent administration of protease inhibitors or its incorporation in several drug delivery systems with insulin has resulted in improved absorption, pharmacological activity and bioavailability of oral insulin. However, its use in long-term therapy of diabetes is dubious as several undesired effects such as stimulation of protease secretion, disturbance of digestion of nutritive proteins and absorption of unwanted proteins along with insulin would be unavoidable.

ALTERNATIVE STRATEGIES TO PREVENT INSULIN DEGRADATION IN GUT

Micro encapsulation is a process by which core materials such as solids, liquids or even gases may be enclosed in microscopic particles of polymers or several other substances. Such processes isolate the core from its harmful external environments such as isolating vitamins from the deteriorating effects of oxygen, volatile core from evaporation and isolating a reactive core from chemical attack. The selection of appropriate coating material decides the physical and chemical properties of the final encapsulated product. Generally hydrophilic polymers, hydrophobic polymers or a combination of both are used for the microencapsulation process. The polymer should be capable of forming a film that is cohesive with the core material. It should be chemically compatible, non-reactive with the core material and provide the desired coating properties such as strength, flexibility, impermeability, optical properties and stability.

Different techniques may be employed for encapsulation to incorporate reasonably high concentrations of the drug stabile for sufficiently long period to be clinically useful. Degradation of oral insulin by proteolytic enzymes has been minimized by encapsulating it in coatings such as enteric coating (Hosny et al., 2002; Qi and Ping, 2004; Li et al., 2012) mucoadhesive coatings (Woitiski et al., 2011; Deat-Laine et al., 2013a; Li et al., 2013a).

LOW PERMEABILITY OF INSULIN AND UTILIZING PENETRATION ENHANCERS

Human skin provides a very efficient transport barrier to delivery of protein molecules like insulin, due to their large size and weakly hydrophobic nature. Intestinal permeation enhancement might be employed for oral insulin delivery so as to promote absorption through oral route. Hydrophilic molecules including insulin are adsorbed to the apical membrane and are internalized by endocytosis or via paracellular transport (Agarwal and Khan, 2001). Tight junctions between each of the cells in the epithelium prevent water and aqueous soluble compounds from moving through cells. Hence, approaches for modulating tight-junction permeability to increase paracellular transport have been studied (Salamat-Miller and Johnston, 2005). A number of absorption enhancers are available that may open these tight junctions transiently thus allowing water-soluble proteins to pass. These include substances like bile salts (Yamamoto et al., 1992; Uchiyama et al., 1999; Degim et al., 2004; Lane et al., 2005; Lane and Corrigan, 2006) surfactants (Touitou et al., 1980; Lane et al., 2005; Lane and Corrigan, 2006; Karamanidou et al., 2015), cell penetrating peptides (Liang and Yang, 2005; Morishita et al., 2007; Kamei et al., 2008, 2013; He et al., 2013; Nielsen et al., 2014; Zhu et al., 2014, 2015) Zonula occludens toxin (Fasano and Uzzau, 1997) and chelating agents like EDTA (Yamamoto et al., 1992; Uchiyama et al., 1999; Li and Deng, 2004).

Employing penetration enhancers or its incorporation in several drug delivery systems has resulted in improved absorption, pharmacological activity and bioavailability of oral insulin. However, its use in long-term therapy of diabetes is harmful as these are not specific and are relatively toxic which may damage cell membrane. Moreover, absorption of undesired materials such as toxins and pathogens to systemic circulation along with insulin is highly likely which may prove dangerous.

ALTERNATIVE STRATEGIES TO INCREASE INTESTINAL PERMEABILITY OF INSULIN

Encapsulations of insulin in mucoadhesive microspheres or nanoparticles were found to enhance intestinal permeability of insulin. Several authors have reported increased permeation of insulin when encapsulated in Trimethyl chitosan-cysteine conjugate microsphere (Yin et al., 2009), thiolated polymethacrylic acid-polyethylene glycol-chitosan based hydrogel microparticles (Sajeesh et al., 2010a), multilayered nanoparticles of alginate and dextran sulfate (Woitiski et al., 2011), lauroylsulphated chitosan microparticles (Shelma and Sharma, 2013), whey protein and alginate microsphere (Deat-Laine et al., 2013b).

Methyl-β-cyclodextrin (MCD) complexed insulin encapsulated in polymethacrylic acid (PMMA) hydrogel microparticles was evaluated for permeability of insulin using Caco-2 cell monolayers and excised intestinal tissue with an Using chamber (Sajeesh et al., 2010b). The MCD complexation was found effective in enhancing insulin transport across Caco-2 cell monolayers, when applied in combination with the PMAA hydrogel system.

DRUG DELIVERY SYSTEMS REPORTING ENHANCED ABSORPTION OF ORAL INSULIN

Several drug delivery systems have been developed and evaluated in an attempt to achieve clinically sufficient bio availability of insulin after oral delivery. These systems either protected insulin degradation or enhanced intestinal permeability or uptake of insulin resulting in enhanced absorption and hence bio availability of oral insulin. Some drug delivery systems have combined both the strategies.

Liposomes developed for oral delivery of insulin: Liposomes are phospholipids vesicles with aqueous cavity which are formed when phospholipids are combined with water (Gowthamarajan and Kulkarni, 2003). These phospholipid vesicles can encapsulate both hydrophobic and hydrophilic drug. These are non toxic, non-immunogenic, biocompatible and biodegradable. The drugs encapsulated in liposomes are sufficiently protected from external harsh environment. The lipid bilayer of liposomes releases the drugs entrapped by fusion with other bilayers such as the cell membrane thus making them useful for delivery of poorly soluble and unstable drugs (Vemuri and Rhodes, 1995). This review revealed a huge number of conventional as well as modified liposomes for enhanced absorption and bioavailability of oral insulin (Dapergolas and Gregoriadis, 1976; Rowland and Woodley, 1981; Arrieta-Molero et al., 1982; Das et al., 1988; Petkowicz et al., 1989; Choudhari et al., 1994; Takeuchi et al., 1996; Kisel et al., 2001; Katayama et al., 2003; Wu et al., 2004; Degim et al., 2004; Zhang et al., 2005, 2014a, b; Park et al., 2011; Manosroi et al., 2011; Niu et al., 2011, 2012, 2014; Agrawal et al., 2014; Cui et al., 2015). The hypoglycaemic effect of insulin encapsulated in liposomes has been found to depend on the lipid composition, physical state and number of phospholipid bilayer and surface charge (Choudhari et al., 1994).

Physical instability, short shelf-life, low drug loading, leakage of entrapped drug and high production cost make it impractical for commercialization especially for treatment of chronic diseases such as diabetes (Gowthamarajan and Kulkarni, 2003).

Nanoparticles developed for oral delivery of insulin: Nanoparticles are spherical microscopic structures having at least one dimension less than 100 nm which may be either solid (nanospheres) or hollow (nanocapsules). Recently nanoparticles have been proposed as colloidal drug carriers with advantages such as improved bioavailability due to enhanced aqueous solubility, improved drug stability, targeted drug delivery. Thus over all, improved therapeutic index and decreased unwanted effects are the major benefits provided by such colloidal drug delivery carriers (Irving, 2007). The nanoparticle releases the entrapped drug either by controlled diffusion or by erosion from the core across the polymeric membrane (Mohanraj and Chen, 2006). Nanoparticles have advantage over liposomes in term of better physical stability, better drug loading capacity and longer duration of action. These are prepared from natural or synthetic polymers. Natural polymers studied for preparation and evaluation of oral insulin nanoparticles include chitosan (Lin et al., 2007b; Sadeghi et al., 2008; Rekha and Sharma, 2009; Yin et al., 2009; Avadi et al., 2010; Su et al., 2012; Fonte et al., 2012; Chuang et al., 2013; Li et al., 2013b; Mansourpour et al., 2015), alginate (Kadir et al., 2013; Wong and Sumiran, 2014), gelatin, albumin (Rieux et al., 2006; Woitiski et al., 2011) and lectin (Ghilzai, 2003). Synthetic polymers used for nanoparticle formulation include acrylates and its derivatives (Sajeesh and Sharma, 2006; Damge et al., 2010; Perera et al., 2009; Socha et al., 2009), polylactic-co-glycolic acid derivatives (Carino et al., 2000; Shi et al., 2008; Han et al., 2009; Zhu et al., 2015). The biological effect of insulin nanoparticles depends on the amount of both insulin and polymer. The nature of polymers strongly influences the nanoparticle size and release profile (Rieux et al., 2006).

Polymric nanoparticles have several disadvantages like low stability, low drug carrying capacity, leakage of entrapped drug and toxicity of the residual solvents and surfactants used (Gowthamarajan and Kulkarni, 2003). Solid lipid nanoparticles (SLNs) have been proposed as an alternative liposomes and polymeric nanoparticles, due to various advantages, such as feasibility of incorporation of lipophilic and hydrophilic drugs, improved physical stability, low cost, ease of scale-up and manufacturing (Muller and Peters, 1998; Kumar, 2000; Mukherjee et al., 2009). Several studies of insulin-loaded solid lipid-based nanoparticles have reported to enhance either oral bioavailability or pharmacological activity of oral insulin or both (Wang et al., 2009; Zhang et al., 2006, 2009, 2012; Sarmento et al., 2007; Liu et al., 2007; Fonte et al., 2012; Ansari et al., 2015).

Microemulsions and nanoemulsions investigated for oral delivery of insulin: Microemulsions are isotropic, transparent, thermodynamically stable liquids which are composed of oil, water and amphiphilic compounds like surfactant and co-surfactant (Lawrence and Rees, 2000). As size of the dispersed particles is much smaller than the wavelength of visible light, microemulsions are transparent and their structure cannot be observed through an optical microscope. Microemulsions have attracted increasing attention as potential drug delivery systems and as bioavailability enhancers for poorly water soluble drugs, due to their unique spontaneous energy-less formulation technique and capability of solubilizing drugs. These are known to protect water soluble drug molecules, in particular proteins and peptides from metabolism and to overcome physical barriers. Microemulsions enhance the bioavailability of poorly soluble drugs by maintaining them in molecular dispersion in the gastrointestinal tract and extending the absorption window available in the gastrointestinal lumen. Improved oral delivery of insulin from microemulsion systems have been reported by several authors (Cho and Flynn, 1989; Patel et al., 1991; Kraeling and Ritschel, 1992; Watnasirichaikul et al., 2000, 2002; Cilek et al., 2005; Graf et al., 2009; Sharma et al., 2010; Karamanidou et al., 2015; Rachmawati et al., 2015).

Self-microemulsifying drug delivery systems are are "Latent" microemulsions in the form of a stable, water-free combination of surfactants, co-surfactants and lipophilic phase, which creates a microemulsion when diluted in water or body fluids. These are developed to deliver sensitive drugs that would undergo hydrolysis in aqueous formulations. Hydrophobic drugs can also be delivered through these systems for oral administration as drug dispersed in gastrointestinal tract form a fine oil in water emulsion with mild agitations provided by gastric mobility which can subsequently be absorbed by lymphatic pathways (Tang et al., 2007; Kohli et al., 2010). Several self-microemulsifying or nanoemulsifying drug delivery systems have been developed and evaluated for oral delivery of insulin (Singnurkar and Gidwani, 2008; Sakloetsakun et al., 2013; Li et al., 2013a, 2014). These carriers have several demerits like low drug loading capacity and drug precipitation upon dilution.

Hydrogels and mucoadhesive drug delivery systems developed for oral delivery of insulin: Hydrogels are three-dimensional, cross-linked networks of water-soluble polymers. Hydrogels absorb large amounts of water and swell, while maintaining their three-dimensional structure. Hydrogels can be made from virtually any water-soluble polymer. The unique physical properties of hydrogels have sparked particular interest in their use in drug delivery applications. Their highly porous structure can easily be tuned by controlling the density of cross-links in the gel matrix and the affinity of the hydrogels for the aqueous environment in which they are swollen. Their porosity also permits loading of drugs into the gel matrix and subsequent drug release at a rate dependent on the diffusion coefficient of the small molecule or macromolecule through the gel network (Hoare and Kohane, 2008).

Complexation hydrogels are suitable candidates for oral delivery of proteins and peptides due to their abilities to respond to changes in pH in the GI tract and provide protection to the drugs from the harsh environment of the GI tract (Kavimandan et al., 2006; Nakamura et al., 2004). Hydrogels with improved delivery of oral insulin have been prepared by utilizing acrylates and their derivatives (Ichikawa and Peppas, 2003; Wood et al., 2010; Yin et al., 2010; Sajeesh et al., 2010a; Sonia and Sharma, 2013) and alginates (Woitiski et al., 2011; Deat-Laine et al., 2013a).

Hydrogels have several limitations such as non-homogeneity of drug loading, low tensile strength, low load-bearing, premature dissolution due to high water content and large pore sizes, flow away of the hydrogel from a target site (Hoare and Kohane, 2008).

Bio adhesive or mucoadhesive drug delivery systems are developed by incorporation of adhesive molecules into some kind of pharmaceutical formulation intended to stay in close contact with the absorption tissue, releasing the drug near to the action site, thereby increasing its bioavailability and promoting local or systemic effects (Woodley, 2001). Mucoadhesive drug delivery systems adhere to the mucous gel layer covering mucosal membranes establishing a high concentration gradient across intestinal epithelium leading to enhanced absorption of drugs (Ahuja et al., 1997; Andrews et al., 2009). Mucoadhesive delivery systems for improved insulin absorption includes chitosan (Pan et al., 2002; Rekha and Sharma, 2009, 2015; Fonte et al., 2012; Shelma and Sharma, 2013), acrylates (Whitehead et al., 2004; Sonia and Sharma, 2013) sodium salicylate and polyoxyethylene-9-lauryl ether (Hosny et al., 2001) have been proposed. The bioadhesive systems may however be affected by the mucous turnover of the GIT, which varies based on the site of absorption (Plate et al., 2002; Gowthamarajan and Kulkarni, 2003; Morishita and Peppas, 2006).

CONCLUSION

The development of drug delivery systems for oral administration of insulin continues to be pursued actively in academic institutions and pharmaceutical research centers. However, without much success so far, as no technique was able to deliver insulin orally with sufficient bioavailability. The different approaches have been used to reduce the enzymatic degradation of insulin and to increase its uptake or permeability across intestine so as to enhance the oral absorption. However, each approach has its own advantage and disadvantages. Formulations of insulin with protease inhibitors have typically shown inconsistent results. Penetration enhancers are not specific hence cause toxic consequences. Surfactants cause lysis of mucous membrane and may damage the lining of the gastrointestinal tract. Chelators such as EDTA cause depletion of calcium ions, which may be dangerous for the cell membrane. Release of insulin from colloidal carriers is inconsistent and slow causing slow and insufficient absorption. The hydrogels and mucoadhesive systems are affected by the mucous turnover of the gastrointestinal tract showing high variability in results. Despite of extensive research being perused for the sake of developing oral insulin, it has not yet been possible to come up with an efficient delivery system which could provide clinically significant bioavailability of oral insulin. An oral delivery system of insulin if developed successfully would have several advantages such as better control of diabetes, better compliance on insulin treatment and avoidance of side effects associated with long term injection therapy of insulin.

REFERENCES

  • Agarwal, V. and M.A. Khan, 2001. Current status of the oral delivery of insulin. Pharmaceutical Technology, October 2001, pp: 76-90. http://www.highbeam.com/doc/1P3-93246096.html.


  • Agrawal, A.K., H. Harde, K. Thanki and S. Jain, 2014. Improved stability and antidiabetic potential of insulin containing folic acid functionalized polymer stabilized multilayered liposomes following oral administration. Biomacromolecules, 15: 350-360.
    CrossRef    Direct Link    


  • Ahuja, A., R.K. Khar and J. Ali, 1997. Mucoadhesive drug delivery systems. Drug Dev. Ind. Pharm., 23: 489-515.
    CrossRef    Direct Link    


  • Allen, C., T. LeCaire, M. Palta, K. Daniels, M. Meredith and D.J. D'Alessio, 2001. Risk factors for frequent and severe hypoglycemia in type 1 diabetes. Diabetes Care, 24: 1878-1881.
    CrossRef    Direct Link    


  • American Diabetes Association, 2006. Diagnosis and classification of diabetes mellitus. Diabetes Care, 29: S43-S48.
    Direct Link    


  • American Diabetes Association, 2009. Diagnosis and classification of diabetes mellitus. Diabetes Care, 32: S62-S67.
    CrossRef    Direct Link    


  • Andrews, G.P., T.P. Laverty and D.S. Jones, 2009. Mucoadhesive polymeric platforms for controlled drug delivery. Eur. J. Pharm. Biopharm., 71: 505-518.
    CrossRef    PubMed    Direct Link    


  • Ansari, M.J., M.K. Anwer, S. Jamil, R. Al-Shdefat, B.E. Ali, M.M. Ahmad and M.N. Ansari, 2015. Enhanced oral bioavailability of insulin-loaded solid lipid nanoparticles: pharmacokinetic bioavailability of insulin-loaded solid lipid nanoparticles in diabetic rats. Drug delivery,
    CrossRef    


  • Arrieta-Molero, J.F., K. Aleck, M.K. Sinha, C.M. Brownscheidle, L.J. Shapiro and M.A. Sperling, 1982. Orally administered liposome-entrapped insulin in diabetic animals. Horm. Res. Paediatr., 16: 249-256.
    CrossRef    Direct Link    


  • Avadi, M.R., A.M.M. Sadeghi, N. Mohammadpour, S. Abedin, F. Atyabi, R. Dinarvand and M. Rafiee-Tehrani, 2010. Preparation and characterization of insulin nanoparticles using chitosan and Arabic gum with ionic gelation method. Nanomedicine, 6: 58-63.
    CrossRef    Direct Link    


  • Balabushevich, N.G., M.A. Pechenkin, E.D. Shibanova, D.V. Volodkin and E.V. Mikhalchik, 2013. Multifunctional polyelectrolyte microparticles for oral insulin delivery. Macromol. Biosci., 13: 1379-1388.
    CrossRef    Direct Link    


  • Bernkop-Schnurch, A., 1998. The use of inhibitory agents to overcome the enzymatic barrier to perorally administered therapeutic peptides and proteins. J. Controlled Release, 52: 1-16.
    CrossRef    Direct Link    


  • Boateng, J.S., J.C. Mitchell, H. Pawar and I. Ayensu, 2014. Functional characterisation and permeation studies of lyophilised thiolated chitosan xerogels for buccal delivery of insulin. Protein Peptide Lett., 21: 1163-1175.
    Direct Link    


  • Bott, S., U. Bott, M. Berger and I. Muhlhauser, 1997. Intensified insulin therapy and the risk of severe hypoglycaemia. Diabetologia, 40: 926-932.
    CrossRef    Direct Link    


  • Carino, G.P., J.S. Jacob and E. Mathiowitz, 2000. Nanosphere based oral insulin delivery. J. Controlled Release, 65: 261-269.
    CrossRef    Direct Link    


  • Chang, L.L., L.E. Stout, W.D. Wong, J.G. Buls and D.A. Rothenberger et al., 1997. Immunohistochemical localization of insulin-degrading enzyme along the rat intestine, in the human colon adenocarcinoma cell line (Caco-2) and in human ileum. J. Pharm. Sci., 86: 116-119.
    CrossRef    Direct Link    


  • Cho, Y.W. and M. Flynn, 1989. Oral delivery of insulin. Lancet, 2: 1518-1519.


  • Choudhari, K.B., V. Labhasetwar and A.K. Dorle, 1994. Liposomes as a carrier for oral administration of insulin: Effect of formulation factors. J. Microencapsulation, 11: 319-325.
    CrossRef    Direct Link    


  • Chuang, E.Y., K.J. Lin, F.Y. Su, H.L. Chen and B. Maiti et al., 2013. Calcium depletion-mediated protease inhibition and apical-junctional-complex disassembly via an EGTA-conjugated carrier for oral insulin delivery. J. Controlled Release, 169: 296-305.
    CrossRef    Direct Link    


  • Cilek, A., N. Celebi, F. Tirnaksiz and A. Tay, 2005. A lecithin-based microemulsion of rh-insulin with aprotinin for oral administration: Investigation of hypoglycemic effects in non-diabetic and STZ-induced diabetic rats. Int. J. Pharm., 298: 176-185.
    CrossRef    Direct Link    


  • Cui, M., W. Wu, L. Hovgaard, Y. Lu, D. Chen and J. Qi, 2015. Liposomes containing cholesterol analogues of botanical origin as drug delivery systems to enhance the oral absorption of insulin. Int. J. Pharm., 489: 277-284.
    CrossRef    Direct Link    


  • Damge, C., M. Socha, N. Ubrich and P. Maincent, 2010. Poly(ε-caprolactone)/eudragit nanoparticles for oral delivery of aspart-insulin in the treatment of diabetes. J. Pharm. Sci., 99: 879-889.
    CrossRef    Direct Link    


  • Dapergolas, G. and G. Gregoriadis, 1976. Hypoglycaemic effect of liposome-entrapped insulin administered intragastrically into rats. Lancet, 308: 824-827.
    CrossRef    Direct Link    


  • Das, N., M.K. Basu and M.K. Das, 1988. Oral application of insulin encapsulated liposomes. Biochem. Int., 16: 983-989.
    Direct Link    


  • Deat-Laine, E., V. Hoffart, G. Garrait, J.F. Jarrige, J.M. Cardot, M. Subirade and E. Beyssac, 2013. Efficacy of mucoadhesive hydrogel microparticles of whey protein and alginate for oral insulin delivery. Pharm. Res., 30: 721-734.
    CrossRef    Direct Link    


  • Deat-Laine, E., V. Hoffart, G. Garrait and E. Beyssac, 2013. Whey protein and alginate hydrogel microparticles for insulin intestinal absorption: Evaluation of permeability enhancement properties on Caco-2 cells. Int. J. Pharm., 453: 336-342.
    CrossRef    Direct Link    


  • Degim, Z., N. Unal, D. Essiz and U. Abbasoglu, 2004. The effect of various liposome formulations on insulin penetration across Caco-2 cell monolayer. Life Sci., 75: 2819-2827.
    CrossRef    Direct Link    


  • Del Curto, M.D., A. Maroni, A. Foppoli, L. Zema, A. Gazzaniga and M.E. Sangalli, 2009. Preparation and evaluation of an oral delivery system for time-dependent colon release of insulin and selected protease inhibitor and absorption enhancer compounds. J. Pharm. Sci., 98: 4661-4669.
    CrossRef    Direct Link    


  • Fasano, A. and S. Uzzau, 1997. Modulation of intestinal tight junctions by zonula occludens toxin permits enteral administration of insulin and other macromolecules in an animal model. J. Clin. Invest., 99: 1158-1164.
    CrossRef    Direct Link    


  • Fix, J.A., 1996. Oral Controlled release technology for peptides: Status and future prospects. Pharm. Res., 13: 1760-1764.
    CrossRef    Direct Link    


  • Fonte, P., F. Andrade, F. Araujo, C. Andrade, J. dos Neves and B. Sarmento, 2012. Chitosan-coated solid lipid nanoparticles for insulin delivery. Methods Enzymol., 508: 295-314.
    CrossRef    PubMed    Direct Link    


  • Funnell, M.M., 2007. Overcoming barriers to the initiation of insulin therapy. Clin. Diabetes, 25: 36-38.
    CrossRef    Direct Link    


  • Ghilzai, N.M.K., 2003. New developments in insulin delivery. Drug Dev. Ind. Pharm., 29: 253-265.
    CrossRef    Direct Link    


  • Gowthamarajan, K. and G.T. Kulkarni, 2003. Oral insulin-fact or fiction? Possibilities of achieving oral delivery for insulin. Resonance, 8: 38-46.
    CrossRef    Direct Link    


  • Graf, A., T. Rades and S.M. Hook, 2009. Oral insulin delivery using nanoparticles based on microemulsions with different structure-types: Optimisation and in vivo evaluation. Eur. J. Pharm. Sci., 37: 53-61.
    CrossRef    Direct Link    


  • Han, Y., H. Tian, P. He, X. Chen and X. Jing, 2009. Insulin nanoparticle preparation and encapsulation into poly(lactic-co-glycolic acid) microspheres by using an anhydrous system. Int. J. Pharm., 378: 159-166.
    CrossRef    Direct Link    


  • He, H., J. Sheng, A.E. David, Y.M. Kwon and J. Zhang et al., 2013. The use of low molecular weight protamine chemical chimera to enhance monomeric insulin intestinal absorption. Biomaterials, 34: 7733-7743.
    Direct Link    


  • Hoare, T.R. and D.S. Kohane, 2008. Hydrogels in drug delivery: Progress and challenges. Polymer, 49: 1993-2007.
    CrossRef    Direct Link    


  • Hosny, E.A., H.I. Al-Shora and M.M.A. Elmazar, 2002. Effect of bioadhesive polymers, sodium salicylate, polyoxyethylene-9-lauryl ether and method of preparation on the relative hypoglycemia produced by insulin enteric-coated capsules in diabetic beagle dogs. Drug Dev. Ind. Pharm., 28: 563-570.
    CrossRef    PubMed    Direct Link    


  • Hosny, E.A., I.M. El-Bagory and F.I. Al-Jenoobi, 2001. Enteric-coated insulin capsules: A combination with or a replacement of oral hypoglycaemic drugs. Bollettino Chimico Farmaceutico, 141: 379-384.
    PubMed    Direct Link    


  • Ichikawa, H. and N.A. Peppas, 2003. Novel complexation hydrogels for oral peptide delivery: In vitro evaluation of their cytocompatibility and insulin‐transport enhancing effects using Caco‐2 cell monolayers. J. Biomed. Mater. Res. Part A, 67: 609-617.
    CrossRef    Direct Link    


  • Irving, B., 2007. Nanoparticle drug delivery systems. Inno. Pharm. Biotechnol., 24: 58-62.


  • Jelvehgari, M., P.Z. Milani, M.R. Siahi-Shadbad, F. Monajjemzadeh, A. Nokhodchi, Z. Azari and H. Valizadeh, 2011. In vitro and in vivo evaluation of insulin microspheres containing protease inhibitor. Arzneimittel-Forschung, 61: 14-22.
    CrossRef    PubMed    Direct Link    


  • Johansson, U.B., S. Amsberg, L. Hannerz, R. Wredling, U. Adamson, H.J. Arnqvist and P.E. Lins, 2005. Impaired absorption of insulin aspart from lipohypertrophic injection sites. Diabetes Care, 28: 2025-2027.
    CrossRef    Direct Link    


  • Jose, S., J.F. Fangueiro, J. Smitha, T.A. Cinu, A.J. Chacko, K. Premaletha and E.B. Souto, 2013. Predictive modeling of insulin release profile from cross-linked chitosan microspheres. Eur. J. Med. Chem., 60: 249-253.
    CrossRef    Direct Link    


  • Kadir, A., M.T.M. Mokhtar and T.W. Wong, 2013. Nanoparticulate assembly of mannuronic acid‐and guluronic acid‐rich alginate: Oral insulin carrier and glucose binder. J. Pharm. Sci., 102: 4353-4363.
    CrossRef    Direct Link    


  • Kamei, N., M. Morishita, Y. Eda, N. Ida, R. Nishio and K. Takayama, 2008. Usefulness of cell-penetrating peptides to improve intestinal insulin absorption. J. Controlled Release, 132: 21-25.
    CrossRef    Direct Link    


  • Kamei, N., Y. Onuki, K. Takayama and M. Takeda‐Morishita, 2013. Mechanistic study of the uptake/permeation of cell‐penetrating peptides across a caco‐2 monolayer and their stimulatory effect on epithelial insulin transport. J. Pharm. Sci., 102: 3998-4008.
    CrossRef    PubMed    Direct Link    


  • Karamanidou, T., K. Karidi, V. Bourganis, K. Kontonikola, O. Kammona and C. Kiparissides, 2015. Effective incorporation of insulin in mucus permeating self-nanoemulsifying drug delivery systems. Eur. J. Pharm. Biopharm., (In Press).
    CrossRef    


  • Katayama, K., Y. Kato, H. Onishi, T. Nagai and Y. Machida, 2003. Double liposomes: Hypoglycemic effects of liposomal insulin on normal rats. Drug Dev. Ind. Pharm., 29: 725-731.
    CrossRef    Direct Link    


  • Kavimandan, N.J., E. Losi and N.A. Peppas, 2006. Novel delivery system based on complexation hydrogels as delivery vehicles for insulin-transferrin conjugates. Biomaterials, 27: 3846-3854.
    CrossRef    Direct Link    


  • Kimura, T., K. Sato, K. Sugimoto, R. Tao, T. Murakami, Y. Kurosaki and T. Nakayama, 1996. Oral administration of insulin as poly (vinyl alcohol)-gel spheres in diabetic rats. Biol. Pharm. Bull., 19: 897-900.
    CrossRef    Direct Link    


  • Kisel, M.A., L.N. Kulik, I.S. Tsybovsky, A.P. Vlasov and M.S. Vorob'yov, E.A. Kholodova and Z.V. Zabarovskaya, 2001. Liposomes with phosphatidylethanol as a carrier for oral delivery of insulin: Studies in the rat. Int. J. Pharm., 216: 105-114.
    CrossRef    Direct Link    


  • Kohli, K., S. Chopra, D. Dhar, S. Arora and R.K. Khar, 2010. Self-emulsifying drug delivery systems: An approach to enhance oral bioavailability. Drug Discovery Today, 15: 958-965.
    CrossRef    Direct Link    


  • Korytkowski, M., 2002. When oral agents fail: Practical barriers to starting insulin. Int. J. Obes. Relat. Metab. Disord., 26: S18-S24.
    PubMed    Direct Link    


  • Kraeling, M.E. and W.A. Ritschel, 1992. Development of a colonic release capsule dosage form and the absorption of insulin. Methods Findings Exp. Clin. Pharmacol., 14: 199-209.
    PubMed    Direct Link    


  • Lane, M.E. and O.I. Corrigan, 2006. Paracellular and transcellular pathways facilitate insulin permeability in rat gut. J. Pharmacy Pharmacol., 58: 271-275.
    CrossRef    Direct Link    


  • Lane, M.E., C.M. O'Driscoll and O.I. Corrigan, 2005. Quantitative estimation of the effects of bile salt surfactant systems on insulin stability and permeability in the rat intestine using a mass balance model. J. Pharm. Pharmacol., 57: 169-175.
    CrossRef    Direct Link    


  • Laurenti, O., A. Piccoli, C. Bravi, M. Cassone‐Faldetta and C. Bellini et al., 1996. Effect of aprotinin on insulin sensitivity in non‐insulin‐dependent diabetes mellitus. Diabetic Med., 13: 642-645.
    CrossRef    Direct Link    


  • Lawrence, M.J. and G.D. Rees, 2000. Microemulsion-based media as novel drug delivery systems. Adv. Drug Delivery Rev., 45: 89-121.
    CrossRef    Direct Link    


  • Li, C.L. and Y.J. Deng, 2004. Oil-based formulations for oral delivery of insulin. J. Pharm. Pharmacol., 56: 1101-1107.
    CrossRef    Direct Link    


  • Li, J., Y. Wang, L. Han, X. Sun, H. Yu and Y. Yu, 2012. Time-action profile of an oral enteric insulin formulation in healthy Chinese volunteers. Clin. Therapeut., 34: 2333-2338.
    CrossRef    Direct Link    


  • Li, L., T. Yi and C.W.K. Lam, 2014. Inhibition of human efflux transporter ABCC2 (MRP2) by Self-emulsifying drug delivery system: Influences of concentration and combination of excipients. J. Pharmacy Pharma. Sci., 17: 447-460.
    PubMed    Direct Link    


  • Li, X., J. Qi, Y. Xie, X. Zhang and S. Hu et al., 2013. Nanoemulsions coated with alginate/chitosan as oral insulin delivery systems: Preparation, characterization and hypoglycemic effect in rats. Int. J. Nanomed., 8: 23-32.
    CrossRef    PubMed    Direct Link    


  • Li, X., S. Guo, C. Zhu, Q. Zhu and Y. Gan et al., 2013. Intestinal mucosa permeability following oral insulin delivery using core shell corona nanolipoparticles. Biomaterials, 34: 9678-9687.
    CrossRef    Direct Link    


  • Li, Y., W. Xu, Z. Liao, B. Yao and X. Chen et al., 2004. Induction of long-term glycemic control in newly diagnosed type 2 diabetic patients is associated with improvement of β-cell function. Diabetes Care, 27: 2597-2602.
    CrossRef    Direct Link    


  • Liang, J.F. and V.C. Yang, 2005. Insulin-cell penetrating peptide hybrids with improved intestinal absorption efficiency. Biochem. Biophys. Res. Commun., 335: 734-738.
    CrossRef    Direct Link    


  • Lin, Y.H., C.T. Chen, H.F. Liang, A.R. Kulkarni, P. Lee, C.H. Chen and H.W. Sung, 2007. Novel nanoparticles for oral insulin delivery via the paracellular pathway. Nanotechnology, Vol. 18.


  • Lin, Y.H., F.L. Mi, C.T. Chen, W.C. Chang, S.F. Peng, H.F. Liang and H.W. Sung, 2007. Preparation and characterization of nanoparticles shelled with chitosan for oral insulin delivery. Biomacromolecules, 8: 146-152.
    CrossRef    Direct Link    


  • Liu, H., R. Tang, W.S. Pan, Y. Zhang and H. Liu, 2003. Potential utility of various protease inhibitors for improving the intestinal absorption of insulin in rats. J. Pharm. Pharmacol., 55: 1523-1529.
    CrossRef    Direct Link    


  • Liu, J., T. Gong, C. Wang, Z. Zhong and Z. Zhang, 2007. Solid lipid nanoparticles loaded with insulin by sodium cholate-phosphatidylcholine-based mixed micelles: Preparation and characterization. Int. J. Pharm., 340: 153-162.
    CrossRef    Direct Link    


  • Manosroi, A., N. Khositsuntiwong, C. Komno, W. Manosroi, R.G. Werner and J. Manosoi, 2011. Chemical stability and cytotoxicity of human insulin loaded in cationic DPPC/CTA/DDAB liposomes. J. Biomed. Nanotechnol., 7: 308-316.
    Direct Link    


  • Mansourpour, M., R. Mahjub, M. Amini, S.N. Ostad, E.S. Shamsa, M. Rafiee-Tehrani and F.A. Dorkoosh, 2015. Development of acid-resistant alginate/trimethyl chitosan nanoparticles containing cationic β-cyclodextrin polymers for insulin oral delivery. AAPS PharmSciTech., 16: 952-962.
    CrossRef    Direct Link    


  • Mohanraj, V.J. and Y. Chen, 2006. Nanoparticles: A review. Trop. J. Pharma. Res., 5: 561-573.
    Direct Link    


  • Morimoto, K., Y. Uehara, K. Iwanaga and M. Kakemi, 2000. Effects of sodium glycocholate and protease inhibitors on permeability of TRH and insulin across rabbit trachea. Pharmaceutica Acta Helvetiae, 74: 411-415.
    CrossRef    Direct Link    


  • Morishita, I., M. Morishita, K. Takayama, Y. Machida and T. Nagai, 1993. Enteral insulin delivery by microspheres in 3 different formulations using Eudragit L100 and S100. Int. J. Pharm., 91: 29-37.
    CrossRef    Direct Link    


  • Morishita, M. and N.A. Peppas, 2006. Is the oral route possible for peptide and protein drug delivery? Drug Discovery Today, 11: 905-910.
    CrossRef    Direct Link    


  • Morishita, M., N. Kamei, J. Ehara, K. Isowa and K. Takayama, 2007. A novel approach using functional peptides for efficient intestinal absorption of insulin. J. Controlled Release, 118: 177-184.
    CrossRef    Direct Link    


  • Mukherjee, S., S. Ray and R.S. Thakur, 2009. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. Indian J. Pharmaceut. Sci., 71: 349-358.
    CrossRef    PubMed    Direct Link    


  • Nakamura, K., R.J. Murray, J.I. Joseph, N.A. Peppas, M. Morishit and A.M. Lowman, 2004. Oral insulin delivery using P(MAA-g-EG) hydrogels: Effects of network morphology on insulin delivery characteristics. J. Control Rel., 95: 589-599.
    CrossRef    PubMed    Direct Link    


  • Nielsen, E.J.B., S. Yoshida, N. Kamei, R. Iwamae and E.S. Khafagyet al., 2014. In vivo proof of concept of oral insulin delivery based on a co-administration strategy with the cell-penetrating peptide penetratin. J. Controlled Release, 189: 19-24.
    CrossRef    Direct Link    


  • Niu, M., Y. Lu, L. Hovgaard and W. Wu, 2011. Liposomes containing glycocholate as potential oral insulin delivery systems: Preparation, in vitro characterization and improved protection against enzymatic degradation. Int. J. Nanomed., 6: 1155-1166.
    CrossRef    Direct Link    


  • Niu, M., Y. Lu, L. Hovgaard, P. Guan and Y. Tan et al., 2012. Hypoglycemic activity and oral bioavailability of insulin-loaded liposomes containing bile salts in rats: the Effect of cholate type, particle size and administered dose. Eur. J. Pharm. Biopharm., 81: 265-272.
    CrossRef    PubMed    Direct Link    


  • Niu, M., Y. Tan, P. Guan, L. Hovgaard and Y. Lu et al., 2014. Enhanced oral absorption of insulin-loaded liposomes containing bile salts: A mechanistic study. Int. J. Pharm., 460: 119-130.
    CrossRef    PubMed    Direct Link    


  • Ogiso, T., M. Iwaki, T. Tanino, S. Nishioka, K. Higashi and M. Kamo, 1997. In vitro skin penetration and degradation of enkephalin, elcatonin and insulin. Biol. Pharm. Bull., 20: 54-60.
    CrossRef    Direct Link    


  • Pamnani, D., 2008. Reality check on oral insulin. Pharma Express, 3: 16-31.
    Direct Link    


  • Pan, Y., Y.J. Li, H.Y. Zhao, J.M. Zheng and H. Xu et al., 2002. Bioadhesive polysaccharide in protein delivery system: Chitosan nanoparticles improve the intestinal absorption of insulin in vivo. Int. J. Pharm., 249: 139-147.
    CrossRef    PubMed    Direct Link    


  • Park, S.H., J.H. Kwon, S.H. Lim, H.W. Park and C.W. Kim, 2007. Characterization of human insulin microcrystals and their absorption enhancement by protease inhibitors in rat lungs. Int. J. Pharm., 339: 205-212.
    CrossRef    Direct Link    


  • Park, S.J., S.G. Choi, E. Davaa and J.S. Park, 2011. Encapsulation enhancement and stabilization of insulin in cationic liposomes. Int. J. Pharm., 415: 267-272.
    CrossRef    Direct Link    


  • Patel, D.G., W.A. Ritschel, P. Chalasani and S. Rao, 1991. Biological activity of insulin in microemulsion in mice. J. Pharm. Sci., 80: 613-614.
    PubMed    Direct Link    


  • Patki, V.P. and S.H. Jagasia, 1996. Progress made in non-invasive insulin delivery. Indian J. Pharmacol., 28: 143-151.
    Direct Link    


  • Perera, G., M. Greindl, T.F. Palmberger and A. Bernkop-Schnurch, 2009. Insulin-loaded poly(acrylic acid)-cysteine nanoparticles: Stability studies towards digestive enzymes of the intestine. Drug Delivery, 16: 254-260.
    CrossRef    Direct Link    


  • Petkowicz, J., A. Byra and T. Szumilo, 1989. Hypoglycemic effect of liposome-entrapped insulin administered by various routes into normal rats. Polish J. Pharmacol. Pharm., 41: 299-304.
    PubMed    Direct Link    


  • Plate, N.A., I.L. Valuev, G.A. Sytov and L.I. Valuev, 2002. Mucoadhesive polymers with immobilized proteinase inhibitors for oral administration of protein drugs. Biomaterials, 23: 1673-1677.
    CrossRef    Direct Link    


  • Qi, R. and Q.N. Ping, 2004. Gastrointestinal absorption enhancement of insulin by administration of enteric microspheres and SNAC to rats. J. Microencapsulation: Micro Nano Carriers, 21: 37-45.
    CrossRef    PubMed    Direct Link    


  • Rachmawati, H., B.M. Haryadi, K. Anggadiredja and V. Suendo, 2015. Intraoral film containing insulin-phospholipid microemulsion: Formulation and in vivo hypoglycemic activity study. AAPS PharmSciTech., 16: 692-703.
    CrossRef    Direct Link    


  • Radwan, M.A. and H.Y. Aboul-Enein, 2002. The effect of oral absorption enhancers on the in vivo performance of insulin-loaded poly(ethylcyanoacrylate) nanospheres in diabetic rats. J. Microencapsulation: Micro Nano Carriers, 19: 225-235.
    CrossRef    Direct Link    


  • Rekha, M.R. and C.P. Sharma, 2009. Synthesis and evaluation of lauryl succinyl chitosan particles towards oral insulin delivery and absorption. J. Control Release, 135: 144-151.
    CrossRef    Direct Link    


  • Rekha, M.R. and C.P. Sharma, 2015. Simultaneous effect of thiolation and carboxylation of chitosan particles towards mucoadhesive oral insulin delivery applications: An in vitro and in vivo evaluation. J. Biomed. Nanotechnol., 11: 165-176.
    Direct Link    


  • Rieux, A.D., V. Fievez, M. Garinot, Y.J. Schneider and V. Preat, 2006. Nanoparticles as potential oral delivery systems of proteins and vaccines: A mechanistic approach. J. Controlled Release, 116: 1-27.
    CrossRef    Direct Link    


  • Rowland, R.N. and J.F. Woodley, 1981. Uptake of free and liposome-entrapped insulin by rat intestinal sacs in vitro. Biosci. Rep., 1: 345-352.
    CrossRef    Direct Link    


  • Ryan, E.A., S. Imes and C. Wallace, 2004. Short-term intensive insulin therapy in newly diagnosed type 2 diabetes. Diabetes Care, 27: 1028-1032.
    CrossRef    Direct Link    


  • Sadeghi, A.M.M., F.A. Dorkoosh, M.R. Avadi, M. Weinhold and A. Bayat et al., 2008. Permeation enhancer effect of chitosan and chitosan derivatives: Comparison of formulations as soluble polymers and nanoparticulate systems on insulin absorption in caco-2 cells. Eur. J. Pharm. Biopharm., 70: 270-278.
    CrossRef    PubMed    Direct Link    


  • Saffran, M., B. Pansky, G.C. Budd and F.E. Williams, 1997. Insulin and the gastrointestinal tract. J. Controlled Release, 46: 89-98.
    CrossRef    Direct Link    


  • Sajeesh, S. and C.P. Sharma, 2006. Cyclodextrin-insulin complex encapsulated polymethacrylic acid based nanoparticles for oral insulin delivery. Int. J. Pharm., 325: 147-154.
    CrossRef    PubMed    Direct Link    


  • Sajeesh, S., C. Vauthier, C. Gueutin, G. Ponchel and C.P. Sharma, 2010. Thiol functionalized polymethacrylic acid-based hydrogel microparticles for oral insulin delivery. Acta Biomaterialia, 6: 3072-3080.
    CrossRef    Direct Link    


  • Sajeesh, S., K. Bouchemal, V. Marsaud, C. Vauthier and C.P. Sharma, 2010. Cyclodextrin complexed insulin encapsulated hydrogel microparticles: An oral delivery system for insulin. J. Controlled Release, 147: 377-384.
    CrossRef    Direct Link    


  • Sakloetsakun, D., S. Dunnhaupt, J. Barthelmes, G. Perera and A. Bernkop-Schnurch, 2013. Combining two technologies: Multifunctional polymers and self-nanoemulsifying drug delivery system (SNEDDS) for oral insulin administration. Int. J. Biol. Macromolecules, 61: 363-372.
    CrossRef    Direct Link    


  • Salamat-Miller, N. and T.P. Johnston, 2005. Current strategies used to enhance the paracellular transport of therapeutic polypeptides across the intestinal epithelium. Int. J. Pharm., 294: 201-216.
    CrossRef    Direct Link    


  • Sarmento, B., S. Martins, D. Ferreira and E.B. Souto, 2007. Oral insulin delivery by means of solid lipid nanoparticles. Int. J. Nanomed., 2: 743-749.
    Direct Link    


  • Scarlett, J.A., R.S. Gray, J. Griffin, J.M. Olefsky and O.G. Kolterman, 1982. Insulin treatment reverses the insulin resistance of type II diabetes mellitus. Diabetes Care, 5: 353-363.
    CrossRef    Direct Link    


  • Shah, R.B., F. Ahsan and M.A. Khan, 2002. Oral delivery of proteins: Progress and prognostication. Crit. Rev. Ther. Drug Carrier Syst., 19: 135-169.
    CrossRef    Direct Link    


  • Sharma, G., K. Wilson, C.F. van der Walle, N. Sattar, J.R. Petrie and M.R. Kumar, 2010. Microemulsions for oral delivery of insulin: Design, development and evaluation in streptozotocin induced diabetic rats. Eur. J. Pharm. Biopharm., 76: 159-169.
    CrossRef    Direct Link    


  • Shelma, R. and C.P. Sharma, 2013. In vitro cell culture evaluation and in vivo efficacy of amphiphilic chitosan for oral insulin delivery. J. Biomed. Nanotechnol., 9: 167-176.
    Direct Link    


  • Shi, K., F. Cui, H. Yamamoto and Y. Kawashima, 2008. Investigation of drug loading and in vitro release mechanisms of insulin-lauryl sulfate complex loaded PLGA nanoparticles. Die Pharmazie-Int. J. Pharm. Sci., 63: 866-871.
    Direct Link    


  • Singnurkar, P.S. and S.K. Gidwani, 2008. Insulin-egg yolk dispersions in self microemulsifying system. Indian J. Pharm. Sci., 70: 727-732.
    CrossRef    Direct Link    


  • Socha, M., A. Sapin, C. Damge and P. Maincent, 2009. Influence of polymers ratio on insulin-loaded nanoparticles based on poly-ε-caprolactone and Eudragit® RS for oral administration. Drug Delivery, 16: 430-436.
    CrossRef    Direct Link    


  • Sonia, T.A. and C.P. Sharma, 2013. In vitro evaluation of quaternized polydimethylaminoethylmethacrylate sub-microparticles for oral insulin delivery. J. Biomater. Applic., 28: 62-73.
    CrossRef    Direct Link    


  • Su, F.Y., K.J. Lin, K. Sonaje, S.P. Wey and T.C. Yen et al., 2012. Protease inhibition and absorption enhancement by functional nanoparticles for effective oral insulin delivery. Biomaterials, 33: 2801-2811.
    Direct Link    


  • Takeuchi, H., H. Yamamoto, T. Niwa, T. Hino and Y. Kawashima, 1996. Enteral absorption of insulin in rats from mucoadhesive chitosan-coated liposomes. Pharm. Res., 13: 896-901.
    CrossRef    Direct Link    


  • Tang, J.L., J. Sun and Z.G. He, 2007. Self-emulsifying drug delivery systems: Strategy for improving oral delivery of poorly soluble drugs. Curr. Drug Ther., 2: 85-93.
    Direct Link    


  • Tasaka, Y., K. Marumo, Y. Inoue and Y. Hirata, 1989. Degradation of 125I-glucagon,-pancreatic polypeptide and-insulin by acid saline extract of rat submaxillary gland and their protection by proteinase inhibitors. Endocrinologia Japonica, 36: 47-53.
    CrossRef    Direct Link    


  • Touitou, E., M. Donbrow and A. Rubinstein, 1980. Effective intestinal absorption of insulin in diabetic rats using a new formulation approach. J. Pharm. Pharmacol., 32: 108-110.
    CrossRef    Direct Link    


  • Tozaki, H., J. Nishioka, J. Komoike, N. Okada and T. Fujita et al., 2001. Enhanced absorption of insulin and (Asu1,7)eel-calcitonin using novel azopolymer-coated pellets for colon-specific drug delivery. J. Pharm. Sci., 90: 89-97.
    CrossRef    Direct Link    


  • Turner, R.C., C.A. Cull, V. Frighi, R.R. Holman and UK Prospective Diabetes Study Group, 1999. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus: Progressive requirement for multiple therapies (UKPDS 49). J. Am. Med. Assco., 281: 2005-2012.
    CrossRef    Direct Link    


  • Uchiyama, T., T. Sugiyama, Y.S. Quan, A. Kotani and N. Okada et al., 1999. Enhanced permeability of insulin across the rat intestinal membrane by various absorption enhancers: Their intestinal mucosal toxicity and absorption-enhancing mechanism of n-Lauryl-β-D-maltopyranoside. J. Pharm. Pharmacol., 51: 1241-1250.
    CrossRef    Direct Link    


  • Vardar, B. and S. Kizilci, 2007. Incidence of lipohypertrophy in diabetic patients and a study of influencing factors. Diabetes Res. Clin. Pract., 77: 231-236.
    CrossRef    Direct Link    


  • Vemuri, S. and C.T. Rhodes, 1995. Preparation and characterization of liposomes as therapeutic delivery systems: A review. Pharmaceutica Acta Helvetiae, 70: 95-111.
    CrossRef    Direct Link    


  • WHO., 2005. Diabetes mellitus. Fact sheet No. 138, Media Centre-World Health Organization, Geneva, Switzerland. http://www.who.int/mediacentre/factsheets/fs138/en/.


  • Wahl, P.W., P.J. Savage, B.M. Psaty, T.J. Orchard, J.A. Robbins and R.P. Tracy, 1998. Diabetes in older adults: Comparison of 1997 American Diabetes Association classification of diabetes mellitus with 1985 WHO classification. Lancet, 352: 1012-1015.
    CrossRef    Direct Link    


  • Wang, S.L., S.Y. Xie, L.Y. Zhu, F.H. Wang and W.Z. Zhou, 2009. Effects of poly (lactic-co-glycolic acid) as a co-emulsifier on the preparation and hypoglycaemic activity of insulin-loaded solid lipid nanoparticles. IET Nanobiotechnol., 3: 103-108.
    CrossRef    Direct Link    


  • Wang, W., 1996. Oral protein drug delivery. J. Drug Target., 4: 195-232.
    CrossRef    Direct Link    


  • Watnasirichaikul, S., N.M. Davies, T. Rades and I.G. Tucker, 2000. Preparation of biodegradable insulin nanocapsules from biocompatible microemulsions. Pharm. Res., 17: 684-689.
    CrossRef    Direct Link    


  • Watnasirichaikul, S., T. Rades, I.G. Tucker and N.M. Davies, 2002. In‐vitro release and oral bioactivity of insulin in diabetic rats using nanocapsules dispersed in biocompatible microemulsion. J. Pharm. Pharmacol., 54: 473-480.
    CrossRef    Direct Link    


  • Weng, J., Y. Li, W. Xu, L. Shi and Q. Zhang et al., 2008. Effect of intensive insulin therapy on β-cell function and glycaemic control in patients with newly diagnosed type 2 diabetes: A multicentre randomised parallel-group trial. Lancet, 371: 1753-1760.
    CrossRef    Direct Link    


  • Whitehead, K., Z. Shen and S. Mitragotri, 2004. Oral delivery of macromolecules using intestinal patches: Applications for insulin delivery. J. Controlled Release, 98: 37-45.
    CrossRef    Direct Link    


  • Woitiski, C.B., B. Sarmento, R.A. Carvalho, R.J. Neufeld and F. Veiga, 2011. Facilitated nanoscale delivery of insulin across intestinal membrane models. Int. J. Pharm., 412: 123-131.
    CrossRef    Direct Link    


  • Wong, T.W. and N. Sumiran, 2014. Oral calcium pectinate‐insulin nanoparticles: Influences of alginate, sodium chloride and Tween 80 on their blood glucose lowering performance. J. Pharm. Pharmacol., 66: 646-657.
    CrossRef    Direct Link    


  • Wood, K.M., G.M. Stone and N.A. Peppas, 2010. The effect of complexation hydrogels on insulin transport in intestinal epithelial cell models. Acta Biomaterialia, 6: 48-56.
    CrossRef    Direct Link    


  • Woodley, J., 2001. Bioadhesion, new possibilities for drug administration? Clin. Pharmacokinet., 40: 77-84.
    CrossRef    Direct Link    


  • Wu, Z.H., Q.N. Ping, Y. Wei and J.M. Lai, 2004. Hypoglycemic efficacy of chitosan-coated insulin liposomes after oral administration in mice. Acta Pharmacologica Sinica, 25: 966-972.
    Direct Link    


  • Yamamoto, A., E. Hayakawa, Y. Kato, A. Nishiura and V.H. Lee, 1992. A mechanistic study on enhancement of rectal permeability to insulin in the albino rabbit. J. Pharmacol. Exp. Ther., 263: 25-31.
    Direct Link    


  • Yamamoto, A., T. Taniguchi, K. Rikyuu, T. Tsuji, T. Fujita, M. Murakami and S. Muranishi, 1994. Effects of various protease inhibitors on the intestinal absorption and degradation of insulin in rats. Pharm. Res., 10: 1496-1500.
    CrossRef    PubMed    Direct Link    


  • Yin, L., J. Ding, C. He, L. Cui, C. Tang and C. Yin, 2009. Drug permeability and mucoadhesion properties of thiolated trimethyl chitosan nanoparticles in oral insulin delivery. Biomaterials, 30: 5691-5700.
    CrossRef    Direct Link    


  • Yin, L., J. Ding, J. Zhang, C. He, C. Tang and C. Yin, 2010. Polymer integrity related absorption mechanism of superporous hydrogel containing interpenetrating polymer networks for oral delivery of insulin. Biomaterials, 31: 3347-3356.
    CrossRef    Direct Link    


  • Zhang, N., Q. Ping, G. Huang, W. Xu, Y. Cheng and X. Han, 2006. Lectin-modified solid lipid nanoparticles as carriers for oral administration of insulin. Int. J. Pharm., 327: 153-159.
    CrossRef    Direct Link    


  • Zhang, N., Q.N. Ping, G.H. Huang and W.F. Xu, 2005. Investigation of lectin-modified insulin liposomes as carriers for oral administration. Int. J. Pharm., 294: 247-259.
    CrossRef    Direct Link    


  • Zhang, X.W., J. Qi, Y. Lu, X.W. Hu, W. He and W. Wu, 2014. Enhanced hypoglycemic effect of biotin-modified liposomes loading insulin: Effect of formulation variables, intracellular trafficking and cytotoxicity. Nanoscale Res. Lett., Vol. 9.
    CrossRef    


  • Zhang, X.W., J. Qi, Y. Lu, X.Y. Li, W. He and W. Wu, 2014. Biotinylated liposomes as potential carriers for the oral delivery of insulin. Nanomed.: Nanotechnol. Biol. Med., 10: 167-176.
    CrossRef    PubMed    Direct Link    


  • Zhang, Z., H. Lv and J. Zhou, 2009. Novel solid lipid nanoparticles as carriers for oral administration of insulin. Die Pharmazie-Int. J. Pharm. Sci., 64: 574-578.
    PubMed    Direct Link    


  • Zhang, Z.H., Y.L. Zhang, J.P. Zhou and H.X. Lv, 2012. Solid lipid nanoparticles modified with stearic acid-octaarginine for oral administration of insulin. Int. J. Nanomed., 7: 3333-3339.
    CrossRef    Direct Link    


  • Zhao, L., D. Sun, F. Cao, T. Yin and H. Wang, 2009. Can insulin resistance be reversed by insulin therapy? Med. Hypotheses, 72: 34-35.
    Direct Link    


  • Zhu, S., S. Chen, Y. Gao, F. Guo and F. Li et al., 2015. Enhanced oral bioavailability of insulin using PLGA nanoparticles co-modified with cell-penetrating peptides and Engrailed secretion peptide (Sec). Drug Delivery.
    CrossRef    


  • Zhu, X., W. Shan, P. Zhang, Y. Jin and S. Guan et al., 2014. Penetratin derivative-based nanocomplexes for enhanced intestinal insulin delivery. Mol. Pharm., 11: 317-328.
    CrossRef    Direct Link    


  • Ziv, E., O. Lior and M. Kidron, 1987. Absorption of protein via the intestinal wall: A quantitative model. Biochem. Pharmacol., 36: 1035-1039.
    CrossRef    Direct Link    


  • Lisa, N., 2007. New criteria for diagnosing metabolic syndrome in teens. Medscape Medical News, February 21, 2007. http://www.medscape.org/viewarticle/552466.


  • Kumar, M.N.V.R., 2000. Nano and microparticles as controlled drug delivery devices. J. Pharm. Pharmaceut. Sci., 3: 234-258.
    PubMed    Direct Link    


  • Muller, R.H. and K. Peters, 1998. Nanosuspensions for the formulation of poorly soluble drugs: I. Preparation by a size-reduction technique. Int. J. Pharma., 160: 229-237.
    CrossRef    Direct Link    

  • © Science Alert. All Rights Reserved