769 International Journal of Progressive Sciences and Technologies (IJPSAT) ISSN: 2509-0119. © 2020 International Journals of Sciences and High Technologies http://ijpsat.ijsht-journals.org Vol. 24 No. 1 December 2020, pp. 586-596

Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

Maged Naser1, Mohamed M.Nasr2.and Lamia H. Shehata3 1Mazahmiya Hospital, Ministry of Health, Kingdom of Saudi Arabia, Department of ob/gyn, 2King Fahd Hospital, Ministry of Health, Kingdom of Saudi Arabia, Department of , 3Care National hospital. Department of

Abstract – Customary techniques for require steady and tedious glucose monitoring (GM) and insulin infusions, affecting quality of life. The worldwide diabetic population is required to increment to 439 million, with roughly US$490 billion in medical services consumptions by 2030, forcing a huge trouble on medical care systems around the world. Ongoing advances in nanotechnology have arisen as promising elective methodologies for the management of diabetes. For instance, implantable nano sensors are being created for nonstop GM, new nanoparticle (NP)- based imaging approaches that evaluate subtle changes in β cell mass can encourage early diagnosis, and nano technology-based insulin delivery strategies are being investigated as novel treatments. Here, we give an all- encompassing rundown of this quickly propelling field gathering all viewpoints relating to the management of diabetes.

Keywords – Diabetes Mellitus, Nanotechnology, Mnps, Novel Drug Delivery System.

I. INTRODUCTION Diabetes mellitus is a gathering of metabolic issues that is portrayed by raised blood glucose levels driven by insulin insufficiency or opposition [1]. The number of people influenced by this issue is consistently expanding with an expected 7.7% of the overall grown-up population (439 million individuals) experiencing the problem by 2030, carrying with it a related increment in the overall monetary weight from US$376 billion out of 2010 to US$490 billion by 2030 [2]. The two types of diabetes were first arranged by Percival Hemsworth in 1936 [3]. Type 1 diabetes represents ~10% of the diabetic patient population, and is related with an inadequacy in insulin [4] brought about via immune system decimation of islet β cells in the pancreas [5]. Type 2 diabetes, by contrast, is related with insulin resistance, the hosing of the body's reaction to insulin, which thus prompts . This type of diabetes has been firmly associated to way of life propensities and can develop over time in an individual [6]. As per the American Diabetes Association, appropriate glucose control (i.e., b150 mg/dl fasting glucose or 7% hemoglobin A1c) is significant for forestalling downstream inconveniences in diabetic patients [7]. prolonged hyperglycemia, for instance, has been linked to a multitude of microvascular (e.g., eye, nerve, and kidney disease) and macrovascular complications (cardiovascular disease) [7]. Accordingly, ordinary treatments for the two kinds of diabetes ordinarily comprise of successive GM and insulin organization (e.g., through subcutaneous infusions or insulin pumps) all through the day. Be that as it may, irregular GM and poor patient adherence, driven by a multitude of factors including pain and the repetitiveness of the technique [8], frequently lead to unpredictable insulin doses, which could bring about uncontrolled hyperglycemia, , seizures, unconsciousness, or death [9]. Furthermore, in adolescents with diabetes, developmental, psychosocial, and caregiver challenges add further complexities to diabetes care [10].

Corresponding Author: Maged Naser 586

Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

Albeit persistent glucose monitors and insulin pumps have been developed to address these issues, and ongoing shut circle systems that incorporate the two devices (wherein pumps). Following accomplishment in other clinical territories, nanotechnology is as of now creating immense promise for alternative strategies for diabetes management. Magnetic nanoparticle-based imaging of β cell mass and immune cell activity, as well as quantum spot (QD)- empowered gas sensors, offer potential new roads for early recognition. Multiwalled-carbon nanotubes, graphene nanocomposites and QDs provide discrete sensors with improved sensitivity and efficiency for glucose and insulin monitoring. Cell supported by nanofiber-based scaffolds or immunoselection membranes, and glucose-responsive nanogels and nanovesicles offer novel approaches to controlled insulin release. intelligently respond by delivering appropriate insulin amounts in response to glucose data, have been introduced, glucose management within an acceptable range has been shown to occur at best 70% of the time using these systems [11,12]. As such, there is still a need for improved diabetes management tools. Recent advances in nanotechnology have shown promise for the management of a wide variety of medical conditions [13–17]. Here, we provide a holistic over view on this rapidly growing field, compiling all aspects pertaining to the management of diabetes, including diagnosis, monitoring, and treatment.

II. PATHOPHYSIOLOGY OF DIABETES The homeostasis of glucose in the body is kept up by various hormones. Any way two hormones to be specific, insulin and glucagon play a prevailing part in the guideline of glucose homeostasis [18]. Insulin is discharged by β cells when the grouping of glucose rises. Insulin diminishes the degree of blood glucose by the same token a) By repressing the creation of glucose from liver by glycogenolysis what's more, gluconeogenesis [19], or b) By expanding the take-up of glucose by liver, muscle and fat tissue [19]. Glucagon is discharged by α cells of pancreas when the grouping of glucose is low. Glucagon acts by; a) Antagonizing the impact of insulin by improving the cycles like glycogenolysis and gluconeogenesis in liver, (b) notwithstanding glucagon, cortisol and catecholamines additionally increments the plasma glucose levels [20]. Different hormones which are engaged with support of ordinary glucose level are amylin (a 37 amino acid peptide), glucagon like Peptide – 1 (GLP-1) (a 30 amino acid peptide) and Glucose subordinate insulinotropic polypeptide (GIP) (a 42 amino acid peptide) [21,22]. Amylin is emitted alongside insulin. It diminishes gastric discharging, which upgrades glucose ingestion after a supper consumption. GLP and GIP are incretin or peptide got from the gut. These incretins encourage the union and discharge of insulin from β cells of pancreas [23]. Glucose isn't retained from digestive tract or by cells requiring energy unreservedly. So, the appropriation of glucose to the cells is finished by glucose carriers. The Glucose carriers are a group of film bound glycoproteins and are characterized into two sorts [24]. Sodium glucose co-carrier (SGLT) Facilitative glucose carrier (GLUT) Diabetes mellitus is arranged into two significant sub-types and the causes related remaining parts differential [25]. • Type – I DM (T1DM): The insusceptible system mistakenly attacks the β cells of pancreas where genes play a vital role. • Type – II DM (T2DM): Interplay of hereditary genes and way of life factors plays an imperative role, being obese or overweight increases the associated risks.

III. BIOMEDICINE AND PHARMACOTHERAPY • In the Event Of T1DM, Being An Adult Or Teen, Risk Of Diabetes Increments In The Event That The Parent Or Kin Is Diabetic [26].

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 587 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

• In the Event Of T2DM, Hazard Increments Because Of Specific Variables Like Being Overweight, Diet Propensities, Age More Than 45yrs, having history of diabetes in the family, actually less dynamic, or having pre-diabetes or then again gestational diabetes, elevated cholesterol or fatty acids levels [27–30]. • Risk of gestational diabetes increments if the individual is overweight with the age more than 25 years, having gestational diabetes during past pregnancy, brought forth an infant weighing around more than 9 pounds, have a family background of T2DM and have polycystic ovary disorder (PCOS) [31]. Diabetes has a great deal of entanglements related with it as high blood sugar harms organs and tissues all through the body. The more extended the body manages high glucose levels, the risk of extra inconveniences surfaces up. Inconveniences of diabetes can be microvascular which incorporates nephropathy, retinopathy, loss of vision and macrovascular like heart sicknesses, cardiovascular failure, stroke, and neuropathy, diseases what's more, injuries that don't recuperate bacterial and contagious contaminations, sorrow and dementia [32]. Any individual with striking manifestations of diabetes mellitus or at a danger of it should be tried consistently, for diagnosing prediabetes and diabetes, a few blood tests can be done, in particular: • Fasting plasma glucose (FPG): helps in estimating blood glucose after fasting for 8 hours. • HbA1C test: helps in estimating glucose levels over the time of past a quarter of a year. For diagnosing gestational diabetes, blood tests are done inside 24th also, 28th weeks of pregnancy, and tried for glucose challenge test and three-hour glucose tolerance test [33,34].

IV. NANOTECHNOLOGY APPLICATIONS IN DIAGNOSTICS Early and reliable diagnostic testing might identify individuals in whom early intervention with lifestyle management or pharmacological approaches might prevent dysglycemia or even the onset of disease. Previous studies demonstrated that early glycemia management prevented or delayed a number of disease-related complications [37,38], suggesting that earlier diagnosis and subsequent management should be of priority. However, traditional testing methods often fall short in this regard [39]. Conventional diagnostic techniques for diabetes include analyzing fasting glucose levels, hemoglobin A1c levels, or oral glucose tolerance tests. In the setting of clinical trials, the measurement of autoantibodies is often used as a diagnostic test to identify individuals at high risk for the development of diabetes, and in the clinical setting autoantibodies are now and then used to recognize those with type 1 diabetes when the nature of diabetes is unclear [40]. These techniques are considered painful by certain patients, and depend on glucose estimations or antibodies titers, which can differ dependent on numerous elements, including age, difficult period, and other physiological conditions [41]. Also, the sign of disease side effects, for example, hyperglycemia, regularly doesn't turn out to be clinically obvious until years after disease onset, which precludes early diagnosis [22]. To address the shortcomings of conventional demonstrative apparatuses, extraordinary kinds of nanotechnologies have been created to possibly empower earlier and non-invasive detection of diabetes. Portrayal of Immune Cell Activity and β Cell Mass In spite of the fact that modifications in β cell work (i.e., loss of insulin secretion) are apparent in both type 1 and type 2 diabetes, evaluation of β cell mass can possibly distinguish those people in whom potential β cell restoration treatments might be helpful. Modifications in β cell mass are not just a hallmark of immune system type 1 diabetes, however, could likewise be accelerated by supported insulin resistance under type 2 diabetes [4,23]. New advancements fit for estimating changes in β cell mass as well as related insusceptible cell action might prompt improved location and earlier prognosis [23], preceding the appearance of illness related indications. Ongoing advances in NP-based imaging innovations have given a novel course to measuring β cell mass in diabetic patients [24–27]. Attractive NPs (MNPs), for instance, have special actual properties that make them great contrast agents for magnetic resonance imaging (MRI) [28,29], and unique approaches have along these lines been created to upgrade their difference and biocompatibility [30,31].

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 588 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

Until this point, MNPs have just been actualized in a wide scope of ailments, counting malignant growth [32] and cardiovascular diseases [33], possibly making them an appealing diagnostics tool for diabetes. Various types of MNPs have now been molded as contrasts agents for β cell monitoring under MRI [34], and when these NPs are disguised by the host's cells, MRI can precisely and proficiently recognize the particles to encourage noninvasive representation of the β cells or pancreas [35]. Superparamagnetic iron oxide nanoparticles (SPIONs), which are profoundly biocompatible [36], have been formed with ferumoxran-10 or exendin-4 for resistant or β cell focusing on, individually [24–26]. Fermoxran-10-formed SPIONs have been created and tried in preclinical and clinical investigations to screen resistant cell penetration, MNPs are regularly utilized as contrast agents in MRI. These agents are regularly evolved from a paramagnetic metal center and covered with explicit materials to improve biocompatibility [30,31]. Improving Contrast in MNPs contrast agents and other attractive organic particles are adjusted and energized from the strong magnetic field and radiofrequency beat during an ordinary MRI. The time taken for the influenced particles to return to their lower energy levels or then again ground state is known as the unwinding time and is straightforwardly connected with contrast in MRI [30,31]. Unwinding properties are diverse for each metal. Metals, for example, gadolinium and iron have been generally utilized for contrast agents due to their moderately short unwinding times contrasted with different metals. MNP coatings, frequently utilized for biocompatibility upgrades, have likewise been utilized to improve unwinding times. β-Cyclodextrin-and hyaluronic acid- based coatings have illustrated improved unwinding times and differentiation [30]. i. Improving Contrast in MNPs MNPs have been created from biocompatible metals or covered with hydrophilic materials to improve biocompatibility [30,31]. Attractive metals, for example, iron oxides and manganese give low harmfulness levels. Also, slim polymer or sugar coatings are regularly used to forestall total development, changes in unique structure, natural debasement of the metal and negative natural responses. Studies examining MNP coatings have given significant understanding into material/sub-atomic properties that can give improved natural cooperation’s [30]. Polyethylene glycol, dextran, silica, what's more, gold coatings have been found to improve MNP biocompatibility [31]. Also, adversely charged materials were discovered to be less genotoxic than emphatically charged materials in vitro considers [30]. Target MNPs to the pancreas opens new roads for noninvasive imaging techniques that can follow the development of the disease just as treatment modalities. ii. Gas Sensor Ongoing examinations have taken a gander at the utilization of quantum dabs (QDs) for the advancement of option symptomatic methodologies for diabetes. QDs are profoundly touchy and savvy fluorescent semiconductor nanoparticles that range in size from 2 nm to 10 nm [37–39]. While their biocompatibility should be concentrated further [40], they can produce fluorescence in the close infrared reach, making them ideal tools for bioimaging applications because of diminished photon dissipating, diminished tissue assimilation, and decreased foundation autofluorescence, which prompts upgraded difference and tissue entrance [41,42]. In general, QDs are of interest in nanotechnology-based imaging/identification applications because of their wide scope of optoelectronic properties (e.g., thin band emanations), which vary altogether from the comparing mass material, and can be finely tuned dependent on size and shape [43]. Also, extra surface adjustments (e.g., zinc sulfide coatings) can empower upgraded properties, including biocompatibility and optical execution [44], for a wide reach of medical applications. Past diabetes, QDs are effectively being explored for different signs, for example, cancer theranostics [45], tissue designing [46], and microorganism discovery [47]. Liu et al. built up a QD-driven breath sensor that could analyze diabetes dependent on the acetone levels, which could increment from ~300–900 ppb for a sound individual to ~1.8 ppm for a diabetic patient [38]. Quickly, the QD metal oxide surface backings the adsorption of acetone particles, which brings down the voltage obstruction by moving electrons up the conduction band, coming about in an expanded progression of electrons. Such changes lead to modifications in the electrical resistance/ conductivity of the sensor, which encourages precise evaluation of the acetone levels present in a solitary breath under recreated diabetic versus sound conditions. Notwithstanding, while the sensor displayed high specifically to acetone contrasted with other possibly meddling analyte gases (e.g., ethanol, methanol, and so on), changes in relative moistness could have a huge hosing impact on the sensor's reaction [38]. Accordingly, future investigations focused on better controlling variances in relative mugginess will be key to empowering clinical implementation of this nanotechnology.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 589 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

V. NANOTECHNOLOGY APPLICATIONS IN MONITORING The most well-known way to deal with observing under diabetes includes the utilization of standard glucose meters, which expect patients to physically prick themselves continually for the duration of the day in request to follow changes in glucose levels. In spite of the fact that reliable, this strategy has a few impediments, counting poor patient compliance and questionable glucose estimations because of an assortment of components, including eating times, age, and so on [41]. Moreover, standard GM can't be directed during certain ordinary exercises, for example, resting or driving. In this way, the discontinuous idea of regular observing techniques infer that a patient can miss possibly risky glycemic vacillations between tests, putting patients in risk for serious difficulties [42,43]. In the course of the last three decades different endeavors have been made towards building up a hassle- free technique for GM. With the inevitable improvement of implantable biosensors, this thought got attainable and come about in persistent GM systems, which can give steady GM to as long as 10 days [44]. The original of these devices included amperometry sensors that are embedded subcutaneously. These sensors transmit a perceptible electric flow as an element of glucose focus [45]. While a step forward in continuous GM, these devices have a few disadvantages including: (I) absence of strength because of signal lag and sensor float; (ii) the requirement for week after week subcutaneous implantation also, alignment strategies; and (iii) high affectability to changes in different physiological boundaries, for example, pH and temperature [45,46]. Nanotechnology-based biosensors have abilities that can conceivably go around these constraints. Fluorescence-based nano sensors discharge a specific fluorescence signal after binding glucose. These sensors normally are related with glucose-binding atoms connected to QDs [47], carbon nanotubes (CNTs) [48], or nano-optodes [49], which convert the fluorescence energy related with the coupling occasion into a move in spectra or voltametric output. A few glucose-binding particles from regular (e.g., lectins) and engineered (e.g., phenylboronic acid) mixes have been utilized in these sensors [50]. Two of the most widely recognized proteins utilized for glucose detecting are concanavalin A (ConA), a lectin which has a high explicitness for glucose, what's more, glucose/galactose-binding protein, which modifies its compliance after binding glucose [51]. A significant favorable position of these devices contrasted with current systems is that they are most certainly not subject to battery life, in this way having the capacity to conceivably work constantly for delayed periods of time [51]. Liao et al. built up a fiberoptic glucose sensor dependent on ConA-functionalized QDs [47], with hair-like size and adaptability, which empowers discrete implantation in the skin for constant checking of glucose in the interstitial liquid. Glucose diffuses uninhibitedly into the sensor and ties to ConA, which causes a move in fluorescence that can be related with glucose focus. This sensor was effectively used to quickly and precisely recognize changes in glucose concentrations (0–500 mg/dl) in vitro in physiological arrangements for as long as 7 weeks. All the more as of late, amino-functionalized silicon QDs have likewise been exhibited as successful glucose detectors [52]. Practicing comparative standards as the CoAfunctionalized QDs, the amino-functionalized QDs demonstrated clear changes in power as a capacity of glucose focus in weakened human blood, just as high particularity for glucose over other important/comparable atoms, for example, sucrose, Na+, and so forth [52]. In any case, while this system shows high sensitivity, impediments in the recognition wrath of the sensor and the requirement for diluted blood tests could represent a test to clinical arrangement. Thusly, extra innovative headways, perhaps determined by miniature/nanofluidic stages, will be vital to empowering robotized test preprocessing (e.g., blood diluting) for appropriate checking of glucose variances. A. Insulin Monitoring Current insulin recognition strategies generally require lumbering and long ex vivo investigations dependent on ELISA or fluorescence spectroscopy [53,54]. Improving insulin detection, and furthermore, checking, be that as it may, may empower improved following of disease progression and additionally restorative delivery in both kind 1 and type 2 diabetes [55]. Past investigations have provided details regarding the advancement of improved advances for persistent insulin checking dependent on multiwalled CNTs, which are fundamentally level sheets of carbon particles stacked and folded into nanoscale tubes, and can be customized to show upgraded sensitivity to various biomolecules [56,57]. Insulin concentration as low as 1 μM can then be observed dependent on electrochemical location driven by insulin oxidation. Later examinations have likewise taken a gander at the utilization of graphene– polypyrrole nanocomposites for electrochemical insulin detection [58]. Improved insulin observing advancements won't simply be vital to encouraging aberrant following of disease progression (i.e., β cell mass/activity) or treatment delivery, yet may likewise discover applications in the screening of islet arrangements preceding transplantation for therapeutic purposes.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 590 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

VI. NANOTECHNOLOGY APPLICATIONS IN Customary for diabetes comprise of controlling manufactured insulin or insulin-based treatments [55]. While numerous types of insulin (e.g., injectable, oral, inhalable, and so forth) [59] and insulin analogs (e.g., insulin degludec U-100 and U200, insulin as part, and so forth) [60] are as of now accessible to treat type 1 diabetes, absolute recovery from this condition has not yet been accounted for. Most current medicines convey the risk of adverse effects (e.g., hypoglycemia) [61,62], and are regularly unequipped for keeping up euglycemia for prolonged period of time [55]. The main portrayal of 'brilliant' glucose-responsive insulin-based treatments, which possibly enact insulin discharge when glucose levels are high, was distributed in 1979 [63]. From that point forward, nanotechnology has played a huge function in expanding the effectiveness, usability, and security of insulin substitution treatments. Numerous durable nanotechnology-empowered restorative strategies have now been created to give more tight glycemic control for patients and limit the requirement for persistent and monotonous manual infusions [63]. Here, we talk about a couple of these novel insulin delivery strategies, including cell therapies, for the treatment of diabetes. 1. Insulin Patch Noninvasive delivery of insulin can possibly address resistance issues and keep away from complications related with poor glycemic control. The transdermal [64] and oral courses [65] have been perceived as patient friendly due to their noninvasive, moderately easy, and basic nature. Notwithstanding, restricted transport across epithelial barriers, poor bioavailability, and the harsh environmental of the gastrointestinal tract have restricted the achievement of past endeavors at insulin delivery by means of these routes [65]. Yu et al. built up a microneedle-based fix with glucose-responsive insulin-loaded nanoscale vesicles for insignificantly minimal invasive delivery of insulin [66]. The fix can be applied on the skin to controllably deliver insulin in light of changes in glucose levels, giving more tight glycemic control [67]. The glucose-responsive vesicles that contain insulin also, glucose oxidase is produced using hyaluronic acid formed with a hypoxia-sensitive component, 2-nitroinidazole. Under hyperglycemia, the oxygen is immediately consumed because of glucose oxidation, which is catalyzed by the glucose oxidase inside the vesicles. This at that point brings about limited hypoxia, setting off the breakdown of the vesicles and resulting arrival of insulin by means of the decrease of 2-nitroindazole [66]. Notwithstanding, while at the same time promising, future changes may even now be expected to forestall unreasonable biofouling under long haul usage, conceivably through nanocoating or nanofabrication advancements [68-69]. 2. Insulin Nanogel Glucose-responsive insulin-stacked hydrogels have arisen as an elective procedure for insulin delivery. Past gel-based systems demonstrated moderate reaction times, needed mechanical strength, furthermore, had issues with the insulin spilling out wildly [70]. Gu et al. presented a profoundly compelling insulin delivery systems dependent on an injectable gel produced using pH-sensitive dextranNPs stacked with glucose oxidase, which changes over glucose into gluconic acid. At the point when glucose levels are high, improved glucose dissemination into the gel brings about enormous amounts of gluconic acid. The subsequent acidic microenvironment at that point causes the dextran circles to continuously debase what's more, discharge insulin. Such systems have been utilized effectively to keep up glycemic control in diabetic mice for as long as 10 days after a system yet with various and more assorted sciences (e.g., polyethene glycol-based nanogels) [71], which instigated sufficient glucose responsiveness in diabetic mice for roughly 2 hours during a glucose tolerance test. Cell Therapies Albeit allogeneic islet transplantation has been appeared to prompt improved glycemic control in diabetic patients, donor tissue scarcity and the requirement for fundamental immunosuppression regimens have ruined boundless usage [72,73]. To address these issues, a few bunches have chipped away at creating improved procedures to separate actuated pluripotent stem cells, which can be autologous in nature, into insulin-creating cells (IPCs) [74,75]. Nanofiber-based polymeric (e.g., polylactic corrosive/polyvinyl liquor and polyether sulfone) systems, for instance, have been effectively used to improve the usefulness (e.g., glucose responsiveness) and separation efficiencies of IPCs [76,77]. Paradoxically, to limit the need for foundational immunosuppression related with donor derived cell therapies, a number of gatherings have taken a gander at different encapsulation ways to protect cellular grafts (e.g., allogeneic islets) from immune cell rejection [78]. Alginate is normally utilized in encapsulation strategies; nonetheless, recent studies have demonstrated that alginate can evoke adverse immune reactions by itself [78]. nanoscale surface adjustments with triazole stores could shorten such reactions, allowing implantation of alginate microspheres in the two rodents and nonhuman primates with altogether decreased unfamiliar body reaction

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 591 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

(e.g., fibrotic tissue testimony, related macrophages, and so on) for as long as 6months [79]. Extra investigations have focused on the advancement of slim (i.e., nanometer scale) exemplification layers with nanoscale pores for immune isolation purposes [80– 82]. The nanoscale thickness of the layer in mix with the little pore size is vital to empowering islet graft protecting from the immune system, while simultaneously encouraging legitimate oxygen, glucose, and insulin transport across the layer, which is an improvement over past alginate-based plans [81,82]. Wang et al., for instance, detailed successful islet transplantation without the utilization of immunosuppressive medications in a nonhuman primate model for 90 days, utilizing nanoscale coatings with a tightened nanopore course system that altogether decreased immune cell just as cytokine cooperation with the embedded islets, without affecting mass transport mechanisms key to graft survivability and functionality [83]. In spite of the fact that nanoscale innovations have obviously empowered numerous mechanical progressions in diabetes the board, likely obstacles for far reaching execution versus the proceeded utilization of customary techniques incorporate money saving advantage contemplations, just as the absence of utilization of customary techniques incorporate money saving advantage contemplations, just as the absence of lucidity in a few cases with respect to administrative rules as well as progress measurements/norms [84]. Extra preclinical and bigger scope clinical studies are as yet expected to completely assess the degree to which various kinds of nanotechnologies can prompt improved diabetic quiet results. For instance, while MNPs encourage insignificantly obtrusive intrapancreatic imaging, more proof is as yet expected to decide if inconspicuous changes in β cell mass as well as invulnerable cell action can be dependably used to analyze diabetes before. Likewise, in spite of the fact that blood glucose estimations have been the highest quality level regarding diagnostics and checking, further exploration is justified to assess not just the favorable circumstances, cost-adequacy, also, clinical pertinence of utilizing nanotechnology regarding expanded glucose affectability, exactness, and toughness, yet additionally as tool to assess other metabolic yields, other than glucose, that could be connected to disease beginning/progress. Conversely, as far as treatment, there is a great deal of preclinical proof recommending that nanotechnology can prompt improved systems for glucose detecting combined with insulin delivery. Nonetheless, more studies are needed to assess the drawn- out adequacy of such systems to securely control glycemic journeys in diabetic patients. Likewise, gene and cell treatments may empower novel answers for diabetes by adjusting resistant cell reactions as well as encouraging the improvement of insulin producing β-like cells through controlled cell separation or reconstructing [85]. What's more, while a wide assortment of nanoscale innovations has been produced for controlled quality conveyance furthermore, cell separation in vitro and in vivo [86,88], there is as yet restricted data with respect to the utilization of these methodologies inside the setting of diabetes treatments. As the information what's more, expansiveness of the nanotechnology field expands, the likely applications for diabetes the board become progressively more reasonable and promising.

VII. OUTSTANDING QUESTIONS Will a lack of precise regulatory guidelines on nanotechnology negatively impact clinical translation and wide spread implementation for some of the most promising nanotechnologies for diabetes management? Should we be putting more effort into establishing clear and widely accepted metrics/standards for risk/cost–benefit considerations to determine which nanotechnologies continue to move along the translational pipeline? Will some of these nanotechnologies end up being cost-prohibitive for certain pockets of the diabetic/prediabetic patient population, and/or for nations with limited healthcare budgets? What should be the role of governments, industry, academia, and non-profit-making organizations in terms of making sure these nanotechnologies are more affordable? Should the field be equally vested in developing nanotechnologies that are solely aimed at helping us understand better the fundamentals of diabetes? Or should the field focus instead on developing translational nanotechnologies with more commercial potential?

CONFLICT OF INTEREST All authors declare no conflicts of interest.

AUTHORS CONTRIBUTION Authors have equally participated and shared every item of the work.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 592 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

REFERENCES [1]- American Diabetes Association. (2019). 2. Classification and diagnosis of diabetes: standards of medical care in diabetes— 2019. Diabetes care, 42(Supplement 1), S13-S28. [2]- Shaw, J. E., Sicree, R. A., & Zimmet, P. Z. (2010). Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes research and clinical practice, 87(1), 4-14. [3]- Karamanou, M., Protogerou, A., Tsoucalas, G., Androutsos, G., & Poulakou-Rebelakou, E. (2016). Milestones in the history of diabetes mellitus: The main contributors. World journal of diabetes, 7(1), 1. [4]- Dabelea, D. (2009). The accelerating epidemic of childhood diabetes. Lancet (London, England), 373(9680), 1999-2000. [5]- Lieberman, S. M., & DiLorenzo, T. P. (2003). A comprehensive guide to antibody and T‐cell responses in type 1 diabetes. Tissue antigens, 62(5), 359-377. [6]- Donath, M. Y., & Shoelson, S. E. (2011). Type 2 diabetes as an inflammatory disease. Nature Reviews , 11(2), 98-107. [7]- Care, D. (2019). 6. Glycemic targets: standards of medical care in diabetes—2019. Diabetes Care, 42(Supplement 1), S61-70. [8]- Zitkus, B. S. (2014). Update on the American Diabetes Association standards of medical care. The Nurse Practitioner, 39(8), 22-32. [9]- Schulman, R., Moshier, E., Rho, L., Casey, M., Godbold, J., & Mechanick, J. (2014). Association of glycemic control parameters with clinical outcomes in chronic critical illness. Endocrine Practice, 20(9), 884-893. [10]- Streisand, R., & Monaghan, M. (2014). Young children with type 1 diabetes: challenges, research, and future directions. Current diabetes reports, 14(9), 520. [11]- Pickup, J. C. (2012). Insulin-pump therapy for type 1 diabetes mellitus. New England Journal of , 366(17), 1616- 1624. [12]- Brown, S. A., Kovatchev, B. P., Raghinaru, D., Lum, J. W., Buckingham, B. A., Kudva, Y. C., ... & Dassau, E. (2019). Six- month randomized, multicenter trial of closed-loop control in type 1 diabetes. New England Journal of Medicine, 381(18), 1707- 1717. [13]- Davis, M. E., Zuckerman, J. E., Choi, C. H. J., Seligson, D., Tolcher, A., Alabi, C. A., ... & Ribas, A. (2010). Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature, 464(7291), 1067-1070. [14]- Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. (2007). Nanocarriers as an emerging platform for cancer therapy. Nature nanotechnology, 2(12), 751-760. [15]- Kim, B. Y., Rutka, J. T., & Chan, W. C. (2010). . New England Journal of Medicine, 363(25), 2434-2443. [16]- LaVan, D. A., Lynn, D. M., & Langer, R. (2002). Moving smaller in drug discovery and delivery. Nature Reviews Drug Discovery, 1(1), 77-84. [17]- Whitesides, G. M. (2003). The'right'size in nanobiotechnology. Nature biotechnology, 21(10), 1161-1165. [18]- Okur, M. E., Karantas, I. D., & Siafaka, P. I. (2017). Diabetes Mellitus: a review on pathophysiology, current status of oral pathophysiology, current status of oral medications and future perspectives. ACTA Pharmaceutica Sciencia, 55(1). [19]- Mayorov, A. Y. E. (2011). Insulin resistance in pathogenesis of type 2 diabetes mellitus. Diabetes mellitus, 14(1), 35-45. [20]- Ojha, A., Ojha, U., Mohammed, R., Chandrashekar, A., & Ojha, H. (2019). Current perspective on the role of insulin and glucagon in the pathogenesis and treatment of type 2 diabetes mellitus. Clinical Pharmacology: Advances and Applications, 11, 57. [21]- Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. (2007). Nanocarriers as an emerging platform for cancer therapy. Nature nanotechnology, 2(12), 751-760. [22]- Kim, B. Y., Rutka, J. T., & Chan, W. C. (2010). Nanomedicine. New England Journal of Medicine, 363(25), 2434-2443. [23]- Hieronymus, L., & Griffin, S. (2015). Role of amylin in type 1 and type 2 diabetes. The Diabetes Educator, 41(1_suppl), 47S- 56S. [24]- Stringer, D. M., Zahradka, P., & Taylor, C. G. (2015). Glucose transporters: cellular links to hyperglycemia in insulin resistance and diabetes. Nutrition Reviews, 73(3), 140-154. [25]- Rang, H. P., Dale, M. M., Ritter, J. M., & Moore, P. K. (2003). Pharmacology, 5th edn. Churchill Livingstone. Edinburgh, Scotland. [26]- Streisand, R., & Monaghan, M. (2014). Young children with type 1 diabetes: challenges, research, and future directions. Current diabetes reports, 14(9), 520.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 593 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

[27]- Olokoba, A. B., Obateru, O. A., & Olokoba, L. B. (2012). Type 2 diabetes mellitus: a review of current trends. Oman medical journal, 27(4), 269. [28]- Khazrai, Y. M., Defeudis, G., & Pozzilli, P. (2014). Effect of diet on type 2 diabetes mellitus: a review. Diabetes/metabolism research and reviews, 30(S1), 24-33. [29]- Yokono, M., Takasu, T., Hayashizaki, Y., Mitsuoka, K., Kihara, R., Muramatsu, Y., ... & Tomiyama, H. (2014). SGLT2 selective inhibitor ipragliflozin reduces body fat mass by increasing fatty acid oxidation in high-fat diet-induced obese rats. European journal of pharmacology, 727, 66-74. [30]- Boles, A., Kandimalla, R., & Reddy, P. H. (2017). Dynamics of diabetes and obesity: epidemiological perspective. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 1863(5), 1026-1036. [31]- Gambineri, A., Patton, L., Altieri, P., Pagotto, U., Pizzi, C., Manzoli, L., & Pasquali, R. (2012). Polycystic ovary syndrome is a risk factor for type 2 diabetes: results from a long-term prospective study. Diabetes, 61(9), 2369-2374. [32]- Papatheodorou, K., Banach, M., Bekiari, E., Rizzo, M., & Edmonds, M. (2018). Complications of diabetes 2017. [33]- K. Akhalya, S. Sreelatha, Rajeshwari, K. Shruthi, A review article- gestational diabetes mellitus, Endocrinol. Metab. Int. J. 7 (2019) 26–39 [34]- Dirar, A. M., & Doupis, J. (2017). Gestational diabetes from A to Z. World journal of diabetes, 8(12), 489. [35]- Diabetes Control and Complications Trial Research Group. (1995). Progression of retinopathy with intensive versus conventional treatment in the Diabetes Control and Complications Trial. , 102(4), 647-661. [36]- Evans, M. (1998). The UK prospective diabetes study. The Lancet, 352(9144), 1932-1933. [37]- Andralojc, K., Srinivas, M., Brom, M., Joosten, L., De Vries, I. J. M., Eizirik, D. L., ... & Gotthardt, M. (2012). Obstacles on the way to the clinical visualisation of beta cells: looking for the Aeneas of molecular imaging to navigate between Scylla and Charybdis. Diabetologia, 55(5), 1247-1257. [38]- Veiseh, O., Gunn, J. W., & Zhang, M. (2010). Design and fabrication of magnetic nanoparticles for targeted drug delivery and imaging. Advanced drug delivery reviews, 62(3), 284-304. [39]- Dallo, F. J., & Weller, S. C. (2003). Effectiveness of diabetes mellitus screening recommendations. Proceedings of the National Academy of Sciences, 100(18), 10574-10579. [40]- Regnell, S. E., & Lernmark, Å. (2017). Early prediction of autoimmune (type 1) diabetes. Diabetologia, 60(8), 1370-1381. [41]- American Diabetes Association. (2019). 2. Classification and diagnosis of diabetes: standards of medical care in diabetes— 2019. Diabetes care, 42(Supplement 1), S13-S28. [42]- Care, D. (2019). 6. Glycemic targets: standards of medical care in diabetes—2019. Diabetes Care, 42(Supplement 1), S61-70. [43]- Schulman, R., Moshier, E., Rho, L., Casey, M., Godbold, J., & Mechanick, J. (2014). Association of glycemic control parameters with clinical outcomes in chronic critical illness. Endocrine Practice, 20(9), 884-893. [44]- Edelman, S. V., Argento, N. B., Pettus, J., & Hirsch, I. B. (2018). Clinical implications of real-time and intermittently scanned continuous glucose monitoring. Diabetes Care, 41(11), 2265-2274. [45]- Hovorka, R., Nodale, M., Haidar, A., & Wilinska, M. E. (2013). Assessing performance of closed-loop insulin delivery systems by continuous glucose monitoring: drawbacks and way forward. Diabetes technology & therapeutics, 15(1), 4-12. [46]- Schmid, C., Haug, C., Heinemann, L., & Freckmann, G. (2013). System accuracy of blood glucose monitoring systems: impact of use by patients and ambient conditions. Diabetes technology & therapeutics, 15(10), 889-896. [47]- Liao, K. C., Hogen-Esch, T., Richmond, F. J., Marcu, L., Clifton, W., & Loeb, G. E. (2008). Percutaneous fiber-optic sensor for chronic glucose monitoring in vivo. Biosensors and Bioelectronics, 23(10), 1458-1465. [48]- Barone, P. W., & Strano, M. S. (2009). Single walled carbon nanotubes as reporters for the optical detection of glucose. [49]- Balaconis, M. K., Billingsley, K., Dubach, J. M., Cash, K. J., & Clark, H. A. (2011). The design and development of fluorescent nano-optodes for in vivo glucose monitoring. Journal of diabetes science and technology, 5(1), 68-75. [50]- Schultz, J. S., Mansouri, S., & Goldstein, I. J. (1982). Affinity sensor: a new technique for developing implantable sensors for glucose and other metabolites. Diabetes Care, 5(3), 245-253. [51]- Klonoff, D. C. (2012). Overview of fluorescence glucose sensing: a technology with a bright future. [52]- Du, L., Li, Z., Yao, J., Wen, G., Dong, C., & Li, H. W. (2019). Enzyme free glucose sensing by amino-functionalized silicon quantum dot. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 216, 303-309. [53]- Pørksen, N., Nyholm, B., Veldhuis, J. D., Butler, P. C., & Schmitz, O. (1997). In humans at least 75% of insulin secretion arises from punctuated insulin secretory bursts. American Journal of Physiology- and Metabolism, 273(5), E908- E914.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 594 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

[54]- Qian, W. J., Peters, J. L., Dahlgren, G. M., Gee, K. R., & Kennedy, R. T. (2004). Simultaneous monitoring of Zn2+ secretion and intracellular Ca2+ from islets and islet cells by fluorescence microscopy. Biotechniques, 37(6), 922-933. [55]- American Diabetes Association. (2019). 7. Diabetes technology: standards of medical care in diabetes—2019. Diabetes Care, 42(Supplement 1), S71-S80. [56]- Snider, R. M., Ciobanu, M., Rue, A. E., & Cliffel, D. E. (2008). A multiwalled carbon nanotube/dihydropyran composite film electrode for insulin detection in a microphysiometer chamber. Analytica chimica acta, 609(1), 44-52. [57]- Meetoo, D., & Lappin, M. (2009). Nanotechnology and the future of diabetes management. Journal of Diabetes Nursing, 13(8), 288-297. [58]- Ebrahimiasl, S., Fathi, E., & Ahmad, M. (2018). Electrochemical detection of insulin in blood serum using Ppy/GF nanocomposite modified pencil graphite electrode. Nanomedicine Research Journal, 3(4), 219-228. [59]- Amulya, C. et al. (2019) A review on alternative routes for insulin administration. World J. Pharm. Res. 8, 1471–1479 [60]- Rodbard, H. W., & Rodbard, D. (2020). Biosynthetic human insulin and insulin analogs. American Journal of Therapeutics, 27(1), e42-e51. [61]- McCrimmon, R. J., & Frier, B. M. (1994). Hypoglycaemia, the most feared complication of insulin therapy. Diabete et metabolisme, 20(6), 503-512. [62]- Johnson‐Rabbett, B., & Seaquist, E. R. (2019). Hypoglycemia in diabetes: The dark side of diabetes treatment. A patient‐ centered review. Journal of diabetes, 11(9), 711-718. [63]- Brownlee, M., & Cerami, A. (1979). A glucose-controlled insulin-delivery system: semisynthetic insulin bound to lectin. Science, 206(4423), 1190-1191. [64]- Pegoraro, C., MacNeil, S., & Battaglia, G. (2012). Transdermal drug delivery: from micro to nano. Nanoscale, 4(6), 1881- 1894. [65]- Ensign, L. M., Cone, R., & Hanes, J. (2012). Oral drug delivery with polymeric nanoparticles: the gastrointestinal mucus barriers. Advanced drug delivery reviews, 64(6), 557-570. [66]- Yu, J., Zhang, Y., Ye, Y., DiSanto, R., Sun, W., Ranson, D., ... & Gu, Z. (2015). Microneedle-array patches loaded with hypoxia-sensitive vesicles provide fast glucose-responsive insulin delivery. Proceedings of the National Academy of Sciences, 112(27), 8260-8265. [67]- Schoellhammer, C. M., Blankschtein, D., & Langer, R. (2014). Skin permeabilization for transdermal drug delivery: recent advances and future prospects. Expert opinion on drug delivery, 11(3), 393-407. [68]- Hang, T., Xiao, S., Yang, C., Li, X., Guo, C., He, G., ... & Deng, S. (2019). Hierarchical graphene/nanorods-based H2O2 electrochemical sensor with self-cleaning and anti-biofouling properties. Sensors and Actuators B: Chemical, 289, 15-23. [69]- Archana, S., & Sundaramoorthy, B. (2019). Review on biofouling prevention using nanotechnology. J. Entomol. Zool. Stud., 7, 640-648. [70]- Gu, Z., Aimetti, A. A., Wang, Q., Dang, T. T., Zhang, Y., Veiseh, O., ... & Anderson, D. G. (2013). Injectable nano-network for glucose-mediated insulin delivery. ACS nano, 7(5), 4194-4201. [71]- Li, C., Liu, X., Liu, Y., Huang, F., Wu, G., Liu, Y., ... & An, Y. (2019). Glucose and H 2 O 2 dual-sensitive nanogels for enhanced glucose-responsive insulin delivery. Nanoscale, 11(18), 9163-9175. [72]- Habener, J. F. (2004). A perspective on pancreatic stem/progenitor cells. Pediatric Diabetes, 5, 29-37. [73]- Shapiro, A. M. J., Lakey, J. R., Paty, B. W., Senior, P. A., Bigam, D. L., & Ryan, E. A. (2005). Strategic opportunities in clinical islet transplantation. Transplantation, 79(10), 1304-1307. [74]- Enderami, S. E., Mortazavi, Y., Soleimani, M., Nadri, S., Biglari, A., & Mansour, R. N. (2017). Generation of insulin‐ producing cells from human‐induced pluripotent stem cells using a stepwise differentiation protocol optimized with platelet‐rich plasma. Journal of cellular physiology, 232(10), 2878-2886. [75]- Abazari, M. F., Soleimanifar, F., Aleagha, M. N., Torabinejad, S., Nasiri, N., Khamisipour, G., ... & Hashemi, J. (2018). PCL/PVA nanofibrous scaffold improve insulin-producing cells generation from human induced pluripotent stem cells. Gene, 671, 50-57. [76]- Mansour, R. N., Soleimanifar, F., Abazari, M. F., Torabinejad, S., Ardeshirylajimi, A., Ghoraeian, P., ... & Enderami, S. E. (2018). Collagen coated electrospun polyethersulfon nanofibers improved insulin producing cells differentiation potential of human induced pluripotent stem cells. Artificial Cells, Nanomedicine, and Biotechnology, 46(sup3), S734-S739.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 595 Nanotechnology in Diagnosis and Treatment of Diabetes Mellitus: Review

[77]- Enderami, S. E., Kehtari, M., Abazari, M. F., Ghoraeian, P., Nouri Aleagha, M., Soleimanifar, F., ... & Askari, H. (2018). Generation of insulin-producing cells from human induced pluripotent stem cells on PLLA/PVA nanofiber scaffold. Artificial cells, nanomedicine, and biotechnology, 46(sup1), 1062-1069. [78]- de Vos, P., Faas, M. M., Strand, B., & Calafiore, R. (2006). Alginate-based microcapsules for immunoisolation of pancreatic islets. Biomaterials, 27(32), 5603-5617. [79]- Vegas, A. J., Veiseh, O., Doloff, J. C., Ma, M., Tam, H. H., Bratlie, K., ... & Fenton, P. (2016). Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates. Nature biotechnology, 34(3), 345-352. [80]- Lou, S., Zhang, X., Zhang, J., Deng, J., Kong, D., & Li, C. (2017). Pancreatic islet surface bioengineering with a heparin- incorporated starPEG nanofilm. Materials Science and Engineering: C, 78, 24-31. [81]- Song, S., Faleo, G., Yeung, R., Kant, R., Posselt, A. M., Desai, T. A., ... & Roy, S. (2016). Silicon nanopore membrane (SNM) for islet encapsulation and immunoisolation under convective transport. Scientific reports, 6(1), 1-9. [82]- Yang, J., Jiang, S., Guan, Y., Deng, J., Lou, S., Feng, D., ... & Li, C. (2019). Pancreatic islet surface engineering with a starPEG-chondroitin sulfate nanocoating. Biomaterials science, 7(6), 2308-2316. [83]- Wang, T. (2018). Successful diabetes management without immunosuppressivedrugs in NHP model has been demonstrated. Encapsulation system with taperednanopore conduits achieved normal glycaemia with regulated insulin release. Artificial cells, nanomedicine, and biotechnology, 46(sup3), S1162-S1168. [84]- Havel, H., Finch, G., Strode, P., Wolfgang, M., Zale, S., Bobe, I., ... & Liu, M. (2016). : from bench to bedside and beyond. The AAPS journal, 18(6), 1373-1378. [85]- Xiao, X., Guo, P., Shiota, C., Zhang, T., Coudriet, G. M., Fischbach, S., ... & Piganelli, J. D. (2018). Endogenous reprogramming of alpha cells into beta cells, induced by viral gene therapy, reverses autoimmune diabetes. Cell stem cell, 22(1), 78-90. [86]- Whitehead, K. A., Langer, R., & Anderson, D. G. (2009). Knocking down barriers: advances in siRNA delivery. Nature reviews Drug discovery, 8(2), 129-138. [87]- Gallego-Perez, D., Pal, D., Ghatak, S., Malkoc, V., Higuita-Castro, N., Gnyawali, S., ... & Singh, K. (2017). Topical tissue nano-transfection mediates non-viral stroma reprogramming and rescue. Nature nanotechnology, 12(10), 974-979. [88]- Gallego-Perez, D., Otero, J. J., Czeisler, C., Ma, J., Ortiz, C., Gygli, P., ... & Ghatak, S. (2016). Deterministic transfection drives efficient nonviral reprogramming and uncovers reprogramming barriers. Nanomedicine: Nanotechnology, Biology and Medicine, 12(2), 399-409.

Vol. 24 No. 1 December 2020 ISSN: 2509-0119 596