Next Article in Journal
Presence of p25alpha-Domain in Seed Plants (Spermatophyta): Microbial/Animal Contaminations and/or Orthologs
Previous Article in Journal
Pathogenesis of Alcoholic Fatty Liver a Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Technologies for Managing Type 1 Diabetes Mellitus and Their Impact on Quality of Life—A Narrative Review

by
Viviana Elian
1,2,
Violeta Popovici
3,*,
Emma-Adriana Ozon
4,*,
Adina Magdalena Musuc
5,
Ancuța Cătălina Fița
4,
Emilia Rusu
1,6,
Gabriela Radulian
1,2 and
Dumitru Lupuliasa
4
1
Department of Diabetes, Nutrition and Metabolic Diseases, “Carol Davila” University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050471 Bucharest, Romania
2
Department of Diabetes, Nutrition and Metabolic Diseases, “Prof. Dr. N. C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 030167 Bucharest, Romania
3
Department of Microbiology and Immunology, Faculty of Dental Medicine, Ovidius University of Constanta, 7 Ilarie Voronca Street, 900684 Constanta, Romania
4
Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania
5
Romanian Academy, “Ilie Murgulescu” Institute of Physical Chemistry, 202 Spl. Independentei, 060021 Bucharest, Romania
6
Department of Diabetes, N. Malaxa Clinical Hospital, 12 Vergului Street, 022441 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
Life 2023, 13(8), 1663; https://doi.org/10.3390/life13081663
Submission received: 31 May 2023 / Revised: 27 July 2023 / Accepted: 28 July 2023 / Published: 30 July 2023
(This article belongs to the Section Physiology and Pathology)

Abstract

:
Type 1 diabetes mellitus is a chronic autoimmune disease that affects millions of people and generates high healthcare costs due to frequent complications when inappropriately managed. Our paper aimed to review the latest technologies used in T1DM management for better glycemic control and their impact on daily life for people with diabetes. Continuous glucose monitoring systems provide a better understanding of daily glycemic variations for children and adults and can be easily used. These systems diminish diabetes distress and improve diabetes control by decreasing hypoglycemia. Continuous subcutaneous insulin infusions have proven their benefits in selected patients. There is a tendency to use more complex systems, such as hybrid closed-loop systems that can modulate insulin infusion based on glycemic readings and artificial intelligence-based algorithms. It can help people manage the burdens associated with T1DM management, such as fear of hypoglycemia, exercising, and long-term complications. The future is promising and aims to develop more complex ways of automated control of glycemic levels to diminish the distress of individuals living with diabetes.

1. Introduction

According to the World Health Organization, diabetes is a chronic, metabolic disease characterized by elevated levels of blood glucose, which leads over time to severe damage to the heart, blood vessels, eyes, kidneys, and nerves [1]. With more than 420 million people living with this disease worldwide, diabetes is at the top of global death causes [2].
Type 1 diabetes mellitus (T1DM) occurs when insulin production is severely impaired by the pancreatic beta cells’ autoimmune destruction [3]. Patients are usually diagnosed when 80–90% of the beta cells are destroyed. Recently, Roep et al. [4] proposed an alternative point of view in which abnormal β-cells are the main contributors to T1DM pathogenesis. Based on previously reported results of immunotherapy in T1DM [5,6,7], they showed that β-cells with limited self-defense are vulnerable to biosynthetic stress [4].
Novel strategies for preventing and managing T1DM are investigated [8,9]. One research direction in this field is antigen vaccination (with oral insulin or peptides) and stem-cell-based replacement therapies [10] to prevent and reverse diabetes autoimmunity. FDA approved in November 2022 the first drug—Tzield (teplizumab-mzwv) injection—that can delay the onset of T1DM [11,12,13], resulting in years of disease remission [14]. Data also suggest that verapamil and glucagon-like peptide-1 receptor agonists could act as beta cell protective agents (in stress conditions).
Even though there are new therapeutical approaches and promising results, insulin remains the only long-term effective treatment for T1DM. Commonly, insulin is administered as multiple daily subcutaneous injections [15,16] with doses adjusted upon the determined glycemic level (fasting, preprandial, or at the time of the injection) and the meal carb count. It is also possible to deliver insulin by continuous subcutaneous insulin infusion [17] or insulin pump.
There are concerns related to access to insulin for people living with T1DM [18] in countries with limited government expenditures for health because they must pay out-of-pocket for all or part of their diabetes care, including insulin and syringes, blood glucose meters, delivery devices, and necessary health education [19,20]. Some patients reported on specific questionnaires cost-related underuse of insulin or cost-related change in insulin type, a scenario caused by low personal income. More than a third of them did not discuss this problem with their physicians. Data suggested that these individuals have poor glycemic control and are exposed to complications [21].
T1DM remains a chronic disease associated with high morbidity [22], mortality, and a diminished work life [23,24,25]. When diabetes coexists with other diseases [26,27,28,29,30,31], the management of a patient’s condition is even more difficult.
The previously mentioned aspects have complex implications on psychology and quality of life (QoL) [32]. The risk of developing psychiatric comorbidities (depressive disorders) increases with the earlier onset of T1DM [33,34]. These conditions are defined as diabetes distress (DD) and are linked to poor compliance, low glycemic control, and long-term complications (Figure 1) [33,35].
Generally, younger people [36,37], women [38], and people with poor glycemic control [39] have an increased risk for high levels of DD. Older people have a lower DD incidence [40].
T1DM management includes complex and precise self-care measures during a person’s entire life. This continuous process could overwhelm them, making them angry, anxious, frustrated, and/or discouraged [41].
Due to the previously described conditions, the new technologies for DM management must provide clinical advantages and high usability without compromising safety [42]. Adherence and compliance to the prescribed therapy are also essential, thus improving their QoL [43]. Moreover, the accessibility of the new system through healthcare coverage [44] is critical; they must be able to afford the new devices and accessories and their maintenance [45,46].
In this context, our work aims to analyze the most recent technologies regarding continuous glucose monitors (CGMs) and continuous subcutaneous insulin infusion (CSII) systems, their psychosocial impact [47], and their capacity to improve T1DM patients’ life quality [48] in a narrative review.
We performed a literature review searching original published articles on type 1 diabetes in humans. We used a data filter on insulin therapy, insulin pumps, and advanced technologies in medical devices for insulin infusion. The selection targeted the publications in scholarly peer-reviewed journals in the past 10 years (2013–2022) written in English (no country restriction). The initial selection was from Google Scholar search. Due to the high document variety, we accessed three other primary databases: PubMed®/MEDLINE, ScienceDirect, and Web of Science®. The following keywords were used: type 1 diabetes mellitus technology (3046 journal articles), continuous glucose monitoring system (4273), continuous insulin infusion (7150), automated insulin delivery (533), patch insulin pump (153), closed-loop insulin pump (874), hybrid closed-loop insulin pump (218), artificial pancreas (3389), bionic pancreas (76). We realized our search only on articles edited in the English language and performed the final search on 15 April 2023.

2. Continuous Glucose Monitoring Systems

Monitoring glycemia rigorously is essential for T1DM management. Adjusting insulin doses requires six or more measurements of blood glucose level daily. The basic technology in determining blood glucose, still widely used around the world, is represented by the blood glucose meters (BGM) [49]. These devices are not very expensive and easy to use by individuals to perform different changes in the day-to-day management of their diabetes [50].
Daily glucose level monitoring was significantly improved through advanced technologies that offer continuous glucose monitoring (CGM) systems [51,52,53]. Two types of CGMs are available: professional CGM (data can only be downloaded and seen by the physician) and personal CGM (data seen in real time by the person wearing the device) comprising intermittently scanned CGMs (IS-CGM) [54] and real-time ones (RT-CGM) [55]. CGMs are based on minimally invasive [56] and non-invasive [57] techniques.
An IS-CGM consists of a sensor that measures glucose levels in the interstitial fluid every minute and stores data automatically every 15 min without transmitting them [58,59]. Data can be seen when the person scans the sensor with a reader or a smartphone.
RT-CGMs automatically measure glucose levels day and night and transmit them every 1/5 min to a receiver (reader, smartphone, insulin pump) [60,61]. They have alarm systems for out-of-range glucose levels and inform patients/caregivers about immediate/long-term events (hyperglycemia or hypoglycemia) [62]. Knowing glucose levels in real-time can help make more informed daily decisions about food balance, physical activity, and insulin doses [63].

2.1. Description

A CGM system comprises a sensor, transmitter, receiver, application software, and insertion tool. CGMs evaluate glucose levels in the interstitial fluid (ISF) from the subcutaneous adipose tissue [64]. The measured glucose concentrations in ISF are processed through various algorithms [65] to predict blood glucose levels (BGL).
A CGMS performance is evaluated by the mean absolute relative difference (MARD) [66]. The MARD calculation involves temporally matched glucose data from CGM systems compared to BG measurements of all subjects included in a clinical study. A high-performance CGM implies MARD value < 10%. Therefore, the BG evaluation 2–4 times daily with a glucometer ensures an optimal calibration of CGM systems, even for those without mandatory calibration, thus avoiding significant differences between CGM data and BG values.
The clinical utility of CGMs consists of therapeutic decisions [67] based on real-time glucose evaluation:
Insulin bolus size calculation both in MDI and CSII regimens [68];
Carbs intake for preventing/treating hypoglycemia;
Insulin corrections for increased/increasing glycemia;
Regulation of basal rates in sensor-augmented insulin pumps [69] or automated insulin delivery (AID) systems [70];
Micro-bolusing in AID.
Different companies commercialize minimally invasive continuous glucose monitoring devices (MID): GlySens Incorporated (San Diego, CA, USA), Senseonics Holdings, Inc. (Germantown, MD, USA), Abbott Laboratories (Chicago, IL, USA), Medtronic plc (Minneapolis, MN, USA), F. Hoffmann-La Roche Ltd. (Basel, Switzerland), Dexcom, Inc. (San Diego, CA, USA), A. Menarini Diagnostics (Florence, Italy), LifeScan IP Holdings LLC (Malvern, PA, USA), Echo Therapeutics, Inc. (Philadelphia, PA, USA), Johnson & Johnson (New Brunswick, NJ, USA), Terumo Corporation (Shibuya City, Japan), and B. Braun Melsungen AG (Melsungen, Germany). In Table 1, we have exemplified the latest and most frequently used CGMs in the EU and the USA.
In Table 1, the most known FDA-approved CGMs are comparatively analyzed.
CGMS should not be exposed to magnetic resonance imaging (MRI) equipment, diathermy devices, or other devices that generate strong magnetic fields (for example, X-ray, CT scan, or other types of radiation). Exposure to a strong magnetic field has not been evaluated and can cause the device to malfunction, result in serious injury, or be unsafe.
Registered data show that the most complex and expensive CGM is Eversense E3 CGM. It also has the most prolonged period of sensor-wearing but is suitable only for adults. The other CGMS have RT under 15 days but can also be used for children.
The measured glucose levels are strongly influenced by numerous factors related to the insertion place [71]. The abdominal region [71] is the most common location, followed by the upper arm. Measurements from the right abdominal site tend to be more diminished than those from the left. Studies on CGM sensors involving sleep [72] and belt compression [73] reported a potential influence of diminished blood flow on the sensors’ measurements. Similarly, Mensh et al. [64] demonstrated that the susceptibility of CGMs to abnormal nocturnal glucose readings was related to sleeping positions. On the abdominal surface, various physiologic conditions [74,75] could be related to different CGM readings in the left versus right part. In various BMI values, the subcutaneous adipose tissue size could influence CGM measurement accuracy [76].
The sensors’ biocompatibility could determine the discrepancy between abdominal sites [77]. Sensor insertion traumatizes the afferent zone, inducing inflammatory reactions [78], with increased blood flow and glucose [79]. Local proteolytic enzymes and reactive oxygen species [80] could negatively affect the sensors. During wound healing, capillary neoangiogenesis can supply additional glucose at the insertion site [25].
Recently, a non-invasive CGM manufactured by Afon Technology Ltd. (Caldicot, Monmouthshire, UK) is in the stage of clinical studies. It could be available soon as an alternative technology to traditional blood glucose monitoring tools. This CGM system utilizes high-frequency microwaves, a technology not yet used in this application [81].
The most important features regarding used materials, techniques, and methods are displayed in Figure 2.

2.2. Benefits

Compared with a standard blood glucometer, every day using a CGM system can help to:
Maintain a constant glycemic level daily;
Diminish time spent in hypoglycemia and severe hyperglycemia;
Reduce the number of finger pricks;
Decrease BG variability and HbA1c levels.

2.3. Limits

CGM systems are more expensive than standard glucometers;
The finger prick glucose test is needed twice daily for some CGMs to calibrate;
Sporadic, unpredictable errors [66];
Invasiveness;
Short lifespan;
Biocompatibility.

2.4. Potential Adverse Effects Related to the Insertion, Removal, and Wear of the Sensor

Allergies to adhesives;
Bleeding and bruising;
Infection, pain, or discomfort;
Sensor destruction during extraction;
Skin inflammation, scarring, thinning, discoloration, or redness.
Other potential unwanted effects are associated with decisions made in the case of inaccurate device measurements:
Excessive insulin administration could increase the risk of hypoglycemia;
Inappropriate administration of carbohydrates increases the risk of hyperglycemia and acute diabetic ketoacidosis;
Inaccurate calculation of the glucose change rate could increase the incidence of hypo or hyperglycemia.

3. Continuous Subcutaneous Insulin Delivery Systems (CSII)

An insulin pump is an electronic device that releases rapid insulin according to the body’s daily needs [82,83].
Insulin pumps deliver insulin in two primary ways:
A continuous infusion of small amounts of rapid insulin throughout the day and night (basal rate);
A one-time dose of rapid-acting insulin for meals or high blood glucose correction (bolus).
The ideal individuals for insulin pump use are:
  • People with T1DM or insulin-dependent T2DM;
  • People with multiple-day injections of insulin;
  • People who can assess appropriate blood glucose control;
  • Capable of performing insulin pump therapy initiation and maintenance;
  • Able to maintain frequent contact with the healthcare team;
  • Able to consider insulin pumps as a tool to improve diabetes care;
  • Capable of accurately calculating carbohydrates and insulin bolus;
  • Individuals with critical clinical conditions who have serious difficulties controlling glycemic targets, despite intensive treatment and monitoring;
  • With substantially decompensated diabetes (frequent severe hypoglycemia and/or hyperglycemia);
  • Other associated conditions: extreme insulin sensitivity, gastroparesis, pregnancy, variable schedules or work shifts, significant “dawn phenomenon”, high insulin dose therapy, or severe insulin resistance.

3.1. Conventional Insulin Pumps

An insulin pump is a small digital device that ensures a continuous infusion of rapid-acting insulin (CSII). The infusion set is inserted into the subcutaneous tissue and fixed on the skin with an adhesive. In most insulin pumps, the infusion set connects to the pump by plastic tubing. Insulin infuses from the pump through the tubing to the infusion set cannula and into the subcutaneous tissue. The most common devices are displayed in Table 2.
Table 3 shows the benefits and limitations of CSII pumps [84].

3.2. Insulin Patch Pumps (PPs)

Some insulin pumps are directly attached to the skin (patch insulin pumps). A hand device controls insulin delivery in a PP; however, some devices also allow at least some functionality via the PP. The simple forms of PPs intended for insulin therapy aim to be small and disposable, and easy to handle and carry. There are three categories of PPs: PPs with reduced features, fully equipped PPs, and PPs suitable for automatic insulin delivery (AID) systems.
The reduced-features PP delivers only basal insulin. A fully equipped PP delivers a variable amount of basal insulin over 24 h and has a bolus button that permits prandial insulin to be given in two-unit increments daily (Figure 3 and Table S1 from Supplementary Materials).
Full-featured electromechanical patch pumps generally have an electromechanical structure with an electronic controller. These are all full-featured pumps with different basal rates, individually controllable bolus amounts, and additional means of bolus delivery.
PPs are small, easy to use, and discreet to wear. Moreover, they can interact with the CGM and AID systems’ algorithms. Their benefits and limitations are displayed in Table 4.

3.3. Sensor-Augmented Pump (SAP)

An SAP is a CSII that can integrate data from a CGM system. Glycemia is displayed on the pump in real time. It is used by the pump algorithm to automatically stop the basal insulin infusion (for up to 2 h) as a response to detected/predicted hypoglycemia. Then, the basal insulin infusion is released at the previously programmed rate. This feature helps diminish moderate-to-severe hypoglycemia, especially during nighttime, and reach better glycemic control [88]. SAPs are known as open-loop systems [89].
There are two types of SAPT available on the market:
Low-glucose suspend (SAPT-LGS): Suspends basal rate when hypoglycemia occurs.
Predictive low-glucose management (SAPT-PLGM): Can suspend basal rate before hypoglycemia occurs.
SAP can reduce hypoglycemia by 40–50% (<70 mg/dL) without an increase in glycosylated hemoglobin [90,91,92,93].

3.4. Closed-Loop Insulin Systems (Artificial Pancreas)

A CGM could become a part of a CSII through an algorithm, generating a closed-loop insulin system (Figure 4A). It is a substantially improved system, adjusting insulin delivery in response to real-time sensor glucose levels and other inputs, such as meal intake (Figure 4B). Control algorithms can modulate the insulin needs’ variability between and within individual users, considering CGM accuracy limitations and insulin delivery imprecision [94].
The three main components of a closed-loop system are (Figure 4A):
Glucose measuring device (CGM);
Control device for BG analysis and insulin dosing regulation (computer/microprocessor);
Insulin infusion device (insulin pump).
The control algorithms are continuously adapted to physiological changes with real-time adjustment of closed-loop control parameters (Figure 5).
Various control algorithms were developed [97]: model predictive iterative learning control (MPC) [98,99,100], proportional integral derivative (PID) controllers [101,102], and fuzzy logic control approaches [103,104].
The closed-loop system functions as a pancreas that controls BG levels. Thus, the closed-loop insulin system is known as the artificial pancreas (AP) [105]. When an AP system requires counting and registering the carbohydrate amount from mealtime, it is called a “hybrid” [106] because a part of insulin is provided automatically, and another is infused based on the reported information.
In 2016, the FDA approved the first hybrid closed-loop system [107]. It automatically gives a suitable amount of short-acting insulin at a basal rate. The patient still needs a glucose meter a few times daily, manually adjusting the insulin delivery at mealtimes and when it requires a dose correction. Nowadays, there are several FDA- and EMA-approved systems: Medtronic 770/780G, Tandem Control IQ, Omnipod 5, CamAPS, Diabeloop, etc. (Table 5).
Another artificial pancreas system is known as a hormonal bionic pancreas (BP) [109,110,111]. It has the next-generation technology to deliver insulin and/or glucagon automatically rather than standard-of-care management. Therefore, BP is more effective in maintaining blood glucose levels within the normal range in T1DM people [112]. In May 2023, the Beta Bionics Inc. (Boston, MA, USA) iLet ACE Pump and iLet Dosing Decision Software pancreas system [109,111,113,114,115] received FDA approval.
Like a healthy pancreas, an utterly automated closed-loop system does not request meal announcements; it can react to BG level variations [116]. The benefits and limitations of closed-loop systems are given in Table 6.

Complications

Hypoglycemia occurs when a significant basal rate of insulin is delivered due to a human error in insulin pump programming or a device malfunction.
Hyperglycemia is caused by programming error or device malfunction, leading to a low insulin delivery rate (battery depletion or malposition, cannula occlusion, total pump failure).
If the infusion set is not changed regularly, at 3–4 days, there are irritation and infections at the place of cannula insertion.
Insulin pump therapy discontinuation (18–50%) is the T1DM patient choice for various reasons: unwanted interference with the lifestyle, missing improvements in glycemic control, and infection at the insertion place. It occurs with high incidence in women, younger individuals, pregnancy, and when the patient has psychological comorbidities.

4. Decision Support Systems

As T1DM individuals play a substantial role in their healthcare, artificial intelligence applications known as decision support systems (DSSs) can help them in decision making and reacting in real time to maintain normal BGLs [117]. The DSSs notably impact the users’ decision making, increasing their confidence in their potential to deal with T1DM (Figure 6). They integrate data such as insulin-to-carb ratio, insulin sensitivity, insulin time of action, sick days, and exercise response. A DSS suggests the best therapeutic option for better glycemic control, higher time in range, diminished hypoglycemic events, and decreased HbA1c.

5. The Impact of New Technologies on T1DM People’s Quality of Life

5.1. Evaluation of Diabetes Distress

The T1DM adult questionnaire has a self-report scale with 28 items, evidencing seven critical points of diabetes distress. These seven items refer to crucial feelings of T1DM adults, according to Fisher et al. [119] (Figure 7).
Open discussions with T1DM subjects represent a clinical instrument for their emotional behavior evaluation.

5.2. Satisfaction Survey for Diabetes Technology Users

The Glucose Monitors Satisfaction Survey (GMSS) [120] was conceived for T1DM individuals as a 15-item self-report scale, with three key dimensions:
Openness;
Emotional and behavioral burdens;
Trust.
The Insulin Device Satisfaction Survey (IDSS) is a self-report scale with a 14-item version for T1DM [121], with three key dimensions:
Effectiveness;
Burdensomeness;
Inconvenience.
All these documents are available in various languages and accessible to non-profit institutions for use in clinical care and research. Concomitantly, they can be accessed as copyright with licensing fees by all for-profit companies and institutions on https://behavioraldiabetes.org/scales-and-measures/ (accessed on 9 April 2023).

5.3. Quality of Life Evaluation

Joensen et al. [122] analyzed the effect of continuous care and support for T1DM adults with insulin pump therapy. The flexible and participatory peer support approach augmented individuals’ motivation and empowerment, induced a feeling of serenity about diabetes management, and demonstrated the potential to diminish diabetes distress, thus increasing their QoL. Adequate diabetes-specific social support avoids the risk of participants feeling isolated with peer support groups and allows free interactive dialogue regarding T1DM disease and all the difficulties of coping with it. Evaluation of motivation, serenity in life with diabetes, awareness of own diabetes practices, diabetes empowerment, diabetes loneliness, and diabetes distress are applicable, feasible, and appropriate when measuring the effect of peer support on adults with insulin-pump-treated diabetes.
Rizvi et al. evaluated the impact of frequent interactions between healthcare providers and individuals using CGMs in a diabetes center. It involved virtual video visits with high frequency and an electronic messaging system. CGM data were downloaded proactively every 2 weeks from computer cloud-based device accounts of 146 individuals with diabetes on multiple (2–5) daily insulin injections or insulin pump therapy. Then, the diabetes specialist analyzed and interpreted the glucose profile and individually advised each patient regarding diet, lifestyle, and insulin regimen. The communication used an electronic portal linked to email during telehealth and in-person office visits. As a result, the adherence was approximately 100% [123].
Introducing technology in T1DM management in individuals with a diabetes duration of over 30 years also encounters multiple obstacles. Our team’s experience materialized in a model of the efficiency of this process. Following this model and after the treatment change, we obtained a 1.3% reduction in HbA1c and significantly fewer hypoglycemic events, RR 34% (p < 0.01). People’s acceptance of the new treatment increased in time by 92%. The implementation process effectively achieved this transformation, reached glycemic targets, and changed patients’ lifestyles [124].
Recently, Fanzolla et al. supervised a two-section questionnaire (children/parents) to evaluate the impact of CGM on T1DM children and their families. The data show that CGM devices significantly improved the QoL of children and their parents, as 80% of children reported that CGM subcutaneous placement is much less painful than fingertips. Moreover, the QoL at school and sports is significantly better: diminished anxiety, higher comfort, and better glycemic control. Also, 90% of parents stated that CGM devices remarkably improved the glycemic control of their children, reducing emerging events. A similar percentage (89% of parents) are convinced that the recent technology generated substantial benefits for their children’s QoL [125].
Rusak et al. [126] evaluated the quality of life and satisfaction in children under 7 years of age with T1DM using the RT-CGM system integrated with an insulin pump, investigating their caregivers’ opinions. They reported high satisfaction with CGM use (68% in groups aged 5–7 years and 92% in 2–4 years), and 71% of caregivers confirmed the positive effect of CGM on their sleep quality.
Polonsky et al. [127] analyzed the CGM impact on the quality of life in T1DM adults. The participants completed a QoL evaluation (regarding overall well-being, health status, DD, confidence, and hypoglycemic fear) and a CGM satisfaction survey. CGM satisfaction was not considerably associated with glycemic levels. The satisfaction was directly correlated with diminishing diabetes distress and hypoglycemic fear and increasing well-being and hypoglycemic confidence [127]. Therefore, the CGM group reported higher hypoglycemic confidence and diabetes distress reduction than the SMBG, but with no statistically significant differences.
Fear of hypoglycemic episodes is also why people with T1DM restrain from regular physical activity [128]. However, most consider that physical exercise generates increased well-being and QOL, but there are still many barriers to engaging in physical activity. Lack of time and support from family or friends was also reported. Practicing exercise improved TIR and decreased TAR; eating before and turning off the pump during the exercise were associated with lower TBR after exercise [129].
CGM usage has proven benefits in glycemic control and decreased the frequency of hypoglycemic events during and after exercise. Pump therapy provides the tools for better insulin management by adjusting basal rates. Hybrid closed-loop systems may be the solution, but for the moment, physical exercise remains a challenge for these systems [130].
The Satisfaction Survey on Individuals With Artificial Pancreas [116] did not yet clearly reveal a clear impact on fear of hypoglycemia, adherence, quality of life, depression and anxiety, and diabetes distress.
However, T1DM children from a summer camp [131] revealed the benefits of a bionic pancreas in the following ways:
Reducing their fear of hypoglycemia;
Decreasing their sense of regimen burden;
Diminishing their worries about out-of-range blood sugar levels;
Improving their overall freedom to engage in activities that they enjoyed.
In addition, their concerns about the BP included wishing the system responded to out-of-range blood sugar levels more quickly and the annoyance of carrying several devices around [131].

6. Discussion

BG self-monitoring is crucial to the daily management of T1DM. However, the adherence rate is low. A recent study [132] reported a 61.6% adherence rate to the Spanish Diabetes Society protocol for SMBG. The authors identified the associated factors: the frequency of insulin injections (<3 injections daily), alcohol abstinence, peripheral vascular disease, and retrieval of the reactive strips from the pharmacy. Only 21.4% of individuals had an excellent self-perception of glycemia.
Peralta et al. [133] performed a cross-sectional observational study in adults and children treated with basal-bolus therapy and CSII users, aiming at T1DM management. They observed that metabolic control (expressed as HbA1c) positively correlates with higher educational levels, carbohydrate counting, more daily SMBG, and fewer hypoglycemic episodes. It decreases with T1DM duration, higher insulin total dose, low adherence to diet, and a family history of DM [133].
Spaan et al. [134] reported that adherence to insulin pumps in T1DM adolescents declined with age. The transition from parental care to adolescent self-caring patients led to this. Moreover, more conditions are requested to maintain the T1DM pediatric patients’ adherence to CSII therapy. Giany et al. [135] observed that insulin pumps are underused in T1DM youths. Their study integrated the evaluation of biomedical and psychosocial factors associated with consistent and durable CGM use over time. Their results were confirmed by Trandafir et al. [136], who showed that close family members (parents) and healthcare professionals have an essential role in their adherence to insulin pump therapy, together with other determinants (Figure 8).
The use of CGM by individuals with diabetes is correlated with psychological benefits and burdens [137]. Potential benefits consist of improved QoL and diminished hypoglycemia fear. However, randomized clinical trials versus cohort studies for youth and adults show heterogeneous results. In correspondent studies, adolescents and adults have nearly universally reported substantially perceived satisfaction regarding CGM use. The specific benefits were linked to easier diabetes management and better glycemic control. In contrast, limitations included pain and body issues, communication problems, feeling awestruck by the complex glucose management, and doubts regarding the accuracy of CGM compared to glucometer readings [138]. Increased anxiety of adolescents and parents and poorer parental sleep has also been remarked [139].
De Bock et al. [140] investigated users of sensor-augmented pumps in a 6-month clinical trial. Variable parameters were examined (individuals’ age and gender, the values of HbA1c, diabetes duration, frequency of BG testing, sensor accuracy, and the frequency of insulin pump alarm) and associated with CGM adherence. CGM adherence was 75% (35% to 96%), and age was the only variable significantly correlated with CGM adherence.
On the other hand, younger clinicians managing individuals who used insulin pumps and CGMs recorded more positive attitudes concerning diabetes technology [141] and identified different barriers to patient adherence: device on the body (73% pump; 63% CGM), alarms (61% CGM), and misunderstanding of the procedure (40% pump; 46% CGM). Numerous recent studies showed the positive impact of DSS and closed-loop systems on T1DM users. Thus, Breton et al. [142] reported the benefits of a DSS formed by two real-time advisors (CGM-informed bolus advisor and exercise advisor) and a retrospective insulin titration tool significantly reducing BGL variation in 48 h. In another study [117], the T1DM participants modified their initial decision 20% of the time based on DSS clinical indications. With automated exercise detection as an additional signal, a dual-hormone AP diminished hypoglycemia during exercise in the study period [143]. Technology such as CGM and pumps provides detailed information on glucose variation before, during, and after exercise and can prevent exercise-related hypoglycemic events. Also, the development of new smartphone apps designed for people with type 1 diabetes can improve time in range and diminish time below range by suggesting the appropriate amount of carbs before or after exercise and by helping the patient to adjust his/her insulin regimen [144,145]. Further planned applications of DSS will be in exercise decision and exercise as an adjunct therapy, optimizing meal bolus timing and other time-varying dosing parameters, and pregnancy. In addition, the DSS will be integrated with AID [118].
However, the healthcare resource waste associated with nonadherence to CGM and/or pump therapy and early discontinuation of device usage justifies rigorous consideration in selecting suitable patients for these technologies.
To diminish the unwanted complications of insulin administration and increase the patient’s compliance, various research teams tried to incorporate it in multiple carriers for oral administration [146,147,148]: liposomes [149,150] mixed micelles [151,152], nanoparticles [153,154,155], intestinal patches [156]. Current studies also explore other access ways for insulin administration: intranasal [157,158,159], transdermal [160,161], and sublingual [162,163].
Recent studies analyzed glucose-responsive biomaterials [163], especially for rapid and extended self-regulated insulin delivery [164]. Moreover, using chitosan hydrogels integrated with glucose-responsive microspheres for insulin delivery, Yin et al. [165] developed a synthetic artificial pancreas. However, new biomaterials or a substantial variety of composite ones are necessary to build a functional bioartificial pancreas with proper mechanical strengths and biological activities [166]. These properties can be achieved using 3D bioprinting technology. An innovative process in advanced tissue engineering aims to construct clinically applicable bioartificial pancreatic islet tissue [167] with a native tissue environment. A 3D bio-printed pancreas is expected to have critical applications for future diabetes treatment.

7. Conclusions

All presented data evidence that current emerging technologies and control systems significantly improve T1DM self-management. Moreover, unconditional, continuous medical and social help is essential in increasing their self-confidence, motivation, and adherence to CSII, minimizing the impact of other factors (family incomes, requested education, and the ability for technology use). The quality of life of T1DM individuals could substantially increase when the performances of advanced devices and algorithms are associated with considerable support from family and healthcare providers.
However, the technological systems’ limitations and potential adverse effects and complications lead to continuous worldwide research on finding alternative approaches to T1DM therapy.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/life13081663/s1, Table S1: Various insulin PPs, adapted from [86,87].

Author Contributions

Conceptualization, V.E., V.P., E.-A.O. and D.L.; methodology, V.P. and E.-A.O.; software, V.P.; validation, V.E., E.-A.O. and D.L.; formal analysis, V.P., A.M.M., A.C.F., E.R. and G.R.; investigation, V.P. and E.-A.O.; resources, V.E.; data curation, A.M.M., A.C.F., E.R. and G.R.; writing—original draft preparation, V.P.; writing—review and editing, V.E., V.P., E.R. and G.R.; visualization, V.E., E.-A.O., A.M.M., A.C.F., E.R., G.R. and D.L.; supervision, V.E., E.-A.O. and D.L.; project administration, V.E., E.-A.O. and D.L.; funding acquisition, V.E., E.-A.O. and A.M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available in the MS and Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. King, H.; Rewers, M. Diabetes in adults is now a Third World problem. The WHO Ad Hoc Diabetes Reporting Group. Bull. World Health Organ. 1991, 69, 643–648. [Google Scholar]
  2. Alam, S.; Hasan, M.K.; Neaz, S.; Hussain, N.; Hossain, M.F.; Rahman, T. Diabetes Mellitus: Insights from Epidemiology, Biochemistry, Risk Factors, Diagnosis, Complications and Comprehensive Management. Diabetology 2021, 2, 36–50. [Google Scholar] [CrossRef]
  3. Schmidt, A.M. Highlighting Diabetes Mellitus. Arterioscler. Thromb. Vasc. Biol. 2018, 38, e1–e8. [Google Scholar] [CrossRef] [Green Version]
  4. Roep, B.O.; Thomaidou, S.; van Tienhoven, R.; Zaldumbide, A. Type 1 diabetes mellitus as a disease of the β-cell (do not blame the immune system?). Nat. Rev. Endocrinol. 2021, 17, 150–161. [Google Scholar] [CrossRef]
  5. Pozzilli, P.; Maddaloni, E.; Buzzetti, R. Combination immunotherapies for type 1 diabetes mellitus. Nat. Rev. Endocrinol. 2015, 11, 289–297. [Google Scholar] [CrossRef]
  6. Bone, R.N.; Evans-Molina, C. Combination Immunotherapy for Type 1 Diabetes. Curr. Diab. Rep. 2017, 17, 50. [Google Scholar] [CrossRef]
  7. Thomas, R.; Carballido, J.M.; Wesley, J.D.; Ahmed, S.T. Overcoming Obstacles in the Development of Antigen-Specific Immunotherapies for Type 1 Diabetes. Front. Immunol. 2021, 12, 730414. [Google Scholar] [CrossRef]
  8. von Scholten, B.J.; Kreiner, F.F.; Gough, S.C.L.; von Herrath, M. Current and future therapies for type 1 diabetes. Diabetologia 2021, 64, 1037–1048. [Google Scholar] [CrossRef]
  9. Felton, J.L. Timing of Immunotherapy in Type 1 Diabetes: The Earlier, the Better? Immunohorizons 2021, 5, 535–542. [Google Scholar] [CrossRef]
  10. Niedźwiedzka-Rystwej, P.; Wołącewicz, M.; Cywoniuk, P.; Klak, M.; Wszoła, M. Crosstalk Between Immunity System Cells and Pancreas. Transformation of Stem Cells Used in the 3D Bioprinting Process as a Personalized Treatment Method for Type 1 Diabetes. Arch. Immunol. Ther. Exp. 2020, 68, 13. [Google Scholar] [CrossRef]
  11. Sims, E.K.; Bundy, B.N.; Stier, K.; Serti, E.; Lim, N.; Long, S.A.; Geyer, S.M.; Moran, A.; Greenbaum, C.J.; Evans-Molina, C.; et al. Teplizumab improves and stabilizes beta cell function in antibody-positive high-risk individuals. Sci. Transl. Med. 2021, 13, eabc8980. [Google Scholar] [CrossRef]
  12. Frontino, G.; Guercio, N.S.; Scaramuzza, A.; Annunzio, G.D.; Toni, S.; Citriniti, F.; BOnfanti, R. Prevention of type 1 diabetes: Where we are and where we are going. Minerva Pediatr. 2021, 73, 486–503. [Google Scholar] [CrossRef]
  13. Perdigoto, A.L.; Preston-Hurlburt, P.; Clark, P.; Long, S.A.; Linsley, P.S.; Harris, K.M.; Gitelman, S.E.; Greenbaum, C.J.; Gottlieb, P.A.; Hagopian, W.; et al. Treatment of type 1 diabetes with teplizumab: Clinical and immunological follow-up after 7 years from diagnosis. Diabetologia 2019, 62, 655–664. [Google Scholar] [CrossRef] [Green Version]
  14. Kwong, S.P.; Wang, C. Review: Usnic acid-induced hepatotoxicity and cell death. Environ. Toxicol. Pharmacol. 2020, 80, 103493. [Google Scholar] [CrossRef]
  15. Hermansen, K.; Fontaine, P.; Kukolja, K.K.; Peterkova, V.; Leth, G.; Gall, M.A. Insulin analogues (insulin detemir and insulin aspart) versus traditional human insulins (NPH insulin and regular human insulin) in basal-bolus therapy for patients with Type 1 diabetes. Diabetologia 2004, 47, 622–629. [Google Scholar] [CrossRef] [Green Version]
  16. Lucidi, P.; Porcellati, F.; Andreoli, A.M.; Carriero, I.; Candeloro, P.; Cioli, P.; Bolli, G.B.; Fanelli, C.G. Pharmacokinetics and pharmacodynamics of NPH insulin in type 1 diabetes: The Importance of appropriate resuspension before subcutaneous injection. Diabetes Care 2015, 38, 2204–2210. [Google Scholar] [CrossRef] [Green Version]
  17. Ross, L.J.; Neville, K.A. Continuous subcutaneous insulin infusion versus multiple daily injections for type 1 diabetes. J. Paediatr. Child Health 2019, 55, 718–722. [Google Scholar] [CrossRef]
  18. Savu, O.; Elian, V.; Steriade, O.; Teodoru, I.; Mihut, S.; Tacu, C.; Covic, A.; Serafinceanu, C. The impact of basal insulin analogues on glucose variability in patients with type 2 diabetes undergoing renal replacement therapy for end-stage renal disease. Int. Urol. Nephrol. 2016, 48, 265–270. [Google Scholar] [CrossRef]
  19. Brown-Georgi, J.; Chhabra, H.; Vigersky, R.A. The Rising Cost of Insulin for Pump Users: How Policy Drives Prices. J. Diabetes Sci. Technol. 2020, 15, 1177–1180. [Google Scholar] [CrossRef]
  20. Lawton, J.; Kirkham, J.; Rankin, D.; White, D.A.; Elliott, J.; Jaap, A.; Smithson, W.H.; Heller, S.; Gianfrancesco, C.; Gordon, V.; et al. Who gains clinical benefit from using insulin pump therapy? A qualitative study of the perceptions and views of health professionals involved in the Relative Effectiveness of Pumps over MDI and Structured Education (REPOSE) trial. Diabet. Med. 2016, 33, 243–251. [Google Scholar] [CrossRef]
  21. Herkert, D.; Vijayakumar, P.; Luo, J.; Schwartz, J.I.; Rabin, T.L.; Defilippo, E.; Lipska, K.J. Cost-Related Insulin Underuse among Patients with Diabetes. JAMA Intern. Med. 2019, 179, 112–114. [Google Scholar] [CrossRef]
  22. Moroșan, E.; Popovici, V.; Elian, V.; Dărăban, A.M.; Rusu, A.I.; Licu, M.; Mititelu, M.; Karampelas, O. The Impact of Medical Nutrition Intervention on the Management of Hyperphosphatemia in Hemodialysis Patients with Stage 5 Chronic Kidney Disease: A Case Series. Int. J. Environ. Res. Public Health 2023, 20, 5049. [Google Scholar] [CrossRef]
  23. Stankute, I.; Radzeviciene, L.; Monstaviciene, A.; Dobrovolskiene, R.; Danyte, E.; Verkauskiene, R. Serum Cystatin C as a Biomarker for Early Diabetic Kidney Disease and Dyslipidemia in Young Type 1 Diabetes Patients. Medicina 2022, 58, 218. [Google Scholar] [CrossRef]
  24. Eliasson, B.; Lyngfelt, L.; Strömblad, S.O.; Franzén, S.; Eeg-Olofsson, K. The significance of chronic kidney disease, heart failure and cardiovascular disease for mortality in type 1 diabetes: Nationwide observational study. Sci. Rep. 2022, 12, 17950. [Google Scholar] [CrossRef]
  25. Genuth, S. Insights from the diabetes control and complications trial/epidemiology of diabetes interventions and complications study on the use of intensive glycemic treatment to reduce the risk of complications of type 1 diabetes. Endocr. Pract. 2006, 12, 34–41. [Google Scholar] [CrossRef]
  26. Niazi, A.K.; Kalra, S. Diabetes and tuberculosis: A review of the role of optimal glycemic control. J. Diabetes Metab. Disord. 2012, 11, 28. [Google Scholar] [CrossRef] [Green Version]
  27. da Silva de Sousa, G.G.; Yamamura, M.; de Araújo, M.F.M.; Ramos, A.C.V.; Arcêncio, R.A.; de Jesus Costa, A.C.P.; Pascoal, L.M.; Santos, F.S.; de Oliveira Serra, M.A.A.; Fontoura, I.G.; et al. Vulnerable territories to tuberculosis-diabetes mellitus comorbidity in a northeastern Brazilian scenario. J. Infect. Dev. Ctries. 2022, 16, 813–820. [Google Scholar] [CrossRef]
  28. Kaur, N.; Bhadada, S.K.; Minz, R.W.; Dayal, D.; Kochhar, R. Interplay between type 1 diabetes mellitus and celiac disease: Implications in treatment. Dig. Dis. 2018, 36, 399–408. [Google Scholar] [CrossRef]
  29. Kogut, M.D.; Brinegar, C.H. Addison’s disease and diabetes mellitus. J. Pediatr. 1972, 81, 307–311. [Google Scholar] [CrossRef]
  30. Ingley, M.; Quebedeaux, P.; Schmidbauer, K.; Amghaiab, I.A.; Chan, J. PMON315 Hyperglycemia Dilemma: Concomitant Type 1 Diabetes Mellitus and Cushing’s disease. J. Endocr. Soc. 2022, 6, A624–A625. [Google Scholar] [CrossRef]
  31. Ilie, I.; Ciubotaru, V.; Tulin, A.; Hortopan, D.; Caragheorgheopol, A.; Purice, M.; Neamtu, C.; Elian, V.I.; Banica, A.; Oprea, L.; et al. The multifarious cushing’s–Lessons from a case series. Acta Endocrinol. 2019, 15, 261–269. [Google Scholar] [CrossRef] [PubMed]
  32. Wardian, J.L.; True, M.W.; Folaron, I.; Colburn, J.; Tate, J.M.; Beckman, D.J. The Choice Should Be Yours: Diabetes-Related Distress by Insulin Delivery Method for People with Type 1 Diabetes. Diabetes Technol. Ther. 2020, 22, 42–47. [Google Scholar] [CrossRef]
  33. Tareen, R.S.; Tareen, K. Psychosocial aspects of diabetes management: Dilemma of diabetes distress. Transl. Pediatr. 2017, 6, 383–396. [Google Scholar] [CrossRef] [Green Version]
  34. Barnard, K.; Crabtree, V.; Adolfsson, P.; Davies, M.; Kerr, D.; Kraus, A.; Gianferante, D.; Bevilacqua, E.; Serbedzija, G. Impact of Type 1 Diabetes Technology on Family Members/Significant Others of People with Diabetes. J. Diabetes Sci. Technol. 2016, 10, 824–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Davies, M. Psychological aspects of diabetes management. Medicine 2022, 50, 749–751. [Google Scholar] [CrossRef]
  36. Mianowska, B.; Fedorczak, A.; Michalak, A.; Pokora, W.; Barańska-Nowicka, I.; Wilczyńska, M.; Szadkowska, A. Diabetes related distress in children with type 1 diabetes before and during the covid-19 lockdown in spring 2020. Int. J. Environ. Res. Public Health 2021, 18, 8527. [Google Scholar] [CrossRef]
  37. Luo, J.; Wang, H.; Li, X.; Zhou, Z.; Valimaki, M.; Whittemore, R.; Grey, M.; Guo, J. Factors associated with diabetes distress among adolescents with type 1 diabetes. J. Clin. Nurs. 2021, 30, 1893–1903. [Google Scholar] [CrossRef] [PubMed]
  38. Schmidt, C.B.; Voorhorst, I.; Van De Gaar, V.H.W.; Keukens, A.; Potter Van Loon, B.J.; Snoek, F.J.; Honig, A. Diabetes distress is associated with adverse pregnancy outcomes in women with gestational diabetes: A prospective cohort study. BMC Pregnancy Childbirth 2019, 19, 223. [Google Scholar] [CrossRef] [Green Version]
  39. Skinner, T.C.; Joensen, L.; Parkin, T. Twenty-five years of diabetes distress research. Diabet. Med. 2020, 37, 393–400. [Google Scholar] [CrossRef] [Green Version]
  40. De Wit, M.; Pouwer, F.; Snoek, F.J. How to identify clinically significant diabetes distress using the Problem Areas in Diabetes (PAID) scale in adults with diabetes treated in primary or secondary care? Evidence for new cut points based on latent class analyses. BMJ Open 2022, 12, e056304. [Google Scholar] [CrossRef] [PubMed]
  41. Polonsky, W.H.; Fisher, L.; Earles, J.; Dudl, R.J.; Lees, J.; Mullan, J.; Jackson, R.A. Assessing psychosocial distress in diabetes: Development of the Diabetes Distress Scale. Diabetes Care 2005, 28, 626–631. [Google Scholar] [CrossRef] [Green Version]
  42. Rubin, R.R.; Peyrot, M. Health-related quality of life and treatment satisfaction in the sensor-augmented pump therapy for A1C reduction 3 (STAR 3) trial. Diabetes Technol. Ther. 2012, 14, 143–151. [Google Scholar] [CrossRef] [Green Version]
  43. Klamann, M.; Majkowska, L. New technologies and metabolic control in type 1 diabetes mellitus. Clin. Diabetol. 2017, 6, 111–114. [Google Scholar] [CrossRef] [Green Version]
  44. Kimbell, B.; Lawton, J.; Boughton, C.; Hovorka, R.; Rankin, D. Parents’ experiences of caring for a young child with type 1 diabetes: A systematic review and synthesis of qualitative evidence. BMC Pediatr. 2021, 21, 160. [Google Scholar] [CrossRef] [PubMed]
  45. Pauley, M.E.; Berget, C.; Messer, L.H.; Forlenza, G.P. Barriers to uptake of insulin technologies and novel solutions. Med. Devices Evid. Res. 2021, 14, 339–354. [Google Scholar] [CrossRef]
  46. Pease, A.; Zomer, E.; Liew, D.; Lo, C.; Earnest, A.; Zoungas, S. Cost-effectiveness of health technologies in adults with type 1 diabetes: A systematic review and narrative synthesis. Syst. Rev. 2020, 9, 171. [Google Scholar] [CrossRef]
  47. Prahalad, P.; Tanenbaum, M.; Hood, K.; Maahs, D.M. Diabetes technology: Improving care, improving patient-reported outcomes and preventing complications in young people with Type 1 diabetes. Diabet. Med. 2018, 35, 419–429. [Google Scholar] [CrossRef] [Green Version]
  48. Gonder-Frederick, L.A.; Shepard, J.A.; Grabman, J.H.; Ritterband, L.M. Psychology, technology, and diabetes management. Am. Psychol. 2016, 71, 577–589. [Google Scholar] [CrossRef] [Green Version]
  49. Cohen, M.; Boyle, E.; Delaney, C.; Shaw, J. A comparison of blood glucose meters in Australia. Diabetes Res. Clin. Pract. 2006, 71, 113–118. [Google Scholar] [CrossRef]
  50. Apostolopoulos, A.; Apostolopoulou, D.; Tsoubeli, A. Application of health informatics in the education of diabetic patients for the improvement of self-management and reporting to specialists. J. Inf. Technol. Healthc. 2007, 5, 379–386. [Google Scholar]
  51. Umpierrez, G.E.; Klonoff, D.C. Diabetes technology update: Use of insulin pumps and continuous glucose monitoring in the hospital. Diabetes Care 2018, 41, 1579–1589. [Google Scholar] [CrossRef] [Green Version]
  52. Yeh, T.; Yeung, M.; Mendelsohn Curanaj, F.A. Managing Patients with Insulin Pumps and Continuous Glucose Monitors in the Hospital: To Wear or Not to Wear. Curr. Diab. Rep. 2021, 21, 7. [Google Scholar] [CrossRef]
  53. Olczuk, D.; Priefer, R. A history of continuous glucose monitors (CGMs) in self-monitoring of diabetes mellitus. Diabetes Metab. Syndr. Clin. Res. Rev. 2018, 12, 181–187. [Google Scholar] [CrossRef]
  54. Franceschi, R.; Micheli, F.; Mozzillo, E.; Cauvin, V.; Liguori, A.; Soffiati, M.; Giani, E. Intermittently Scanned and Continuous Glucose Monitor Systems: A Systematic Review on Psychological Outcomes in Pediatric Patients. Front. Pediatr. 2021, 9, 660173. [Google Scholar] [CrossRef]
  55. Visser, M.M.; Charleer, S.; Fieuws, S.; De Block, C.; Hilbrands, R.; Van Huffel, L.; Maes, T.; Vanhaverbeke, G.; Dirinck, E.; Myngheer, N.; et al. Comparing real-time and intermittently scanned continuous glucose monitoring in adults with type 1 diabetes (ALERTT1): A 6-month, prospective, multicentre, randomised controlled trial. Lancet 2021, 397, 2275–2283. [Google Scholar] [CrossRef]
  56. Cappon, G.; Acciaroli, G.; Vettoretti, M.; Facchinetti, A.; Sparacino, G. Wearable continuous glucose monitoring sensors: A revolution in diabetes treatment. Electron. 2017, 6, 65. [Google Scholar] [CrossRef] [Green Version]
  57. Zafar, H.; Channa, A.; Jeoti, V.; Stojanović, G.M. Comprehensive Review on Wearable Sweat-Glucose Sensors for Continuous Glucose Monitoring. Sensors 2022, 22, 638. [Google Scholar] [CrossRef]
  58. Staal, O.M.; Hansen, H.M.U.; Christiansen, S.C.; Fougner, A.L.; Carlsen, S.M.; Stavdahl, Ø. Differences between flash glucose monitor and fingerprick measurements. Biosensors 2018, 8, 93. [Google Scholar] [CrossRef] [Green Version]
  59. Fokkert, M.; Van Dijk, P.R.; Edens, M.A.; Díez Hernández, A.; Slingerland, R.; Gans, R.; Delgado Álvarez, E.; Bilo, H. Performance of the Eversense versus the Free Style Libre Flash glucose monitor during exercise and normal daily activities in subjects with type 1 diabetes mellitus. BMJ Open Diabetes Res. Care 2020, 8, e001193. [Google Scholar] [CrossRef]
  60. Ahmed, I.; Jiang, N.; Shao, X.; Elsherif, M.; Alam, F.; Salih, A.; Butt, H.; Yetisen, A.K. Recent advances in optical sensors for continuous glucose monitoring. Sens. Diagn. 2022, 1, 1098–1125. [Google Scholar] [CrossRef]
  61. Gaynanova, I.; Punjabi, N.; Crainiceanu, C. Modeling continuous glucose monitoring (CGM) data during sleep. Biostatistics 2022, 23, 223–239. [Google Scholar] [CrossRef]
  62. Urakami, T. The Advanced Diabetes Technologies for Reduction of the Frequency of Hypoglycemia and Minimizing the Occurrence of Severe Hypoglycemia in Children and Adolescents with Type 1 Diabetes. J. Clin. Med. 2023, 12, 781. [Google Scholar] [CrossRef]
  63. Kropff, J.; DeVries, J.H. Continuous Glucose Monitoring, Future Products, and Update on Worldwide Artificial Pancreas Projects. Diabetes Technol. Ther. 2016, 18, S253–S263. [Google Scholar] [CrossRef]
  64. Mensh, B.D.; Wisniewski, N.A.; Neil, B.M.; Burnett, D.R. Susceptibility of interstitial continuous glucose monitor performance to sleeping position. J. Diabetes Sci. Technol. 2013, 7, 863–870. [Google Scholar] [CrossRef] [Green Version]
  65. Russell, S.J.; El-Khatib, F.H.; Sinha, M.; Magyar, K.L.; Mckeon, K.; Goergen, L.G.; Hillard, M.A.; Nathan, D.M.; Damiano, E.R. Multiday outpatient glycemic control in adolescents with type 1 diabetes using a bihormonal bionic pancreas: The barton center summer camp study. Diabetes 2014, 63, A62. [Google Scholar]
  66. Shapiro, A.R. Nonadjunctive Use of Continuous Glucose Monitors for Insulin Dosing: Is It Safe? J. Diabetes Sci. Technol. 2017, 11, 833–838. [Google Scholar] [CrossRef] [Green Version]
  67. Beck, R.W.; Riddlesworth, T.D.; Ruedy, K.; Ahmann, A.; Haller, S.; Kruger, D.; McGill, J.B.; Polonsky, W.; Price, D.; Aronoff, S.; et al. Continuous glucose monitoring versus usual care in patients with type 2 diabetes receiving multiple daily insulin injections. Ann. Intern. Med. 2017, 167, 365–374. [Google Scholar] [CrossRef]
  68. Vettoretti, M.; Cappon, G.; Facchinetti, A.; Sparacino, G. Advanced diabetes management using artificial intelligence and continuous glucose monitoring sensors. Sensors 2020, 20, 3870. [Google Scholar] [CrossRef]
  69. Tagougui, S.; Taleb, N.; Rabasa-Lhoret, R. The benefits and limits of technological advances in glucose management around physical activity in patients type 1 diabetes. Front. Endocrinol. 2019, 10, 818. [Google Scholar] [CrossRef]
  70. Von Dem Berge, T.; Biester, S.; Biester, T.; Buchmann, A.K.; Datz, N.; Grosser, U.; Kapitzke, K.; Klusmeier, B.; Remus, K.; Reschke, F.; et al. Recommendations for Diabetes-Treatment with Systems for Automated Insulin Delivery. Diabetol. Und Stoffwechs. 2022, 17, 61–73. [Google Scholar]
  71. Wu, P.T.; Segovia, D.E.; Lee, C.C.; Nguyen, K.L. Consistency of continuous ambulatory interstitial glucose monitoring sensors. Biosensors 2018, 8, 49. [Google Scholar] [CrossRef] [Green Version]
  72. Helton, K.L.; Ratner, B.D.; Wisniewski, N.A. Biomechanics of the sensor-tissue interface-Effects of motion, pressure, and design on sensor performance and foreign body response-Part II: Examples and application. J. Diabetes Sci. Technol. 2011, 5, 647–656. [Google Scholar] [CrossRef] [Green Version]
  73. Petrofsky, J.S.; McLellan, K.; Prowse, M.; Bains, G.; Berk, L.; Lee, S. The effect of body fat, aging, and diabetes on vertical and shear pressure in and under a waist belt and its effect on skin blood flow. Diabetes Technol. Ther. 2010, 12, 153–160. [Google Scholar] [CrossRef] [Green Version]
  74. Ardilouze, J.L.; Karpe, F.; Currie, J.M.; Frayn, K.N.; Fielding, B.A. Subcutaneous adipose tissue blood flow varies between superior and inferior levels of the anterior abdominal wall. Int. J. Obes. 2004, 28, 228–233. [Google Scholar] [CrossRef] [Green Version]
  75. Cengiz, E.; Tamborlane, W.V. A tale of two compartments: Interstitial versus blood glucose monitoring. Diabetes Technol. Ther. 2009, 11, S11–S16. [Google Scholar] [CrossRef]
  76. Weinstein, R.L.; Schwartz, S.L.; Brazg, R.L.; Bugler, J.R.; Peyser, T.A.; McGarraugh, G.V. Accuracy of the 5-Day FreeStyle Navigator Continuous Glucose Monitoring System. Diabetes Care 2007, 30, 1125–1130. [Google Scholar] [CrossRef] [Green Version]
  77. Hoss, U.; Budiman, E.S.; Liu, H.; Christiansen, M.P. Continuous glucose monitoring in the subcutaneous tissue over a 14-day sensor wear period. J. Diabetes Sci. Technol. 2013, 7, 1210–1219. [Google Scholar] [CrossRef] [Green Version]
  78. Borazan, A.; Binici, D.N. Relationship between insulin resistance and inflammation markers in hemodialysis patients. Ren. Fail. 2010, 32, 198–202. [Google Scholar] [CrossRef]
  79. Gerritsen, M.; Jansen, J.A.; Kros, A.; Vriezema, D.M.; Sommerdijk, N.A.J.M.; Nolte, R.J.M.; Lutterman, J.A.; Van Hvell, S.W.F.M.; Van Der Gaag, A. Influence of inflammatory cells and serum on the performance of implantable glucose sensors. J. Biomed. Mater. Res. 2001, 54, 69–75. [Google Scholar] [CrossRef]
  80. Clark, L.C.; Spokane, R.B.; Homan, M.M.; Sudan, R.; Miller, M. Long-term stability of electroenzymatic glucose sensors implanted in mice. An update. ASAIO Trans. 1988, 34, 259–265. [Google Scholar]
  81. Thippeswamy, B.; Naveenkumar, K.J.; Bodharthi, J.G.; Shivaprasad, S.R. Antimicrobial activity of ethanolic extract of Usnea longissima. J. Exp. Sci. 2011, 2, 1–3. [Google Scholar]
  82. Alsaleh, F.M.; Smith, F.J.; Keady, S.; Taylor, K.M.G. Insulin pumps: From inception to the present and toward the future. J. Clin. Pharm. Ther. 2010, 35, 127–138. [Google Scholar] [CrossRef] [PubMed]
  83. Benkhadra, K.; Alahdab, F.; Tamhane, S.U.; McCoy, R.G.; Prokop, L.J.; Murad, M.H. Continuous subcutaneous insulin infusion versus multiple daily injections in individuals with type 1 diabetes: A systematic review and meta-analysis. Endocrine 2017, 55, 77–84. [Google Scholar] [CrossRef] [PubMed]
  84. Pros and Cons of Insulin Pumps. Available online: https://www.diabetes.co.uk/insulin-pumps (accessed on 5 March 2023).
  85. Zeidan, T.; Nikkel, C.; Dziengelewski, B.; Wu, S.; Chen, A.M.H. Clinical Evaluation of Basal-Bolus Therapy Delivered by the V-Go® Wearable Insulin Delivery Device in Patients with Type 2 Diabetes: A Retrospective Analysis. Pharmacy 2020, 8, 215. [Google Scholar] [CrossRef]
  86. Kulzer, B.; Freckmann, G.; Heinemann, L.; Schnell, O.; Hinzmann, R.; Ziegler, R. Diabetes Research and Clinical Practice Patch Pumps: What are the advantages for people with diabetes ? Diabetes Res. Clin. Pract. 2022, 187, 109858. [Google Scholar] [CrossRef]
  87. Thompson, B.; Cook, C.B. Insulin Pumping Patches: Emerging Insulin Delivery Systems. J. Diabetes Sci. Technol. 2019, 13, 8–10. [Google Scholar] [CrossRef] [Green Version]
  88. Berget, C.; Messer, L.H.; Forlenza, G.P. A clinical overview of insulin pump therapy for the management of diabetes: Past, present, and future of intensive therapy. Diabetes Spectr. 2019, 32, 194–204. [Google Scholar] [CrossRef] [PubMed]
  89. Pohar, S.L. Subcutaneous open-loop insulin delivery for type 1 diabetes: Paradigm Real-Time System. Issues Emerg. Health Technol. 2007, 105, 1–6. [Google Scholar]
  90. Danne, T.; Kordonouri, O.; Holder, M.; Haberland, H.; Golembowski, S.; Remus, K.; Bläsig, S.; Wadien, T.; Zierow, S.; Hartmann, R.; et al. Prevention of Hypoglycemia by using low glucose suspend function in sensor-augmented pump therapy. Diabetes Technol. Ther. 2011, 13, 1129–1134. [Google Scholar] [CrossRef]
  91. Bergenstal, R.M.; Klonoff, D.C.; Garg, S.K.; Bode, B.W.; Meredith, M.; Slover, R.H.; Ahmann, A.J.; Welsh, J.B.; Lee, S.W.; Kaufman, F.R. Threshold-Based Insulin-Pump Interruption for Reduction of Hypoglycemia. N. Engl. J. Med. 2013, 369, 224–232. [Google Scholar] [CrossRef] [Green Version]
  92. Abraham, M.B.; Nicholas, J.A.; Smith, G.J.; Fairchild, J.M.; King, B.R.; Ambler, G.R.; Cameron, F.J.; Davis, E.A.; Jones, T.W. Reduction in Hypoglycemia with the predictive low-Glucose management system: A long-term randomized controlled trial in adolescents with type 1 diabetes. Diabetes Care 2018, 41, 303–310. [Google Scholar] [CrossRef] [Green Version]
  93. Forlenza, G.P.; Li, Z.; Buckingham, B.A.; Pinsker, J.E.; Cengiz, E.; Paul Wadwa, R.; Ekhlaspour, L.; Church, M.M.; Weinzimer, S.A.; Jost, E.; et al. Predictive low-glucose suspend reduces Hypoglycemia in adults, adolescents, and children with type 1 diabetes in an at-home randomized crossover study: Results of the PROLOG trial. Diabetes Care 2018, 41, 2155–2161. [Google Scholar] [CrossRef] [Green Version]
  94. Boughton, C.K.; Hovorka, R. New closed-loop insulin systems. Diabetologia 2021, 64, 1007–1015. [Google Scholar] [CrossRef]
  95. Chen, C.; Zhao, X.L.; Li, Z.H.; Zhu, Z.G.; Qian, S.H.; Flewitt, A.J. Current and emerging technology for continuous glucose monitoring. Sensors 2017, 17, 182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. El Youssef, J.; Castle, J.; Ward, W.K. A review of closed-loop algorithms for glycemic control in the treatment of type 1 diabetes. Algorithms 2009, 2, 518–532. [Google Scholar] [CrossRef] [Green Version]
  97. Quiroz, G. The evolution of control algorithms in artificial pancreas: A historical perspective. Annu. Rev. Control 2019, 48, 222–232. [Google Scholar] [CrossRef]
  98. Vigersky, R.A.; Huang, S.; Cordero, T.L.; Shin, J.; Lee, S.W.; Chhabra, H.; Kaufman, F.R.; Cohen, O. Improved HBA1C, total daily insulin dose, and treatment satisfaction with insulin pump therapy compared to multiple daily insulin injections in patients with type 2 diabetes irrespective of baseline C-peptide levels. Endocr. Pract. 2018, 24, 446–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Abuin, P.; Rivadeneira, P.S.; Ferramosca, A.; González, A.H. Artificial pancreas under stable pulsatile MPC: Improving the closed-loop performance. J. Process Control 2020, 92, 246–260. [Google Scholar] [CrossRef]
  100. Incremona, G.P.; Messori, M.; Toffanin, C.; Cobelli, C.; Magni, L. Model predictive control with integral action for artificial pancreas. Control Eng. Pract. 2018, 77, 86–94. [Google Scholar] [CrossRef]
  101. Karpelyev, V.A.; Philippov, Y.I.; Averin, A.V.; Boyarskiy, M.D.; Gavrilov, D.A. Development and in silico validation of the pid-algorithm for the artificial pancreas with intraperitoneal insulin delivery. Diabetes Mellit. 2018, 21, 58–65. [Google Scholar] [CrossRef]
  102. Chakrabarty, A.; Gregory, J.M.; Moore, L.M.; Williams, P.E.; Farmer, B.; Cherrington, A.D.; Lord, P.; Shelton, B.; Cohen, D.; Zisser, H.C.; et al. A new animal model of insulin-glucose dynamics in the intraperitoneal space enhances closed-loop control performance. J. Process Control 2019, 76, 62–73. [Google Scholar] [CrossRef]
  103. Soylu, S.; Danisman, K. In silico testing of optimized Fuzzy P+D controller for artificial pancreas. Biocybern. Biomed. Eng. 2018, 38, 399–408. [Google Scholar] [CrossRef]
  104. Mehmood, S.; Ahmad, I.; Arif, H.; Ammara, U.E.; Majeed, A. Artificial pancreas control strategies used for type 1 diabetes control and treatment: A comprehensive analysis. Appl. Syst. Innov. 2020, 3, 31. [Google Scholar] [CrossRef]
  105. Peyser, T.; Dassau, E.; Breton, M.; Skyler, J.S. The artificial pancreas: Current status and future prospects in the management of diabetes. Ann. N. Y. Acad. Sci. 2014, 1311, 102–123. [Google Scholar] [CrossRef]
  106. Janez, A.; Battelino, T.; Klupa, T.; Kocsis, G.; Kuricová, M.; Lalić, N.; Stoian, A.P.; Prázný, M.; Rahelić, D.; Šoupal, J.; et al. Hybrid Closed-Loop Systems for the Treatment of Type 1 Diabetes: A Collaborative, Expert Group Position Statement for Clinical Use in Central and Eastern Europe. Diabetes Ther. 2021, 12, 107–3135. [Google Scholar] [CrossRef]
  107. De Bock, M.; Dart, J.; Roy, A.; Davey, R.; Soon, W.; Berthold, C.; Retterath, A.; Grosman, B.; Kurtz, N.; Davis, E.; et al. Exploration of the Performance of a Hybrid Closed Loop Insulin Delivery Algorithm That Includes Insulin Delivery Limits Designed to Protect Against Hypoglycemia. J. Diabetes Sci. Technol. 2017, 11, 68–73. [Google Scholar] [CrossRef] [PubMed]
  108. Hybrid Closed-Loop Insulin Delivery Systems for People with Type 1 Diabetes. Available online: https://www.cadth.ca/hybrid-closed-loop-insulin-delivery-systems-people-type-1-diabetes (accessed on 14 March 2023).
  109. Elkhatib, F.; Buckingham, B.A.; Buse, J.B.; Harlan, D.M.; Magyar, K.; Ly, T.T.; Kirkman, M.S.; Malkani, S.; Thompson, M.J.; Lock, J.P.; et al. Home use of a bihormonal bionic pancreas vs. Conventional insulin pump therapy in adults with type 1 diabetes: A multicenter, randomized clinical trial. Diabetes 2016, 65, 369–380. [Google Scholar]
  110. El-Khatib, F.H.; Russell, S.J.; Magyar, K.L.; Sinha, M.; McKeon, K.; Nathan, D.M.; Damiano, E.R. Autonomous and continuous adaptation of a bihormonal bionic pancreas in adults and adolescents with type 1 diabetes. J. Clin. Endocrinol. Metab. 2014, 99, 1701–1711. [Google Scholar] [CrossRef] [Green Version]
  111. Rayannavar, A.; Mitteer, L.M.; Balliro, C.A.; El-Khatib, F.H.; Lord, K.L.; Hawkes, C.P.; Ballester, L.S.; Damiano, E.R.; Russell, S.J.; De Leon, D.D. The bihormonal bionic pancreas improves glycemic control in individuals with hyperinsulinism and post pancreatectomy diabetes: A pilot study. Diabetes Care 2021, 44, 2582–2585. [Google Scholar] [CrossRef]
  112. Beck, R.W.; Russell, S.J.; Damiano, E.R.; El-Khatib, F.H.; Ruedy, K.J.; Balliro, C.; Li, Z.; Calhoun, P. A Multicenter Randomized Trial Evaluating Fast-Acting Insulin Aspart in the Bionic Pancreas in Adults with Type 1 Diabetes. Diabetes Technol. Ther. 2022, 24, 681–696. [Google Scholar] [PubMed]
  113. Wszola, M.; Klak, M.; Kosowska, A.; Olkowska-Truchanowicz, J.; Tymicki, G.; Berman, A.; Bryniarski, T.; Kołodziejska, M.; Uchrynowska-Tyszkiewicz, I.; Kamiński, A. Bionic pancreas: The first results of functionality bionic tissue model with pancreatic islets. Korean J. Transplant. 2021, 35, S44. [Google Scholar] [CrossRef]
  114. Russell, S.; Balliro, C.; Sherwood, J.; Jafri, R.; Hillard, M.; Sullivan, M.; Greaux, E.; Selagamsetty, R.; El-Khatib, F.; Damiano, E. Home use of the iLet bionic pancreas in the bihormonal configuration using dasiglucagon versus the insulin-only configuration in adults with type 1 diabetes. Diabetes Technol. Ther. 2020, 22, S97. [Google Scholar]
  115. NCT03565666. The Insulin-Only Bionic Pancreas Bridging Study. 2018. Available online: https://clinicaltrials.gov/show/NCT03565666 (accessed on 10 February 2023).
  116. Forlenza, G.P.; Messer, L.H.; Berget, C.; Wadwa, R.P.; Driscoll, K.A. Biopsychosocial Factors Associated with Satisfaction and Sustained Use of Artificial Pancreas Technology and Its Components: A Call to the Technology Field. Curr. Diab. Rep. 2018, 18, 114. [Google Scholar] [CrossRef] [PubMed]
  117. Pérez-Gandía, C.; García-Sáez, G.; Subías, D.; Rodríguez-Herrero, A.; Gómez, E.J.; Rigla, M.; Hernando, M.E. Decision Support in Diabetes Care: The Challenge of Supporting Patients in Their Daily Living Using a Mobile Glucose Predictor. J. Diabetes Sci. Technol. 2018, 12, 243–250. [Google Scholar] [CrossRef] [PubMed]
  118. Tyler, N.S.; Jacobs, P.G. Artificial intelligence in decision support systems for type 1 diabetes. Sensors 2020, 20, 3214. [Google Scholar] [CrossRef]
  119. Fisher, L.; Polonsky, W.H.; Hessler, D.M.; Masharani, U.; Blumer, I.; Peters, A.L.; Strycker, L.A.; Bowyer, V. Understanding the sources of diabetes distress in adults with type 1 diabetes. J. Diabetes Complicat. 2015, 29, 572–577. [Google Scholar] [CrossRef] [Green Version]
  120. Polonsky, W.H.; Fisher, L.; Hessler, D.; Edelman, S.V. Development of a New Measure for Assessing Glucose Monitoring Device-Related Treatment Satisfaction and Quality of Life. Diabetes Technol. Ther. 2015, 17, 657–663. [Google Scholar] [CrossRef]
  121. Polonsky, W.H.; Fisher, L.; Hessler, D.; Edelman, S.V. Development of a New Measure for Assessing Insulin Delivery Device Satisfaction in Patients with Type 1 and Type 2 Diabetes. Diabetes Technol. Ther. 2015, 17, 773–779. [Google Scholar] [CrossRef]
  122. Joensen, L.E.; Andersen, M.M.; Jensen, S.; Nørgaard, K.; Willaing, I. The effect of peer support in adults with insulin pump-treated type 1 diabetes: A pilot study of a flexible and participatory intervention. Patient Prefer. Adherence 2017, 11, 1879–1890. [Google Scholar] [CrossRef] [Green Version]
  123. Rizvi, A. IDF21-0043 Technology Trio: Continuous Glucose Monitoring, Telehealth, and Electronic Messaging for Insulin-Treated Patients. Diabetes Res. Clin. Pract. 2022, 186, 109605. [Google Scholar] [CrossRef]
  124. Elian, V.; Musat, M.; Radulian, G.; Negoita, O. A model of efficiency on introducing technology in T1DM management process for patients with 30+year’s duration of diabetes-a single-center interventional study. Diabetes Technol. Ther. 2020, 22, A225. [Google Scholar]
  125. Fanzola, V.; Riboni, S.; Cannalire, G.; Metti, M.; Bensi, G.; Granata, C.; Biasucci, G. The impact of new continuous glucose monitoring (CGM) devices versus self-management of blood glucose (SMBG) on the daily life of parents and children affected by type 1 diabetes mellitus. J. Pediatr. Neonatal Individ. Med. 2022, 11, e110111. [Google Scholar]
  126. Rusak, E.; Ogarek, N.; Wolicka, K.; Mrówka, A.; Seget, S.; Kuźnik, M.; Jarosz-Chobot, P. The quality of life and satisfaction with continuous glucose monitoring therapy in children under 7 years of age with T1D using the rTCGM system integrated with insulin pump—A caregivers point of view. Sensors 2021, 21, 3683. [Google Scholar] [CrossRef] [PubMed]
  127. Polonsky, W.H.; Hessler, D.; Ruedy, K.J.; Beck, R.W. The impact of continuous glucose monitoring on markers of quality of life in adults with type 1 diabetes: Further findings from the DIAMOND randomized clinical trial. Diabetes Care 2017, 40, 736–741. [Google Scholar] [CrossRef] [Green Version]
  128. Brazeau, A.S.; Rabasa-Lhoret, R.; Strychar, I.; Mircescu, H. Barriers to physical activity among patients with type 1 diabetes. Diabetes Care 2008, 31, 2108–2109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Ferreira, M.; Neves, J.S.; Neves, C.; Carvalho, D. Physical exercise and glycemic management in patients with type 1 diabetes on insulin pump therapy—A cross-sectional study. Acta Diabetol. 2023, 60, 881–889. [Google Scholar] [CrossRef] [PubMed]
  130. Chetty, T.; Shetty, V.; Fournier, P.A.; Adolfsson, P.; Jones, T.W.; Davis, E.A. Exercise management for young people with type 1 diabetes: A structured approach to the exercise consultation. Front. Endocrinol. 2019, 10, 326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Weissberg-Benchell, J.; Hessler, D.; Polonsky, W.H.; Fisher, L. Psychosocial Impact of the Bionic Pancreas during Summer Camp. J. Diabetes Sci. Technol. 2016, 10, 840–844. [Google Scholar] [CrossRef] [Green Version]
  132. Vidal Florc, M.; Jansà Morató, M.; Galindo Rubio, M.; Penalba Martínez, M. Factors associated to adherence to blood glucose self-monitoring in patients with diabetes treated with insulin. The dapa study. Endocrinol. Diabetes Nutr. 2018, 65, 99–106. [Google Scholar] [CrossRef]
  133. Gómez-Peralta, F.; Menéndez, E.; Conde, S.; Conget, I.; Novials, A. Clinical characteristics and management of type 1 diabetes in Spain. The SED1 study. Endocrinol. Diabetes Nutr. 2021, 68, 642–653. [Google Scholar] [CrossRef]
  134. Spaans, E.A.J.M.; Kleefstra, N.; Groenier, K.H.; Bilo, H.J.G.; Brand, P.L.P. Adherence to insulin pump treatment declines with increasing age in adolescents with type 1 diabetes mellitus. Acta Paediatr. Int. J. Paediatr. 2020, 109, 134–139. [Google Scholar] [CrossRef]
  135. Giani, E.; Snelgrove, R.; Volkening, L.K.; Laffel, L.M. Continuous Glucose Monitoring (CGM) Adherence in Youth with Type 1 Diabetes: Associations with Biomedical and Psychosocial Variables. J. Diabetes Sci. Technol. 2017, 11, 476–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Trandafir, L.M.; Moisa, S.M.; Vlaiculescu, M.V.; Butnariu, L.I.; Boca, L.O.; Constantin, M.M.L.; Lupu, P.M.; Brinza, C.; Temneanu, O.R.; Burlacu, A. Insulin Pump Therapy Efficacy and Key Factors Influencing Adherence in Pediatric Population-A Narrative Review. Medicina 2022, 58, 1671. [Google Scholar] [CrossRef] [PubMed]
  137. Patton, S.R.; Clements, M.A. Psychological Reactions Associated with Continuous Glucose Monitoring in Youth. J. Diabetes Sci. Technol. 2016, 10, 656–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Markowitz, J.T.; Pratt, K.; Aggarwal, J.; Volkening, L.K.; Laffel, L.M.B. Psychosocial correlates of continuous glucose monitoring use in youth and adults with type 1 diabetes and parents of youth. Diabetes Technol. Ther. 2012, 14, 523–526. [Google Scholar] [CrossRef] [Green Version]
  139. Landau, Z.; Rachmiel, M.; Pinhas-Hamiel, O.; Boaz, M.; Bar-Dayan, Y.; Wainstein, J. TR Parental sleep quality and continuous glucose monitoring system use in children with type 1 diabetes. Diabetes Technol. Ther. 2015, 17, S18–S19. [Google Scholar]
  140. De Bock, M.; Cooper, M.; Retterath, A.; Nicholas, J.; Ly, T.; Jones, T.; Davis, E. Continuous Glucose Monitoring Adherence: Lessons from a Clinical Trial to Predict Outpatient Behavior. J. Diabetes Sci. Technol. 2016, 10, 627–632. [Google Scholar] [CrossRef] [Green Version]
  141. Tanenbaum, M.L.; Adams, R.N.; Hanes, S.J.; Barley, R.C.; Miller, K.M.; Mulvaney, S.A.; Hood, K.K. Optimal Use of Diabetes Devices: Clinician Perspectives on Barriers and Adherence to Device Use. J. Diabetes Sci. Technol. 2017, 11, 484–492. [Google Scholar] [CrossRef] [Green Version]
  142. Breton, M.D.; Patek, S.D.; Lv, D.; Schertz, E.; Robic, J.; Pinnata, J.; Kollar, L.; Barnett, C.; Wakeman, C.; Oliveri, M.; et al. Continuous Glucose Monitoring and Insulin Informed Advisory System with Automated Titration and Dosing of Insulin Reduces Glucose Variability in Type 1 Diabetes Mellitus. Diabetes Technol. Ther. 2018, 20, 531–540. [Google Scholar] [CrossRef]
  143. Castle, J.R.; El Youssef, J.; Wilson, L.M.; Reddy, R.; Resalat, N.; Branigan, D.; Ramsey, K.; Leitschuh, J.; Rajhbeharrysingh, U.; Senf, B.; et al. Randomized Outpatient Trial of Single- and Dual-Hormone Closed-Loop Systems That Adapt to Exercise Using Wearable Sensors. Diabetes Care 2018, 41, 1471–1477. [Google Scholar] [CrossRef] [Green Version]
  144. Kordonouri, O.; Riddell, M.C. Use of apps for physical activity in type 1 diabetes: Current status and requirements for future development. Ther. Adv. Endocrinol. Metab. 2019, 10, 2042018819839298. [Google Scholar] [CrossRef]
  145. Buoite Stella, A.; Assaloni, R.; Tonutti, L.; Manca, E.; Tortul, C.; Candido, R.; Francescato, M.P. Strategies used by Patients with Type 1 Diabetes to Avoid Hypoglycemia in a 24 × 1-Hour Marathon: Comparison with the Amounts of Carbohydrates Estimated by a Customizable Algorithm. Can. J. Diabetes 2017, 41, 184–189. [Google Scholar] [CrossRef]
  146. Wang, M.; Wang, C.; Ren, S.; Pan, J.; Wang, Y.; Shen, Y.; Zeng, Z.; Cui, H.; Zhao, X. Versatile Oral Insulin Delivery Nanosystems: From Materials to Nanostructures. Int. J. Mol. Sci. 2022, 23, 3362. [Google Scholar] [CrossRef]
  147. Mahameed, M.; Xue, S.; Stefanov, B.; Hamri, G.C.; Fussenegger, M. Engineering a Rapid Insulin Release System Controlled by Oral Drug Administration. Adv. Sci. 2022, 9, 2105619. [Google Scholar] [CrossRef]
  148. Martínez-López, A.L.; Carvajal-Millan, E.; Canett-Romero, R.; Prakash, S.; Rascón-Chu, A.; López-Franco, Y.L.; Lizardi-Mendoza, J.; Micard, V. Arabinoxylans-Based Oral Insulin Delivery System Targeting the Colon: Simulation in a Human Intestinal Microbial Ecosystem and Evaluation in Diabetic Rats. Pharmaceuticals 2022, 15, 1062. [Google Scholar] [CrossRef]
  149. Liu, G.; He, S.; Ding, Y.; Chen, C.; Cai, Q.; Zhou, W. Multivesicular liposomes for glucose-responsive insulin delivery. Pharmaceutics 2022, 14, 21. [Google Scholar] [CrossRef]
  150. Sarhadi, S.; Moosavian, S.A.; Mashreghi, M.; Rahiman, N.; Golmohamadzadeh, S.; Tafaghodi, M.; Sadri, K.; Chamani, J.; Jaafari, M.R. B12-functionalized PEGylated liposomes for the oral delivery of insulin: In vitro and in vivo studies. J. Drug Deliv. Sci. Technol. 2022, 69, 103141. [Google Scholar] [CrossRef]
  151. Bahman, F.; Taurin, S.; Altayeb, D.; Taha, S.; Bakhiet, M.; Greish, K. Oral insulin delivery using poly (Styrene co-Maleic acid) micelles in a diabetic mouse model. Pharmaceutics 2020, 12, 1026. [Google Scholar] [CrossRef] [PubMed]
  152. Han, X.; Lu, Y.; Xie, J.; Zhang, E.; Zhu, H.; Du, H.; Wang, K.; Song, B.; Yang, C.; Shi, Y.; et al. Zwitterionic micelles efficiently deliver oral insulin without opening tight junctions. Nat. Nanotechnol. 2020, 15, 605–614. [Google Scholar] [CrossRef] [PubMed]
  153. Wu, H.; Guo, T.; Nan, J.; Yang, L.; Liao, G.; Park, H.J.; Li, J. Hyaluronic-Acid-Coated Chitosan Nanoparticles for Insulin Oral Delivery: Fabrication, Characterization, and Hypoglycemic Ability. Macromol. Biosci. 2022, 22, 2100493. [Google Scholar] [CrossRef] [PubMed]
  154. Li, J.; Qiang, H.; Yang, W.; Xu, Y.; Feng, T.; Cai, H.; Wang, S.; Liu, Z.; Zhang, Z.; Zhang, J. Oral insulin delivery by epithelium microenvironment-adaptive nanoparticles. J. Control. Release 2022, 341, 31–43. [Google Scholar] [CrossRef] [PubMed]
  155. Elkhatib, M.M.; Ali, A.I.; Al-Badrawy, A.S. In vitro and in vivo comparative study of oral nanoparticles and gut iontophoresis as oral delivery systems for insulin. Biol. Pharm. Bull. 2021, 44, 251–258. [Google Scholar] [CrossRef] [PubMed]
  156. Banerjee, A.; Wong, J.; Gogoi, R.; Brown, T.; Mitragotri, S. Intestinal micropatches for oral insulin delivery. J. Drug Target. 2017, 25, 608–615. [Google Scholar] [CrossRef] [PubMed]
  157. Torabi, N.; Nazari, M.; Fahanik-Babaei, J.; Eliassi, A. Long-term administration of intranasal insulin improves peripheral glucose concentration in diabetic male rats. Physiol. Pharmacol. 2020, 24, 268–275. [Google Scholar] [CrossRef]
  158. Roque, P.; Nakadate, Y.; Sato, H.; Sato, T.; Wykes, L.; Kawakami, A.; Yokomichi, H.; Matsukawa, T.; Schricker, T. Intranasal administration of 40 and 80 units of insulin does not cause hypoglycemia during cardiac surgery: A randomized controlled trial. Can. J. Anesth. 2021, 68, 991–999. [Google Scholar] [CrossRef] [PubMed]
  159. Zhu, Y.; Huang, Y.; Yang, J.; Tu, R.; Zhang, X.; He, W.W.; Hou, C.Y.; Wang, X.M.; Yu, J.M.; Jiang, G.H. Intranasal insulin ameliorates neurological impairment after intracerebral hemorrhage in mice. Neural Regen. Res. 2022, 17, 210–216. [Google Scholar]
  160. Jin, X.; Zhu, D.D.; Chen, B.Z.; Ashfaq, M.; Guo, X.D. Insulin delivery systems combined with microneedle technology. Adv. Drug Deliv. Rev. 2018, 127, 119–137. [Google Scholar] [CrossRef]
  161. Chen, S.; Miyazaki, T.; Itoh, M.; Matsumoto, H.; Moro-Oka, Y.; Tanaka, M.; Miyahara, Y.; Suganami, T.; Matsumoto, A. Temperature-Stable Boronate Gel-Based Microneedle Technology for Self-Regulated Insulin Delivery. ACS Appl. Polym. Mater. 2020, 2, 2781–2790. [Google Scholar] [CrossRef]
  162. Cui, C.Y.; Lu, W.L.; Xiao, L.; Zhang, S.Q.; Huang, Y.B.; Li, S.L.; Zhang, R.J.; Wang, G.L.; Zhang, X.; Zhang, Q. Sublingual delivery of insulin: Effects of enhancers on the mucosal lipid fluidity and protein conformation, transport, and in Vivo hypoglycemic activity. Biol. Pharm. Bull. 2005, 28, 2279–2288. [Google Scholar] [CrossRef] [Green Version]
  163. Erzengin, S.; Guler, E.; Eser, E.; Polat, E.B.; Gunduz, O.; Cam, M.E. In vitro and in vivo evaluation of 3D printed sodium alginate/polyethylene glycol scaffolds for sublingual delivery of insulin: Preparation, characterization, and pharmacokinetics. Int. J. Biol. Macromol. 2022, 204, 429–440. [Google Scholar] [CrossRef]
  164. Volpatti, L.R.; Matranga, M.A.; Cortinas, A.B.; Delcassian, D.; Daniel, K.B.; Langer, R.; Anderson, D.G. Glucose-Responsive Nanoparticles for Rapid and Extended Self-Regulated Insulin Delivery. ACS Nano 2020, 14, 488–497. [Google Scholar] [CrossRef] [PubMed]
  165. Yin, R.; He, J.; Bai, M.; Huang, C.; Wang, K.; Zhang, H.; Yang, S.M.; Zhang, W. Engineering synthetic artificial pancreas using chitosan hydrogels integrated with glucose-responsive microspheres for insulin delivery. Mater. Sci. Eng. C 2019, 96, 374–382. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, X. Bioartificial Organ Manufacturing Technologies. Cell Transplant. 2019, 28, 5–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Parvaneh, S.; Kemény, L.; Ghaffarinia, A.; Yarani, R.; Veréb, Z. Three-dimensional bioprinting of functional β-islet-like constructs. Int. J. Bioprinting 2022, 9, 256–279. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Diabetes distress, a schematic presentation; adapted from [33,35].
Figure 1. Diabetes distress, a schematic presentation; adapted from [33,35].
Life 13 01663 g001
Figure 2. Overview of the modern non-invasive CGM approaches for intensive insulin therapy, adapted from [57].
Figure 2. Overview of the modern non-invasive CGM approaches for intensive insulin therapy, adapted from [57].
Life 13 01663 g002
Figure 3. V-Go, a 24 h wearable insulin PP. Reproduction with permission from [85].
Figure 3. V-Go, a 24 h wearable insulin PP. Reproduction with permission from [85].
Life 13 01663 g003
Figure 4. (A) Closed-loop glycemic management system using the “Sense and Act” method for optimal insulin delivery; reproduction with permission from [95]; (B) Schematic representation of closed-loop insulin delivery, adapted from [94]. A CGM wirelessly transmits the information related to interstitial glucose concentration (to a smartphone/insulin pump); the algorithm translates the received data and calculates the appropriate amount of insulin. Then, a rapid-acting insulin analog is delivered by an insulin pump. Insulin infusion is regulated in real time by the control algorithm. CSII—continuous subcutaneous insulin infusion; ISF—interstitial fluid.
Figure 4. (A) Closed-loop glycemic management system using the “Sense and Act” method for optimal insulin delivery; reproduction with permission from [95]; (B) Schematic representation of closed-loop insulin delivery, adapted from [94]. A CGM wirelessly transmits the information related to interstitial glucose concentration (to a smartphone/insulin pump); the algorithm translates the received data and calculates the appropriate amount of insulin. Then, a rapid-acting insulin analog is delivered by an insulin pump. Insulin infusion is regulated in real time by the control algorithm. CSII—continuous subcutaneous insulin infusion; ISF—interstitial fluid.
Life 13 01663 g004
Figure 5. Control system principles applied to glycemic control; reproduction with permission from [96].
Figure 5. Control system principles applied to glycemic control; reproduction with permission from [96].
Life 13 01663 g005
Figure 6. Decision support system (DSS) objectives; adapted from [118].
Figure 6. Decision support system (DSS) objectives; adapted from [118].
Life 13 01663 g006
Figure 7. The principal sources of diabetes distress in T1DM adults, adapted from [119].
Figure 7. The principal sources of diabetes distress in T1DM adults, adapted from [119].
Life 13 01663 g007
Figure 8. Factors negatively impacting insulin pump adherence in pediatric patients, adapted from [136]. Reproduction with permission.
Figure 8. Factors negatively impacting insulin pump adherence in pediatric patients, adapted from [136]. Reproduction with permission.
Life 13 01663 g008
Table 1. Comparison between various commercial CGM systems.
Table 1. Comparison between various commercial CGM systems.
CharacteristicsDexcom G7Guardian 4Libre 3Eversense E3
Manufacturer [60]DexCom, Inc., San Diego, CA, USAMedtronic, Minneapolis, MN, USAAbbott Laboratories, Chicago, IL, USASenseonics Holdings, Germantown, MD, USA
FDA approvalDecember 20222021April 2022February 2022
UsersAdults and children > 2 yearsAdults and children > 7 yearsAdults and children ≥ 4 years
Pregnant women
Adults over
18 years
Days of sensor wear (RT)10
+12 h grace period
714180
Sensing moleculeGlucose oxidaseGlucose oxidaseGlucose oxidaseBoronic-acid derivative
Technique
category [60]
ElectrochemicalElectrochemicalElectrochemicalOptique
Components [60]Sensor, Transmitter, appSensor, Transmitter,
app
Sensor, appSensor, Transmitter, app, insertion tool
Sensor size (mm)
 [60]
24 × 27.3 × 4.638 × 67 × 52 mm21 × 2.9 × 0.11 mm18.3 × 3.5 subcutaneous
37.6 × 48 × 8.8 transmitter
Approved areas of insertion [60]All pts—abdomen and upper arm
2–6 years—also upper buttocks
7–17 years—upper arm, upper buttocks,
Over 18 years—abdomen, upper arm
Back of
upper arm
Upper arm
Accuracy
(MARD%) [60]
8.7%10.8%7.8%8.5%
Daily calibration
frequency (×)
 [60]
0
(factory
calibration)
00
(factory
calibration)
2
(at 12 h)
Warm-up
(min)
301206060
High and low
alarms
YesYesYesYes
High and low
prediction
YesYesYesYes
Integration with an
insulin pump [60]
Tandem t:slim Control-IQMedtronic MiniMed 770G, 780GNoNo
Smartphone
integration
Android, iOs,
Apple Watch
Android,
iOs
Android,
iOs
Android, iOs,
Apple Watch
Distance to phone
(m)
6610-
Operating temperature (°C)10–42 °C0–45 °C10–45 °C5–40 °C
Data sharing≤10 people≤5 people≤20 people≤5 people
A separate receiver is availableYesNoYesNo
Water resistance
depth (m)/time (min, hours)
2.4 m, ≥ 24 h2.5 m, 10 min1 m, 30 min1 m, 30 min
Skin
complications
YesYesYesYes
Interference with drugsHydroxyureaAcetaminophenVitamin C-
MARD—mean absolute relative difference; CGM—continuous glucose monitoring; data adapted from https://www.dexcom.com/en-GB/downloadsandguides/search (accessed on 7 March 2023), https://www.medtronicdiabetes.com/download-library/guardian-4-sensor-transmitter (accessed on 9 March 2023), and https://www.diabetescare.abbott/support/manuals/uk.html (accessed on 9 March 2023).
Table 2. Comparison between various commercially available CSII pumps.
Table 2. Comparison between various commercially available CSII pumps.
AccuChek Spirit ComboMedtronic Paradigm 522/722Medtronic 720GOmipod Patch PumpCellnovo Insulin PumpDana
Diabecare R
ProducerRoche PharmaMedtronicMedtronicInsulet CorporationCellnovoSooil
Weight (g)80100105 3051
Dimensions (mm)80 × 56 × 2051 × 79 × 2096 × 53 × 25Pod: 41 × 61 × 18
PDA: 66 × 110 × 26
NA54 × 75 × 19
Insulin Volume per Infusion Set (mL)315176-300300200150300
Basal Increments (units)0.10.050.0250.050.050.1
Basal Delivery minim (units)3103333
Bolus Increments minim (units)0.1, 0.2, 0.5, 1,20.10.0250.050.050.1–87
Basal Rates/24 h (units)244848482448
Basal Profiles (units)5387204
Bolus Calculator On separate device Yes Yes Yes Yes Yes
Multiple Bolus-type Options Yes Yes Yes Yes Yes Yes
NA = not available.
Table 3. Benefits and limitations of CSII pumps, adapted from [84].
Table 3. Benefits and limitations of CSII pumps, adapted from [84].
Insulin Pump Therapy BenefitsInsulin Pump Therapy Limitations
Better diabetes control.
Fewer injections.
The need to understand the functioning and proper management of the device.
Improved quality of life.High costs, if not covered by the insurance company.
The flexibility of basal insulin delivery during the day and night.
The flexibility of food intake and exercises.
A device that should be worn on the body with tubing that can be caught on objects.
Diminished risk of hypoglycemia.Skin allergies or infections.
Diminished risk of complications.Multiple alerts.
Table 4. Advantages and limitations of PPs, adapted from [86,87].
Table 4. Advantages and limitations of PPs, adapted from [86,87].
AdvantagesLimitations
The devices are tubeless, without request for an insulin infusion systemWaste of insulin when PPs are replaced.
The needle could be automatically inserted; thus, their application could be less painful.The infusion place is poorly visible, and regular inspection is complex.
The needle is not visible.
More convenient than conventional pumps for numerous activities (showering, swimming, sweating, or exercising).
The accuracy of insulin delivery of some PPs is often lower than that of conventional pumps, particularly at low basal doses.
Smaller and lighter than conventional pumps.
PPs can be discreetly carried to various body parts, offering more effortless movement.
Technical properties are often specifically adapted to T1DM individuals’ needs.
Simple education and training are requested for their use.
They have a poor ecological balance due to waste from plastic material and batteries.
Risk of infections.
Diminished price if certain PPs compared to conventional pumps.Higher price than MDI.
IIS—insulin infusion system; PP—patch pump; MDI—multiday injections.
Table 5. Comparison between various commercially available hybrid closed-loop systems [108].
Table 5. Comparison between various commercially available hybrid closed-loop systems [108].
Tandem t:Slim X2 Control IQMedtronic 780GOmnipod 5CamAPS FX
ProducerTandem Diabetes Care,
San Diego, CA, USA
Medtronic plc,
Dublin, Ireland
Insulet Corporation,
Acton, MA, USA
CamDiab Ltd.,
London, UK
PumpTandemMedtronicOmnipodDana, Ypsopump
CGMDexcom G6Guardian 3 and 4Dexcom G6Dexcom G6, Freestyle Libre 3
CGM duration (days)1071010/14
Algorithm typeMPCPIDMPCMPC
Algorithm configurationOn pumpOn pumpOn pumpApp on Android smartphone
Approved for ages (years)6 and above7 and above2 and above1 and above
Weight (g)112105Pod 26
PDA 165
NA
Dimensions (mm)79 × 51 × 1596 × 53 × 25Pod 39 × 52 × 14.5NA
TubelessNoNoYesNo
Insulin volume per infusion set
(mL)
30030085–200300
Minimum basal increments (units)0.0010.0250.05NA
Bolus range (units)0–250–250.05–30
Minimum daily dose (units)10865
Algorithm target (mg/dL)110–160100 or 120110–150104
(80–198)
Meal detectionNoYesNoNo
Data are available in each product’s technical specifications, adapted from https://www.pcdsociety.org/resources/details/diabetes-technology-state-art, accessed on 10 March 2023. NA = not available.
Table 6. Benefits and limitations of closed-loop systems.
Table 6. Benefits and limitations of closed-loop systems.
Closed-Loop Insulin Systems BenefitsClosed-Loop Insulin Systems Limitations
The glucose levels can be continuously monitored. The T1DM patient regularly verifies the devices to ensure that they function correctly.
The control algorithms improve BG control, automatically regulating the amount of insulin. The user must continuously verify the CGM and infusion pump catheter, ensuring they are in a suitable place, and change them when needed.
The system helps the T1DM user avoid emerging events (hypoglycemia and hyperglycemia). The CGM accuracy should be verified, and the CGM sensor must be regularly replaced.
The patient must count the mealtime carbohydrates and enter them into the system.
The control software settings must be verified to ensure that the insulin infusion has a suitable amount.
The extreme BG levels should be regulated if the system is unable.
The pump’s algorithm could not predict exercise.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Elian, V.; Popovici, V.; Ozon, E.-A.; Musuc, A.M.; Fița, A.C.; Rusu, E.; Radulian, G.; Lupuliasa, D. Current Technologies for Managing Type 1 Diabetes Mellitus and Their Impact on Quality of Life—A Narrative Review. Life 2023, 13, 1663. https://doi.org/10.3390/life13081663

AMA Style

Elian V, Popovici V, Ozon E-A, Musuc AM, Fița AC, Rusu E, Radulian G, Lupuliasa D. Current Technologies for Managing Type 1 Diabetes Mellitus and Their Impact on Quality of Life—A Narrative Review. Life. 2023; 13(8):1663. https://doi.org/10.3390/life13081663

Chicago/Turabian Style

Elian, Viviana, Violeta Popovici, Emma-Adriana Ozon, Adina Magdalena Musuc, Ancuța Cătălina Fița, Emilia Rusu, Gabriela Radulian, and Dumitru Lupuliasa. 2023. "Current Technologies for Managing Type 1 Diabetes Mellitus and Their Impact on Quality of Life—A Narrative Review" Life 13, no. 8: 1663. https://doi.org/10.3390/life13081663

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop