Type 1 Diabetes Mellitus

Back

Practice Essentials

Type 1 diabetes is a chronic illness characterized by the body’s inability to produce insulin due to the autoimmune destruction of the beta cells in the pancreas. Although onset frequently occurs in childhood, the disease can also develop in adults.[1]

See Clinical Findings in Diabetes Mellitus, a Critical Images slideshow, to help identify various cutaneous, ophthalmologic, vascular, and neurologic manifestations of DM.

Signs and symptoms

The classic symptoms of type 1 diabetes are as follows:

Other symptoms may include fatigue, nausea, and blurred vision.

The onset of symptomatic disease may be sudden. It is not unusual for patients with type 1 diabetes to present with diabetic ketoacidosis (DKA).

See Clinical Presentation for more detail.

Diagnosis

Diagnostic criteria by the American Diabetes Association (ADA) include the following[2] :

Lab studies

A fingerstick glucose test is appropriate for virtually all patients with diabetes. All fingerstick capillary glucose levels must be confirmed in serum or plasma to make the diagnosis. All other laboratory studies should be selected or omitted on the basis of the individual clinical situation.

An international expert committee appointed by the ADA, the European Association for the Study of Diabetes, and the International Diabetes Association recommended the HbA1c assay for diagnosing type 1 diabetes only when the condition is suspected but the classic symptoms are absent.[3]

Screening

Screening for type 1 diabetes in asymptomatic low-risk individuals is not recommended.[2] However, in patients at high risk (eg, those who have first-degree relatives with type 1 diabetes), it may be appropriate to perform annual screening for anti-islet antibodies before the age of 10 years, along with 1 additional screening during adolescence.[4]

See Workup for more detail.

Management

Glycemic control

The ADA recommends using patient age as one consideration in the establishment of glycemic goals, with different targets for preprandial, bedtime/overnight, and hemoglobin A1c (HbA1c) levels in patients aged 0-6, 6-12, and 13-19 years.[5] Benefits of tight glycemic control include not only continued reductions in the rates of microvascular complications but also significant differences in cardiovascular events and overall mortality.

Self-monitoring

Optimal diabetic control requires frequent self-monitoring of blood glucose levels, which allows rational adjustments in insulin doses. All patients with type 1 diabetes should learn how to self-monitor and record their blood glucose levels with home analyzers and adjust their insulin doses accordingly.

Real-time continuous monitoring of glucose—using continuous glucose monitors (CGMs)—can help patients improve glycemic control.[6, 7] CGMs contain subcutaneous sensors that measure interstitial glucose levels every 1-5 minutes, providing alarms when glucose levels are too high or too low or are rapidly rising or falling.

Insulin therapy

Patients with type 1 diabetes require lifelong insulin therapy. Most require 2 or more injections of insulin daily, with doses adjusted on the basis of self-monitoring of blood glucose levels. Insulin replacement is accomplished by giving a basal insulin and a preprandial (premeal) insulin. The basal insulin is either long-acting (glargine or detemir) or intermediate-acting (NPH). The preprandial insulin is either rapid-acting (lispro, aspart, insulin inhaled, or glulisine) or short-acting (regular).

Common insulin regimens include the following:

Diet and activity

All patients on insulin should have a comprehensive diet plan, created with the help of a professional dietitian, that includes the following:

Exercise is also an important aspect of diabetes management. Patients should be encouraged to exercise regularly.

See Treatment and Medication for more detail.

Background

Type 1 diabetes mellitus (DM) is a multisystem disease with both biochemical and anatomic/structural consequences. It is a chronic disease of carbohydrate, fat, and protein metabolism caused by the lack of insulin, which results from the marked and progressive inability of the pancreas to secrete insulin because of autoimmune destruction of the beta cells.[1] (See Pathophysiology.) (See also Glucose Intolerance.)

Type 1 DM can occur at any age. Although it frequently arises in juveniles, it can also develop in adults. (See Epidemiology.)

Unlike people with type 2 DM, those with type 1 DM usually are not obese and usually present initially with diabetic ketoacidosis (DKA). The distinguishing characteristic of a patient with type 1 DM is that if his or her insulin is withdrawn, ketosis and eventually ketoacidosis develop. Therefore, these patients are dependent on exogenous insulin. (See Presentation.)

Treatment of type 1 DM requires lifelong insulin therapy. A multidisciplinary approach by the physician, nurse, and dietitian, with regular specialist consultation, is needed to control glycemia, as well as to limit the development of its devastating complications and manage such complications when they do occur. (See Treatmentand Medication.)

Despite the differences between type 1 and type 2 DM, the costs of the 2 conditions are often combined. In a study that focused on type 1 alone, Tao et al estimated that in the United States, type 1 DM is responsible for $14.4 billion in medical costs and lost income each year.[8]

Pathophysiology

Type 1 DM is the culmination of lymphocytic infiltration and destruction of insulin-secreting beta cells of the islets of Langerhans in the pancreas. As beta-cell mass declines, insulin secretion decreases until the available insulin no longer is adequate to maintain normal blood glucose levels. After 80-90% of the beta cells are destroyed, hyperglycemia develops and diabetes may be diagnosed. Patients need exogenous insulin to reverse this catabolic condition, prevent ketosis, decrease hyperglucagonemia, and normalize lipid and protein metabolism.

Currently, autoimmunity is considered the major factor in the pathophysiology of type 1 DM. In a genetically susceptible individual, viral infection may stimulate the production of antibodies against a viral protein that trigger an autoimmune response against antigenically similar beta cell molecules.

Approximately 85% of type 1 DM patients have circulating islet cell antibodies, and the majority also have detectable anti-insulin antibodies before receiving insulin therapy. The most commonly found islet cell antibodies are those directed against glutamic acid decarboxylase (GAD), an enzyme found within pancreatic beta cells.

The prevalence of type 1 DM is increased in patients with other autoimmune diseases, such as Graves disease, Hashimoto thyroiditis, and Addison disease. Pilia et al found a higher prevalence of islet cell antibodies (IA2) and anti-GAD antibodies in patients with autoimmune thyroiditis.[9]

A study by Philippe et al used computed tomography (CT) scans, glucagon stimulation test results, and fecal elastase-1 measurements to confirm reduced pancreatic volume in individuals with DM.[10] This finding, which was equally present in both type 1 and type 2 DM, may also explain the associated exocrine dysfunction that occurs in DM.

Polymorphisms of the class II human leukocyte antigen (HLA) genes that encode DR and DQ are the major genetic determinants of type 1 DM. Approximately 95% of patients with type 1 DM have either HLA-DR3 or HLA-DR4. Heterozygotes for those haplotypes are at significantly greater risk for DM than homozygotes. HLA-DQs are also considered specific markers of type 1 DM susceptibility. In contrast, some haplotypes (eg, HLA-DR2) confer strong protection against type 1 DM.[11]

Sensory and autonomic neuropathy

Sensory and autonomic neuropathy in people with diabetes are caused by axonal degeneration and segmental demyelination. Many factors are involved, including the accumulation of sorbitol in peripheral sensory nerves from sustained hyperglycemia. Motor neuropathy and cranial mononeuropathy result from vascular disease in blood vessels supplying nerves.

Angiopathy

Using nailfold video capillaroscopy, Barchetta et al detected a high prevalence of capillary changes in patients with diabetes, particularly those with retinal damage. This reflects a generalized microvessel involvement in both type 1 and type 2 DM.[12]

Microvascular disease causes multiple pathologic complications in people with diabetes. Hyaline arteriosclerosis, a characteristic pattern of wall thickening of small arterioles and capillaries, is widespread and is responsible for ischemic changes in the kidney, retina, brain, and peripheral nerves.

Atherosclerosis of the main renal arteries and their intrarenal branches causes chronic nephron ischemia. It is a significant component of multiple renal lesions in diabetes.

Vitamin D deficiency is an important independent predictor of development of coronary artery calcification in individuals with type 1 DM.[13] Joergensen et al determined that vitamin D deficiency in type 1 diabetes may predict all causes of mortality but not development of microvascular complications.[14]

Nephropathy

In the kidneys, the characteristic wall thickening of small arterioles and capillaries leads to diabetic nephropathy, which is characterized by proteinuria, glomerular hyalinization (Kimmelstiel-Wilson), and chronic renal failure. Exacerbated expression of cytokines such as tumor growth factor beta 1 is part of the pathophysiology of glomerulosclerosis, which begins early in the course of diabetic nephropathy.

Genetic factors influence the development of diabetic nephropathy. Single-nucleotide polymorphisms affecting the factors involved in its pathogenesis appear to influence the risk for diabetic nephropathy in different people with type 1 DM.[15]

Double diabetes

In areas where rates of type 2 DM and obesity are high, individuals with type 1 DM may share genetic and environmental factors that lead to their exhibiting type 2 features such as reduced insulin sensitivity. This condition is termed double diabetes.

In a study that included 207 patients with type 1 DM, Epstein et al used the estimated glucose disposal rate (eGDR) to assess insulin resistance and found that mean eGDR was significantly lower (and, thus, insulin resistance was higher) in black patients (5.66 mg/kg/min) than in either Hispanic patients (6.70 mg/kg/min) or white patients (7.20 mg/kg/min). In addition, low eGDR was associated with an increased risk of vascular complications of diabetes (eg, cardiovascular disease, diabetic retinopathy, or severe chronic kidney disease).[16, 17]

Etiology

Type 1A DM results from autoimmune destruction of the beta cells of the pancreas and involves both genetic predisposition and an environmental component.

Genetic factors

Although the genetic aspect of type 1 DM is complex, with multiple genes involved, there is a high sibling relative risk.[18] Whereas dizygotic twins have a 5-6% concordance rate for type 1 DM,[19] monozygotic twins will share the diagnosis more than 50% of the time by the age of 40 years.[20]

For the child of a parent with type 1 DM, the risk varies according to whether the mother or the father has diabetes. Children whose mother has type 1 DM have a 2-3% risk of developing the disease, whereas those whose father has the disease have a 5-6% risk. When both parents are diabetic, the risk rises to almost 30%. In addition, the risk for children of parents with type 1 DM is slightly higher if onset of the disease occurred before age 11 years and slightly lower if the onset occurred after the parent’s 11th birthday.

The genetic contribution to type 1 DM is also reflected in the significant variance in the frequency of the disease among different ethnic populations. Type 1 DM is most prevalent in European populations, with people from northern Europe more often affected than those from Mediterranean regions.[21] The disease is least prevalent in East Asians.[22]

Genome-wide association studies have identified several loci that are associated with type 1 DM, but few causal relations have been established. The genomic region most strongly associated with other autoimmune diseases, the major histocompatibility complex (MHC), is the location of several susceptibility loci for type 1 DM—in particular, class II HLA DR and DQ haplotypes.[23, 24, 25]

A hierarchy of DR-DQ haplotypes associated with increased risk for type 1 DM has been established. The most susceptible haplotypes are as follows[26] :

Other haplotypes appear to offer protection against type 1 DM. These include the following[26] :

From 90% to 95% of young children with type 1 DM carry HLA-DR3 DQB1*0201, HLA-DR4 DQB1*0302, or both. Carriage of both haplotypes (ie, DR3/4 heterozygotes) confers the highest susceptibility.

These high-risk haplotypes are found primarily in people of European descent; other ethnic groups are less well studied. In African Americans, the DRB1*07:01 - DQA1*03:01 -DQB1*02:01g haplotype is associated with increased risk (OR 3.96), whereas the DRB1*07:01-DQA1*02:01 - DQB1*02:01g haplotype appears to be protective (OR 0.34).[27]

The insulin gene (INS), which encodes for the pre-proinsulin peptide, is adjacent to a variable number of tandem repeats (VNTR) polymorphism at chromosome 11p15.5.[28] Different VNTR alleles may promote either resistance or susceptibility to type 1 DM through their effect on INS transcription in the thymus; for example, protective VNTRs are associated with higher INS expression, which may promote deletion of insulin-specific T cells.[29]

Other genes that have been reported to be involved in the mechanism of type 1 DM include CTLA4 (important in T-cell activation), PTPN22 (produces LYP, a negative regulator of T-cell kinase signaling), and IL2RA (encodes for CD25 which is involved with regulating T-cell function). UBASH3A (also known as STS2), may be involved in the increased risk not only of type 1 DM but also of other autoimmune disease and Down syndrome; it is located on locus chromosome 21q22.3.[30]

In addition, genome-wide association studies have implicated numerous other genes, including the following[31] :

Environmental factors

Extragenetic factors also may contribute. Potential triggers for immunologically mediated destruction of the beta cells include viruses (eg, enterovirus,[32] mumps, rubella, and coxsackievirus B4), toxic chemicals, exposure to cow’s milk in infancy,[33] and cytotoxins.

Combinations of factors may be involved. Lempainen et al found that signs of an enterovirus infection by 12 months of age were associated with the appearance of type 1 DM–related autoimmunity among children who were exposed to cow's milk before 3 months of age. These results suggest an interaction between the 2 factors and provide a possible explanation for the contradictory findings obtained in studies that examined these factors in isolation.[34]

One meta-analysis found a weak but significant linear increase in the risk of childhood type 1 DM with increasing maternal age.[35] However, little evidence supports any substantial increase in childhood type 1 DM risk after pregnancy complicated by preeclampsia.[36]

A study by Simpson et al found that neither vitamin D intake nor 25-hydroxyvitamin D levels throughout childhood were associated with islet autoimmunity or progression to type 1 DM.[37] This study was based in Denver, Colorado, and has been following children at increased risk of diabetes since 1993.

Early upper respiratory infection may also be a risk factor for type 1 diabetes. In an analysis of data on 148 children considered genetically at risk for diabetes, upper respiratory infections in the first year of life were associated with an increased risk for type 1 diabetes .[38, 39] All children in the study who developed islet autoimmunity had at least 2 upper respiratory infections in the first year of life and at least 1 infection within 6 months before islet autoantibody seroconversion.

Children with respiratory infections in the first 6 months of life had the greatest increased hazard ratio (HR) for islet autoantibody seroconversion (HR = 2.27), and the risk was also increased in those with respiratory infections at ages 6 to almost 12 months (HR = 1.32).[38, 39] The rate of islet autoantibody seroconversion was highest among children with more than 5 respiratory infections in the first year of year of life. Respiratory infections in the second year of life were not related to increased risk.[38, 39]

Epidemiology

United States statistics

A 2011 report from the US Centers for Disease Control and Prevention (CDC) estimated that approximately 1 million Americans have type 1 DM.[40] The CDC estimated that each year from 2002 to 2005, type 1 DM was newly diagnosed in 15,600 young people. Among children younger than 10 years, the annual rate of new cases was 19.7 per 100,000 population; among those 10 years or older, the rate was 18.6 per 100,000 population.[40]

Type 1 DM is the most common metabolic disease of childhood. About 1 in every 400-600 children and adolescents has type 1 DM. In adults, type 1 DM constitutes approximately 5% of all diagnosed cases of diabetes.[40]

A study by Mayer-Davis et al indicated that between 2002 and 2012, the incidence of type 1 and type 2 DM saw a significant rise among youths in the United States. According to the report, after the figures were adjusted for age, sex, and race or ethnic group, the incidence of type 1 (in patients aged 0-19 years) and type 2 DM (in patients aged 10-19 years) during this period underwent a relative annual increase of 1.8% and 4.8%, respectively. The greatest increases occurred among minority youths.[41]

International statistics

Internationally, rates of type 1 DM are increasing. In Europe, the Middle East, and Australia, rates of type 1 DM are increasing by 2-5% per year.[42] The prevalence of type 1 DM is highest in Scandinavia (ie, approximately 20% of the total number of people with DM) and lowest in China and Japan (ie, fewer than 1% of all people with diabetes). Some of these differences may relate to definitional issues and the completeness of reporting.

Age-related demographics

Previously referred to as juvenile-onset diabetes, type 1 DM is typically diagnosed in childhood, adolescence, or early adulthood. Although the onset of type 1 DM often occurs early in life, 50% of patients with new-onset type 1 DM are older than 20 years of age.

Type 1 DM usually starts in children aged 4 years or older, appearing fairly abruptly, with the peak incidence of onset at age 11-13 years (ie, in early adolescence and puberty). There is also a relatively high incidence in people in their late 30s and early 40s, in whom the disease tends to present less aggressively (ie, with early hyperglycemia without ketoacidosis and gradual onset of ketosis). This slower-onset adult form of type 1 DM is referred to as latent autoimmune diabetes of the adult (LADA).[40]

A study by Thomas et al, using data from the UK Biobank, determined that in 42% of type 1 DM cases reviewed, disease onset occurred in patients aged 31 to 60 years. The report also found that because type 2 DM is far more common than type 1 in individuals in the 31- to 60-year age group, with type 1 DM making up only 4% of all diabetes cases in this population, identification of type 1 DM is difficult in patients over age 30 years. The presence of type 1 DM was identified in the study using a genetic risk score that employed 29 common genetic variants.[43, 44]

The risk of development of antibodies (anti-islet) in relatives of patients with type 1 DM decreases with increasing age. This finding supports annual screening for antibodies in relatives younger than 10 years and 1 additional screening during adolescence.[4]

Sex- and race-related demographics

Type 1 DM is more common in males than in females. In populations of European origin, the male-to-female ratio is greater than 1.5:1.

Type 1 DM is most common among non-Hispanic whites, followed by African Americans and Hispanic Americans. It is comparatively uncommon among Asians.

Prognosis

Type 1 DM is associated with a high morbidity and premature mortality. More than 60% of patients with type 1 DM do not develop serious complications over the long term, but many of the rest experience blindness, end-stage renal disease (ESRD), and, in some cases, early death. The risk of ESRD and proliferative retinopathy is twice as high in men as in women when the onset of diabetes occurred before age 15 years.[45]

Patients with type 1 DM who survive the period 10-20 years after disease onset without fulminant complications have a high probability of maintaining reasonably good health. Other factors affecting long-term outcomes are the patient’s education, awareness, motivation, and intelligence level. The 2012 American Diabetes Association (ADA) standard of care emphasizes the importance of long-term, coordinated care management for improved outcomes and suggests structural changes to existing systems of long-term care delivery.[5]

The morbidity and mortality associated with diabetes are related to the short- and long-term complications. Such complications include the following:

These complications result in increased risk for ischemic heart disease, cerebral vascular disease, peripheral vascular disease with gangrene of lower limbs, chronic renal disease, reduced visual acuity and blindness, and autonomic and peripheral neuropathy. Diabetes is the major cause of blindness in adults aged 20-74 years, as well as the leading cause of nontraumatic lower-extremity amputation and ESRD.

In both diabetic and non-diabetic patients, coronary vasodilator dysfunction is a strong independent predictor of cardiac mortality. In diabetic patients without coronary artery disease, those with impaired coronary flow reserve have event rates similar to those with prior coronary artery disease, while patients with preserved coronary flow reserve have event rates similar to non-diabetic patients.[46]

Type 1 diabetic patients also have a high prevalence of small-fiber neuropathy.[47, 48] In a prospective study of 27 patients who had type 1 diabetes with a mean disease duration of 40 years, almost 60% of the subjects showed signs or symptoms of neuropathy, including sensory neuropathy symptoms (9 patients), pain (3 patients), and carpal-tunnel symptoms (5 patients).[47, 48] Of the 27 patients, 22 were diagnosed with small-fiber dysfunction by means of quantitative sensory testing.

Abnormal results on intraepidermal nerve-fiber density measurement (IENFD) were seen in 19 patients.[48] IENFD was negatively correlated with HbA1c, but this relation was no longer significant after adjustment for age, body mass index, and height. N-ε-(carboxymethyl) lysine (CML), which is linked to painful diabetic neuropathy, remained independently associated with IENFD even after adjustment for these variables. Large-fiber neuropathy was also common, being found in 16 patients.

Although ESRD is one of the most severe complications of type 1 DM, its incidence is relatively low: 2.2% at 20 years after diagnosis and 7.8% at 30 years after diagnosis.[49] A greater risk is that mild diabetic nephropathy in type 1 diabetic persons appears to be associated with an increased likelihood of cardiovascular disease.[50] Moreover, the long-term risk of an impaired glomerular filtration rate (GFR) is lower in persons treated with intense insulin therapy early in the course of disease than in those given conventional therapy.[51]

Although mortality from early-onset type 1 DM (onset age, 0-14 y) has declined, the same may not be true for late-onset type 1 DM (onset age, 15-29 y). One study suggest that women tend to fare worse in both cohorts and that alcohol and drug use account for more than one third of deaths.[52]

Control of blood glucose, hemoglobin A1c (HbA1c), lipids, blood pressure, and weight significantly affects prognosis. Excess weight gain with intensified diabetes treatment is associated with hypertension, insulin resistance, dyslipidemia and extetnsive atherosclerotic cardiovascular disease.[53]

Patients with diabetes face a lifelong challenge to achieve and maintain blood glucose levels as close to the normal range as possible. With appropriate glycemic control, the risk of both microvascular and neuropathic complications is decreased markedly. In addition, aggressive treatment of hypertension and hyperlipidemia decreases the risk of macrovascular complications.

A study by Zheng et al indicated that HbA1c levels in persons with diabetes are longitudinally associated with long-term cognitive decline, as found using a mean 4.9 cognitive assessments of diabetes patients over a mean 8.1-year follow-up period. The investigators saw a significant link between each 1 mmol/mol rise in HbA1c and an increased rate of decline in z scores for global cognition, memory, and executive function. Patients in the study had a mean age of 65.6 years. The report cited a need for research into whether optimal glucose control in people with diabetes can affect their cognitive decline rate.[54, 55]

The benefits of glycemic control and control of comorbidities must be weighed against the risk of hypoglycemia and the short-term costs of providing high-quality preventive care. However, studies have shown cost savings due to a reduction in acute diabetes-related complications within 1-3 years of starting effective preventive care.

Patient Education

Education is a vital aspect of diabetes management. Patients with new-onset type 1 DM require extensive education if they are to manage their disease safely and effectively and to minimize long-term complications. Such education is best coordinated by the patient’s long-term care providers.

At every encounter, the clinician should educate the patient—and, in the case of children, the parents—about the disease process, management, goals, and long-term complications. In particular, clinicians should do the following:

ADA guidelines urge that attention be paid to older adolescent patients who may be leaving their home and their current health care providers. At the transition between pediatric and adult health care, older teens can become detached from the health care system, putting their medical care and their glycemic control at risk.[5] The guidelines identify the National Diabetes Education Program (NDEP) as a source of materials that can help smooth the transition to adult health care.

Education about an appropriate treatment plan and encouragement to follow the plan are especially important in patients with diabetes. Physicians must ensure that the care for each patient with diabetes includes all necessary laboratory tests, examinations (eg, foot and neurologic examinations), and referrals to specialists (eg, an ophthalmologist or podiatrist).

A dietitian should provide specific diet control education to the patient and family. A nurse should educate the patient about self–insulin injection and performing fingerstick tests for blood glucose level monitoring.

For patient education information, see the Diabetes Center, as well as Diabetes.

History

The most common symptoms of type 1 diabetes mellitus (DM) are polyuria, polydipsia, and polyphagia, along with lassitude, nausea, and blurred vision, all of which result from the hyperglycemia itself.

Polyuria is caused by osmotic diuresis secondary to hyperglycemia. Severe nocturnal enuresis secondary to polyuria can be an indication of onset of diabetes in young children. Thirst is a response to the hyperosmolar state and dehydration.

Fatigue and weakness may be caused by muscle wasting from the catabolic state of insulin deficiency, hypovolemia, and hypokalemia. Muscle cramps are caused by electrolyte imbalance. Blurred vision results from the effect of the hyperosmolar state on the lens and vitreous humor. Glucose and its metabolites cause osmotic swelling of the lens, altering its normal focal length.

Symptoms at the time of the first clinical presentation can usually be traced back several days to several weeks. However, beta-cell destruction may have started months, or even years, before the onset of clinical symptoms.

The onset of symptomatic disease may be sudden. It is not unusual for patients with type 1 DM to present with diabetic ketoacidosis (DKA), which may occur de novo or secondary to the stress of illness or surgery. An explosive onset of symptoms in a young lean patient with ketoacidosis always has been considered diagnostic of type 1 DM.

Over time, patients with new-onset type 1 DM will lose weight, despite normal or increased appetite, because of depletion of water and a catabolic state with reduced glycogen, proteins, and triglycerides. Weight loss may not occur if treatment is initiated promptly after the onset of the disease.

Gastrointestinal (GI) symptoms of type 1 DM are as follows:

Neuropathy affects up to 50% of patients with type 1 DM, but symptomatic neuropathy is typically a late development, developing after many years of chronic prolonged hyperglycemia. Peripheral neuropathy presents as numbness and tingling in both hands and feet, in a glove-and-stocking pattern; it is bilateral, symmetric, and ascending.

History in patients with established diabetes

It is important to inquire about the type and duration of the patient’s diabetes and about the care the patient is receiving for diabetes. Determination of the type of diabetes is based on history, therapy, and clinical judgment. The chronic complications of diabetes are related to the length of time the patient has had the disease.

Ask about the type of insulin being used, delivery system (pump vs injections), dose, and frequency. Also ask about oral antidiabetic agents, if any. Of course, a full review of all medications and over-the-counter supplements being taken is crucial in the assessment of patients with type 1 DM.

Patients using a pump or a multiple-injection regimen have a basal insulin (taken through the pump or with the injection of a long-acting insulin analogue) and a premeal rapid-acting insulin, the dose of which may be determined as a function of the carbohydrate count plus the correction (to adjust for how high the premeal glucose level is). In these patients, ask about the following:

A focused diabetes history should also include the following questions:

In assessing glycemic exposure of a patient with established type 1 DM, review of self-monitored blood glucose levels is necessary. Ideally, this done by uploading time- and date-stamped levels from the patient’s meter to assure full understanding of the frequency of testing and the actual levels.

Questions regarding hypoglycemia and hyperglycemia

Hypoglycemia and hyperglycemia should be considered. Ask the following questions as needed:

Questions regarding microvascular complications

Microvascular complications, such as retinopathy and nephropathy, should be considered as well. Ask the following questions as appropriate:

Questions regarding macrovascular complications

Macrovascular complications should be explored. Questions should include the following:

Questions regarding neuropathy

Potential neuropathy should be taken into account. Ask whether the patient has a history of neuropathy or symptoms of peripheral neuropathy or whether autonomic neuropathy is present (including erectile dysfunction if the patient is a man).

Other questions

The possibility of foot disease should be addressed. Inquire as to whether the patient has a history of foot ulcers or amputations or whether any foot ulcers are present. (See Diabetic Foot and Diabetic Foot Infections.)

The possibility of infection also should be considered. Be sure to inquire about whether frequent infections are a problem and, if so, at what sites.

Physical Examination

In new cases of diabetes, physical examination findings are usually normal. Patients with DKA, however, will have Kussmaul respiration, signs of dehydration, hypotension, and, in some cases, altered mental status.

In established cases, patients should be examined every 3 months for macrovascular and microvascular complications. They should undergo funduscopic examination for retinopathy and monofilament testing for peripheral neuropathy.

Diabetes-focused examination

A diabetes-focused physical examination includes assessment of vital signs, funduscopic examination, limited vascular and neurologic examinations, and foot examination. Other organ systems should be assessed as indicated by the patient’s clinical situation. A comprehensive examination is not necessary at every visit.

Assessment of vital signs

Patients with established diabetes and autonomic neuropathy may have orthostatic hypotension. Orthostatic vital signs may be useful in assessing volume status and in suggesting the presence of an autonomic neuropathy. Measurement of the pulse is important, in that relative tachycardia is a typical finding in autonomic neuropathy, often preceding the development of orthostatic hypotension. If the respiratory rate and pattern suggest Kussmaul respiration, DKA must be considered immediately, and appropriate tests must be ordered.

Funduscopic examination

The funduscopic examination should include a careful view of the retina. Both the optic disc and the macula should be visualized. If hemorrhages or exudates are seen, the patient should be referred to an ophthalmologist as soon as possible. Examiners who are not ophthalmologists tend to underestimate the severity of retinopathy, which cannot be evaluated accurately unless the patients’ pupils are dilated.

Foot examination

The dorsalis pedis and posterior tibialis pulses should be palpated and their presence or absence noted. This is particularly important in patients who have foot infections: poor lower-extremity blood flow can delay healing and increase the risk of amputation.

Documenting lower-extremity sensory neuropathy is useful in patients who present with foot ulcers because decreased sensation limits the patient’s ability to protect the feet and ankles. If peripheral neuropathy is found, the patient should be made aware that foot care (including daily foot examination) is very important for the prevention of foot ulcers and lower-extremity amputation. (See Diabetic Foot and Diabetic Foot Infections.)

Complications

Infections

Infections cause considerable morbidity and mortality in patients with diabetes. Infection may precipitate metabolic derangements, and conversely, the metabolic derangements of diabetes may facilitate infection. (See Infections in Patients with Diabetes Mellitus.)

Patients with long-standing diabetes tend to have microvascular and macrovascular disease with resultant poor tissue perfusion and increased risk of infection. The ability of the skin to act as a barrier to infection may be compromised when the diminished sensation of diabetic neuropathy results in unnoticed injury.

Diabetes increases susceptibility to various types of infections. The most common sites are the skin and urinary tract. Dermatologic infections that occur with increased frequency in patients with diabetes include staphylococcal follicular skin infections, superficial fungal infections, cellulitis, erysipelas, and oral or genital candidal infections. Both lower urinary tract infections and acute pyelonephritis are seen with greater frequency.

A few infections, such as malignant otitis externa, rhinocerebral mucormycosis, and emphysematous pyelonephritis, occur almost exclusively in patients with diabetes, though they are fairly rare even in this population. Infections such as staphylococcal sepsis occur more frequently and are more often fatal in patients with diabetes than in others. Infections such as pneumococcal pneumonia affect patients with diabetes and other patients with the same frequency and severity.[56]

Ophthalmologic complications

Diabetes can affect the lens, vitreous, and retina, causing visual symptoms that may prompt the patient to seek emergency care. Visual blurring may develop acutely as the lens changes shape with marked changes in blood glucose concentrations.

This effect, which is caused by osmotic fluxes of water into and out of the lens, usually occurs as hyperglycemia increases, but it also may be seen when high glucose levels are lowered rapidly. In either case, recovery to baseline visual acuity can take up to a month, and some patients are almost completely unable to read small print or do close work during this period.

Patients with diabetes tend to develop senile cataracts at a younger age than persons without diabetes. Rarely, patients with type 1 DM that is very poorly controlled (eg, those with frequent episodes of DKA) can acutely develop a “snowflake” (or “metabolic”) cataract. Named for their snowflake or flocculent appearance, these cataracts can progress rapidly and create total opacification of the lens within a few days.

Whether diabetes increases the risk of glaucoma remains controversial; epidemiologic studies have yielded conflicting results.[57] Glaucoma in diabetes relates to the neovascularization of the iris (ie, rubeosis iridis diabetica).

Diabetic retinopathy is the principal ophthalmologic complication of DM. (See Diabetic Retinopathy.) Diabetic retinopathy is the leading cause of blindness in the United States in people younger than 60 years and affects the eyes in the following different ways:

Whether patients develop diabetic retinopathy depends on the duration of their diabetes and on the level of glycemic control.[58, 59, 60] The following are the 5 stages in the progression of diabetic retinopathy:

  1. Dilation of the retinal venules and formation of retinal capillary microaneurysms
  2. Increased vascular permeability
  3. Vascular occlusion and retinal ischemia
  4. Proliferation of new blood vessels on the surface of the retina
  5. Hemorrhage and contraction of the fibrovascular proliferation and the vitreous

The first 2 stages of diabetic retinopathy are jointly referred to as background or nonproliferative retinopathy. Initially, the retinal venules dilate, then microaneurysms (tiny red dots on the retina that cause no visual impairment) appear. The microaneurysms or retinal capillaries become more permeable, and hard exudates appear, reflecting leakage of plasma.

Rupture of intraretinal capillaries results in hemorrhage. If a superficial capillary ruptures, a flame-shaped hemorrhage appears. Hard exudates are often found in partial or complete rings (circinate pattern), which usually include multiple microaneurysms. These rings usually mark an area of edematous retina.

The patient may not notice a change in visual acuity unless the center of the macula is involved. Macular edema can cause visual loss; therefore, all patients with suspected macular edema must be referred to an ophthalmologist for evaluation and possible laser therapy. Laser therapy is effective in decreasing macular edema and preserving vision but is less effective in restoring lost vision. (See Macular Edema in Diabetes .)

Preproliferative (stage 3) and proliferative diabetic retinopathy (stages 4 and 5) are the next phases in the progression of the disease. Cotton-wool spots can be seen in preproliferative retinopathy. These represent retinal microinfarcts from capillary occlusion and appear as off-white to gray patches with poorly defined margins.

Proliferative retinopathy is characterized by neovascularization, or the development of networks of fragile new vessels that often are seen on the optic disc or along the main vascular arcades. The vessels undergo cycles of proliferation and regression. During proliferation, fibrous adhesions develop between the vessels and the vitreous. Subsequent contraction of the adhesions can result in traction on the retina and retinal detachment. Contraction also tears the new vessels, which hemorrhage into the vitreous.

Diabetic nephropathy

About 20–30% of patients with type 1 DM develop evidence of nephropathy,[61] and all patients with diabetes should be considered to have the potential for renal impairment unless proven otherwise. Chronically elevated blood pressure contributes to the decline in renal function. The use of contrast media can precipitate acute renal failure in patients with underlying diabetic nephropathy. Although most recover from contrast medium–induced renal failure within 10 days, some have irreversible renal failure. (See Diabetic Nephropathy.)

Diabetic neuropathy

In the peripheral nerves, diabetes causes peripheral neuropathy. (See Diabetic Lumbosacral Plexopathy and Diabetic Neuropathy.) The 4 types of diabetic neuropathy are as follows:

Of these 4 types, distal symmetric sensorimotor polyneuropathy (in a glove-and-stocking distribution) is the most common.[62] Besides causing pain in its early stages, this type of neuropathy eventually results in the loss of peripheral sensation. The combination of decreased sensation and peripheral arterial insufficiency often leads to foot ulceration and eventual amputation.

Acute-onset mononeuropathies in diabetes include acute cranial mononeuropathies, mononeuropathy multiplex, focal lesions of the brachial or lumbosacral plexus, and radiculopathies. Of the cranial neuropathies, the third cranial nerve (oculomotor) is most commonly affected, followed by the sixth nerve (abducens) and the fourth nerve (trochlear).

Patients can present with diplopia and eye pain. In diabetic third-nerve palsy, the pupil is usually spared, whereas in third-nerve palsy due to intracranial aneurysm or tumor, the pupil is affected in 80-90% of cases.

It is important to consider nondiabetic causes of cranial nerve palsies, including intracranial tumors, aneurysms, and brainstem stroke.[63] Therefore, evaluation should include nonenhanced and contrast-enhanced compute4d tomography (CT) or, preferably, magnetic resonance imaging (MRI). Neurologic consultation is recommended. Acute cranial-nerve mononeuropathies usually resolve in 2-9 months. Acute thrombosis or ischemia of the blood vessels supplying the structure involved is thought to cause these neuropathies.

Macrovascular complications

People with diabetes experience accelerated atherosclerosis, affecting the small arteries of the heart, brain, lower extremity, and kidney. Coronary atherosclerosis often occurs at a younger age and is more severe and extensive than in those without diabetes, increasing the risk of ischemic heart disease. Atherosclerosis of the internal carotid and vertebrobasilar arteries and their branches predisposes to cerebral ischemia.

Severe atherosclerosis of the iliofemoral and smaller arteries of the lower legs predisposes to gangrene. Ischemia of a single toe or ischemic areas on the heel are characteristic of diabetic peripheral vascular disease; these result from the involvement of much smaller and more peripheral arteries.

Atherosclerosis of the main renal arteries and their intrarenal branches causes chronic nephron ischemia, which is a significant component of multiple renal lesions in diabetes. However, not all people with type 1 DM are at risk for nephropathy, because there are some polymorphisms in the various factors involved in its pathogenesis, which can modulate the course of this disease from one person to the other.

Risk factors for macrovascular disease

Macrovascular disease is the leading cause of death in patients with diabetes, causing 65-75% of deaths in this group, compared with approximately 35% of deaths in people without diabetes. Diabetes by itself increases the risk of myocardial infarction (MI) 2-fold in men and 4-fold in women, and many patients have other risk factors for MI as well.

The HbA1c value per se, rather than self-reported diabetes status or other established risk factors, robustly predicts MI odds. Each 1% increment in HbA1c independently predicts 19% higher odds for MI.[64] The risk of stroke in people with diabetes is double that of nondiabetic people, and the risk of peripheral vascular disease is 4 times that of people without diabetes.

Patients with diabetes may have an increased incidence of silent ischemia.[65] Diastolic dysfunction is common in patients with diabetes and should be considered in patients who have symptoms of congestive heart failure and a normal ejection fraction.

Laboratory Studies

Plasma glucose

Patients with type 1 diabetes mellitus (DM) typically present with symptoms of uncontrolled hyperglycemia (eg, polyuria, polydipsia, polyphagia). In such cases, the diagnosis of DM can be confirmed with a random (nonfasting) plasma glucose concentration of 200 mg/dL or a fasting plasma glucose concentration of 126 mg/dL (6.99 mmol/L) or higher.[2, 66]

A fingerstick glucose test is appropriate in the emergency department (ED) for virtually all patients with diabetes. All fingerstick capillary glucose levels must be confirmed in serum or plasma to make the diagnosis. All other laboratory studies should be selected or omitted on the basis of the individual clinical situation. Intravenous (IV) glucose testing may be considered for possible early detection of subclinical diabetes.

Individually measured glucose levels may differ considerably from estimated glucose averages calculated from measured hemoglobin A1c (HbA1c) levels.[68] Therefore, caution is urged when the decision is made to estimate rather than actually measure glucose concentration; the difference between the 2 has a potential impact on decision making.

Hemoglobin A

HbA1c is the stable product of nonenzymatic irreversible glycation of the beta chain of hemoglobin by plasma glucose and is formed at rates that increase with increasing plasma glucose levels. HbA1c levels provide an estimate of plasma glucose levels during the preceding 1-3 months. The reference range for nondiabetic people is 6% in most laboratories. Glycated hemoglobin levels also predict the progression of diabetic microvascular complications.

American Diabetes Association (ADA) guidelines recommend measuring HbA1c at least every 6 months in patients with diabetes who are meeting treatment goals and who have stable glycemic control. For patients whose therapy has changed or who are not meeting glycemic goals, the guidelines recommend HbA1c testing every 3 months.[5]

In the past, HbA1c measurements were not considered useful for the diagnosis of DM. Drawbacks included a lack of international standardization and insensitivity for the detection of milder forms of glucose intolerance.

In a 2009 report, however, an international expert committee appointed by the ADA, the European Association for the Study of Diabetes, and the International Diabetes Association recommended the HbA1c assay for diagnosing type 1 and type 2 DM.[3] In the case of type 1 DM, however, the committee recommended using the test only when the condition is suspected but the classic symptoms of type 1 DM—polyuria, polydipsia, polyphagia, a random glucose level of 200 mg/dL, and unexplained weight loss—are absent.

The committee noted the improvement in standardization and cited the following advantages of HbA1c testing over glucose measurement:

Consequently, since 2010 the ADA has included an HbA1c level of 6.5% or higher as a criterion for diabetes diagnosis, with confirmation from repeat testing (unless clinical symptoms are present and the glucose level exceeds 200 mg/dL). HbA1c testing cannot be used in patients with abnormal red blood cell (RBC) turnover (as in hemolytic or iron-deficiency anemia). In children with rapidly evolving type 1 DM, HbA1c may not be significantly elevated despite frank diabetes.[2]

One study found seasonal variability in HbA1c levels of school-age children with higher levels (0.44%) coinciding with colder outdoor temperatures, fewer hours of sunlight, and lower levels of solar irradiance.[69] This effect was seen in school-aged children but not preschoolers and may hold importance for studies using HbA1c as a primary endpoint and HbA1c -based diagnosis of diabetes.

HbA1c cannot be used as an indicator of glycemic control in patients with neonatal diabetes mellitus (NDM) because of the high levels of fetal hemoglobin (HbF) remaining in the blood. A study by Suzuki et al found that glycated albumin, which is not affected by HbF levels, more strongly correlated with 1-month average postprandial blood glucose and was therefore a better marker of diabetes in neonates. This finding is important to neonatologist and those caring for newborns.[70]

Moreover, the overall efficacy of HbA1c testing in diabetes diagnosis remains uncertain. A study presented in 2019, using data derived from 9000 adults, reported diabetes diagnosis with the HbA1c blood test to be unreliable. The investigators found evidence that in comparison with the oral glucose tolerance test, HbA1c testing would lead to a 42% overdiagnosis of glucose tolerance and a 73% underdiagnosis of diabetes, in adults.[71]

ADA guidelines recommend measuring HbA1c at least every 6 months year in patients who are meeting treatment goals and who have stable glycemic control. For patients whose therapy has changed or who are not meeting glycemic goals, the guidelines recommend HbA1c testing every 3 months.[5]

Other laboratory studies

Fructosamine levels also test for glucose levels. Fructosamine is formed by a chemical reaction of glucose with plasma protein and reflects glucose control in the previous 1-3 weeks. This assay, therefore, may show a change in control before HbA1c and often is helpful when applying intensive treatment and in short-term clinical trials.

A white blood cell (WBC) count and blood and urine cultures may be performed to rule out infection.

Urine ketones are not reliable for diagnosing or monitoring diabetic ketoacidosis (DKA), although they may be useful in screening to see whether a hyperglycemic individual may have some degree of ketonemia. The plasma acetone level—specifically, the beta-hydroxybutyrate level—is a more reliable indicator of DKA, along with measurement of plasma bicarbonate or arterial pH as clinically required. (See the Medscape Reference Laboratory Medicine article Ketones.)

Screening for type 1 DM in asymptomatic low-risk individuals is not recommended.[2] However, in patients at high risk (eg, those who have first-degree relatives with type 1 DM), it may be appropriate to perform annual screening for anti-islet antibodies before the age 10 years, along with 1 additional screening during adolescence.[4]

Tests to Differentiate Type 1 from Type 2 Diabetes

Although the oral glucose tolerance test with insulin levels is usually considered unnecessary for diagnosing type 1 DM, the dramatic increase of type 2 DM in the young suggests that assessment of insulin secretion may become more important. The 2011 American Association of Clinical Endocrinologists (AACE) guidelines note that to help distinguish between the 2 types in children, physicians should measure insulin and C-peptide levels and immune markers (eg, glutamic acid decarboxylase [GAD] autoantibodies), as well as obtain a detailed family history.[66]

C-peptide is formed during conversion of proinsulin to insulin. An insulin or C-peptide level below 5 µU/mL (0.6 ng/mL) suggests type 1 DM; a fasting C-peptide level greater than 1 ng/dL in a patient who has had diabetes for more than 1-2 years is suggestive of type 2 (ie, residual beta-cell function). An exception is the individual with type 2 DM who presents with a very high glucose level (eg, >300 mg/dL) and a temporarily low insulin or C-peptide level but who will recover insulin production once normal glucose is restored.

Most patients who present with undiagnosed type 1 DM have the classic symptoms of uncontrolled hyperglycemia, including polyuria, polydipsia, nocturia, fatigue, and weight loss. In these patients, a confirmatory random plasma glucose level of greater than 200 mg/dL is adequate to establish the diagnosis of DM. On occasion, a patient who is ultimately found to have type 1 DM presents with subtle symptoms because of residual insulin secretion.

Islet-cell (IA2), anti-GAD65, and anti-insulin autoantibodies can be present in early type 1 but not type 2 DM. Measurements of IA2 autoantibodies within 6 months of diagnosis can help differentiate between type 1 and type 2 DM. These titers decrease after 6 months. Anti-GAD65 antibodies can be present at diagnosis of type 1 DM and are persistently positive over time. (See also Type 2 Diabetes Mellitus.)

Testing for islet autoantibodies can substitute for expensive genetic testing in those patients suspected of having maturity-onset diabetes of the young (MODY). The prevalence of these antibodies is the same in patients with MODY as in the healthy population. A positive test for positive islet autoantibodies makes MODY highly unlikely.[72]

Approach Considerations

Patients with type 1 diabetes mellitus (DM) require lifelong insulin therapy. Most require 2 or more injections of insulin daily, with doses adjusted on the basis of self-monitoring of blood glucose levels. Long-term management requires a multidisciplinary approach that includes physicians, nurses, dietitians, and selected specialists.

In some patients, the onset of type 1 DM is marked by an episode of diabetic ketoacidosis (DKA) but is followed by a symptom-free “honeymoon period” in which the symptoms remit and the patient requires little or no insulin. This remission is caused by a partial return of endogenous insulin secretion, and it may last for several weeks or months (sometimes for as long as 1-2 years). Ultimately, however, the disease recurs, and patients require insulin therapy.

Often, the patient with new-onset type 1 DM who presents with mild manifestations and who is judged to be compliant can begin insulin therapy as an outpatient. However, this approach requires close follow-up and the ability to provide immediate and thorough education about the use of insulin; the signs, symptoms, and treatment of hypoglycemia; and the need to self-monitor blood glucose levels.

The American Diabetes Association (ADA) recommends using patient age as one consideration in the establishment of glycemic goals, with targets for preprandial, bedtime/overnight, and hemoglobin A1c (HbA1c) levels.[5] In 2014, the ADA released a position statement on the diagnosis and management of type 1 diabetes in all age groups. The statement includes a new pediatric glycemic control target of HbA1c of less than 7.5% across all pediatric age groups, replacing earlier guidelines that specified different glycemic control targets by age. The adult HbA1c target of less than 7% did not change. Individualized lower or higher targets may be used based on patient need.[73, 74]

In addition to diagnosis and management, the new statement also covers screening for long-term complications, workplace management, diabetes in older patients, and diabetes in pregnancy, and recommends unimpeded access to glucose test strips for blood glucose testing and use of continuous glucose monitoring.[73, 74]

Although patients with type 1 DM have normal incretin response to meals, administration of exogenous glucagonlike peptide 1 (GLP-1) reduces peak postprandial glucose by 45%. Long-term effects of exogenously administered GLP-1 analogues warrant further studies.[75]

Pancreatic transplantation for patients with type 1 DM isg a possibility in some referral centers. It is performed most commonly with simultaneous kidney transplantation for end-stage renal disease (ESRD).

The American Diabetes Association's Standards of Medical Care in Diabetes for type 1 and 2 diabetes highlight recommendations most relevant to primary care.[76] (See Guidelines.)

The care of patients with type 1 diabetes mellitus is summarized below.

Tight glycemic control

The association between chronic hyperglycemia and increased risk of microvascular complications in patients with type 1 DM was demonstrated in the Diabetes Control and Complications Trial (DCCT).[77] In that trial, intensive therapy designed to maintain normal blood glucose levels greatly reduced the development and progression of retinopathy, microalbuminuria, proteinuria, and neuropathy, as assessed over 7 years.

The DCCT ended in 1993. However, the Epidemiology of Diabetes Interventions and Complications Study (EDIC), an observational study that continues to follow the patients previously enrolled in the DCCT, has demonstrated continued benefit from intensive treatment.[78, 79]

Benefits

Benefits of tight glycemic control include not only continued reductions in the rates of microvascular complications but also significant differences in cardiovascular events and overall mortality. These benefits occurred even though subjects in the intensively treated group and those in the standard treatment group maintained similar HbA1c levels (about 8%), starting 1 year after the DCCT ended. It is postulated that a “metabolic memory” exists and that better early glycemic control sets the stage for outcomes many years in the future.

Increasing HbA1c levels correlated with increasing risk of developing heart failure in a study of 20,985 patients with type 1 DM. Thus, improved glycemic control should prevent heart failure as well.[80]

Risks

For many patients, the HbA1c target should be less than 7%, with a premeal blood glucose level of 80–130 mg/dL. However, targets should be individualized.

Individuals with recurrent episodes of severe hypoglycemia, cardiovascular disease, advanced complications, substance abuse, or untreated mental illness may require higher targets, such as an HbA1c of less than 8% and preprandial glucose levels of 100-150 mg/dL. The 2011 American Association of Clinical Endocrinologists (AACE) guidelines support the creation of individualized targets that consider these factors as part of a comprehensive treatment plan.[81]

Although tight glycemic control is beneficial, an increased risk of severe hypoglycemia accompanies lower blood glucose levels. The 2011 AACE guidelines for developing a comprehensive care plan emphasize that hypoglycemia should be avoided.[66]

In patients with type 1 DM, recurrent and chronic hypoglycemia has been linked to cognitive dysfunction.[82] This has important implications in the management of children with type 1 DM.[83]

An 18-year follow-up of the DCCT by Jacobson et al found that HbA1c levels and retinal and renal complications were independently linked to cognitive declines. No relation with macrovascular risk factors or severe hypoglycemic events was found. A smoking history was modestly associated with decrements in learning, memory, spatial information processing, and psychomotor efficiency. This information is useful in advising patients with type 1 DM interested in preserving cognitive function.

Self-Monitoring of Glucose Levels

Optimal diabetic control requires frequent self-monitoring of blood glucose levels, which allows rational adjustments in insulin doses. All patients with type 1 DM should learn how to self-monitor and record their blood glucose levels with home analyzers and adjust their insulin doses accordingly.

Insulin-dependent patients ideally should test their plasma glucose daily before meals, in some cases 1-2 hours after meals, and at bedtime. In practice, however, patients often obtain 2-4 measurements each day, including fasting levels and levels checked at various other times (eg, preprandially and at bedtime).

Instruct patients with type 1 DM in the method of testing for urine ketones with commercially available reagent strips. Advise patients to test for urine ketones whenever they develop any of the following:

Continuous Glucose Monitoring

Continuous glucose monitors (CGMs) contain subcutaneous sensors that measure interstitial glucose levels every 1-5 minutes, providing alarms when glucose levels are too high or too low or are rapidly rising or falling. CGMs transmit to a receiver, which either is a pagerlike device or is integral to an insulin pump. Looking at the continuous glucose graph and responding to the alarms can help patients avoid serious hyperglycemia or hypoglycemia.

CGMs have several drawbacks. First, there is a lag between glucose levels in the interstitial space and levels in capillary blood, so that the levels recorded by the CGM may differ from a fingerstick (capillary) glucose reading. For that reason, the trends (ie, whether the glucose levels are rising or falling) tend to be more helpful.

Second, patients may overtreat hyperglycemia (repeatedly giving insulin because the glucose levels do not fall rapidly enough—a phenomenon known as stacking), as well as overtreat low glucose levels (because the glucose levels rise slowly with ingestion of carbohydrate).

Use of CGMs may help to prevent significant glucose variability in patients receiving either multiple daily injection therapy or continuous insulin infusion therapy.[84] Additionally, continuous glucose monitoring is associated with reduced time spent in hypoglycemia.[85] Whether glucose variability is detrimental in the absence of hypoglycemia remains an unresolved question; in any event, variability leads to the expense of frequent testing.

Guidelines from the Endocrine Society[86] recommend the use of real-time CGMs in adult patients with type 1 DM who have demonstrated that they are able to use these devices on a nearly daily basis. The guidelines suggest the intermittent use of CGM systems for short-term retrospective analysis in the following cases[86] :

The 2018 edition of the ADA’s Standards of Medical Care in Diabetes recommends that continuous glucose monitoring be used in all persons aged 18 years or older with type 1 DM (down from the previously recommended age of 25 years or above) in whom glycemic targets are unmet.[87]

A study comparing the performance of three CGM devices—Navigator (Abbott Diabetes Care), Seven Plus (Dexcom), and Guardian (Medtronic)—found the Navigator to be the most accurate.[6, 7] For commercial reasons, however, this device is no longer on the market in the United States, though it remains available in Europe, Israel, Australia, and other areas; the other two CGM devices are still available in the United States.

In September 2013, the US Food and Drug Administration (FDA) approved a sensor-augmented insulin pump system that includes an automated low-glucose suspend safety feature (Medtronic's MiniMed 530G with Enlite) for use by patients aged 16 years and older with type 1 DM.[88, 89] When the continuous glucose sensor detects that blood sugar has fallen below a preset threshold (60-90 mg/dL) and the patient fails to respond to a first alarm, the pump automatically stops insulin delivery. The manufacturer indicates the Elite sensor is 31% more accurate than previous-generation sensors, as well as being 69% smaller and simpler to insert.[88, 89]

In December 2016, Dexcom’s G5 Mobile Continuous Glucose Monitoring System became the first CGM to win FDA approval as a replacement for finger-stick testing for determination of insulin doses, although twice-daily finger-stick testing was still required for calibration.[90]

In March 2018, the FDA approved Medtronic’s stand-alone CGM, Guardian Connect, which eschews use of a receiver and makes data viewable via a smartphone display alone. Receiving CGM data through its smartphone app, the device works with an artificial intelligence app to assess glucose level response to various factors, including an individual’s food intake, insulin dosages, and daily routines. The approval was made for patients with diabetes aged 14-75 years.[91]

Another device, the FreeStyle Libre Flash Glucose Monitoring System (Abbott Diabetes Care), approved by the FDA in September 2017, allows patients to reduce the number of required finger-stick tests by measuring glucose levels via a self-applied sensor inserted into the back of the upper arm.[92]

In June 2018, the FDA approved the Eversense Continuous Glucose Monitoring system (Senseonics), the first continuous glucose monitoring system with a fully implantable glucose sensor, for persons aged 18 years or older with diabetes. Using a fluorescence-based sensor that a physician implants subcutaneously in the patient's upper arm (via an office procedure), the device has a transmitter that is worn above the sensor, with the CGM employing a mobile app to show glucose values and trends. In addition, the app alerts the patient to high and low glucose values, with the transmitter also emitting on-body vibration alerts. The device is intended for adjunctive use, with fingerstick monitoring and twice-daily calibrations required. The implant lasts for up to 3 months before needing replacement.[93, 94]

Artificial pancreas

Closed-loop systems, also known as artificial pancreases, are in development for use in improving glycemic control in type 1 diabetes. These systems include a CGM that is in constant communication with an infusion pump, with a blood glucose device (eg, a glucose meter) utilized for CGM calibration. An external processor, such as a cell phone, runs control algorithm software, receiving data from the CGM. The data is used to perform a series of calculations, producing dosing instructions that are sent to the infusion pump.[95]

The artificial pancreases are being developed to administer either insulin or glucagon or a combination of the two agents.[96] A 1-month study in 20 patients indicated that, with regard to keeping blood glucose levels in the target range over a 24-hour period, round-the-clock use of closed-loop glucose control is more effective than use of a patient-controlled sensor-augmented pump.[97, 98]

In September 2016, the FDA approved the first artificial pancreas, Medtronic's MiniMed 670G, for persons aged 14 years or older with type 1 diabetes. A hybrid closed-loop system, it still requires patients to determine the number of carbohydrates in their food and input that data into the system, manually requesting the insulin dose needed for meals.[99]  In June 2018, the FDA extended the MiniMed 670G’s approval to children aged 7-13 years with type 1 diabetes.[100]

Insulin Therapy

Types of insulin

Rapid-, short-, intermediate-, and long-acting insulin preparations are available. Various pork, beef, and beef-pork insulins were previously used; however, in the United States, recombinant human insulin is now used almost exclusively. Commercially prepared mixtures of insulin are also available.

Rapid-acting insulins include lispro, glulisine, and aspart insulin. Lispro insulin is a form of regular insulin that is genetically engineered with the reversal of the amino acids lysine and proline at B28,29 in the B chain. Glulisine insulin substitutes glutamic acid for lysine in position B29. Aspart insulin substitutes aspartic acid for proline in position 28 of the B chain.

These insulins are absorbed more quickly and have a rapid onset of action (5-10 minutes), a short interval to peak action (45-75 minutes), and a short duration of action (2-4 hours). Therefore, they can be administered shortly before eating. In addition, neutral protamine Hagedorn (NPH) insulin will not inhibit the action of insulin lispro when the 2 agents are mixed together right before injection; this is not true of regular insulin.

A rapid-acting inhaled insulin powder (Afrezza) for types 1 and 2 diabetes mellitus was approved by the FDA in June 2014. It is regular insulin but is considered rapid-acting because it peaks at 12-15 minutes and returns to baseline levels at about 160 minutes. Approval was based on a study involving over 3,000 patients over a 24-week period. In persons with type 1 diabetes, the inhaled insulin was found to be noninferior to standard injectable insulin when used in conjunction with basal insulin at reducing hemoglobin A1c. In persons with type 2 diabetes, the inhaled insulin was compared to placebo inhalation in combination with oral diabetic agents and showed a statistically significant lower hemoglobin A1c.[101, 102]

Short-acting insulin includes regular insulin. Regular insulin is a preparation of zinc insulin crystals in solution. When it is administered subcutaneously, its onset of action occurs in 0.5 hours, its peak activity comes at 2.5-5 hours, and its duration of action is 4-12 hours.

The standard strength of regular insulin is 100 U/mL (U-100), but 500 U/mL (U-500) insulin is increasingly used, albeit mostly in type 2 DM. Accidental prescribing of U-500 rather than U-100 is a potential safety issue.[103] A study by de la Pena et al found that although the overall insulin exposure and effects of 500 U/mL insulin are similar to those of 100 U/mL insulin, peak concentration was significantly lower with U-500, and the effect after the peak was prolonged; areas under the curve were similar for the 2 strengths.[104]

Both regular human insulin and rapid-acting insulin analogues are effective at lowering postprandial hyperglycemia in various basal bolus insulin regimens used in type 1 DM. Rapid-acting insulin analogues may be slightly better at lowering HbA1c and are preferred by most US diabetologists, but the differences are clinically insignificant.[105]

In September 2017, the FDA approved the rapid-acting insulin aspart Fiasp for the treatment of adults with diabetes. This human insulin analog is formulated with niacinamide, which aids in speeding the initial absorption of insulin. Dosing can occur at the beginning of a meal or within 20 minutes after the meal commences. In a study of adult patients with type 1 DM, Fiasp could be detected in the circulation about 2.5 minutes after it was administered. Maximum insulin levels occurred approximately 63 minutes after the drug’s administration.[106, 107]

Semilente insulin is like regular insulin and is a rapid-acting insulin with a slightly slower onset of action. It contains zinc insulin microcrystals in an acetate buffer. It is not readily available in the United States.

Intermediate-acting insulins include NPH insulin, a crystalline suspension of human insulin with protamine and zinc. NPH provides a slower onset of action and longer duration of action than regular insulin does. The onset of action usually occurs at 1-2 hours, the peak effect is noted at 4-12 hours, and the duration of action is normally 14–24 hours.

Lente insulin is a suspension of insulin in buffered water that is modified by the addition of zinc chloride. This insulin zinc suspension is equivalent to a mixture of 30% prompt insulin zinc (Semilente) and 70% extended insulin zinc (Ultralente). It is not used in the United States.

Long-acting insulins used in the United States include insulin glargine (Lantus, Toujeo) and insulin detemir (Levemir). Insulin glargine has no peak and produces a relatively stable level lasting more than 24 hours. In some cases, it can produce a stable basal serum insulin concentration with a single daily injection, though patients requiring lower doses typically are given twice-daily injections. Insulin detemir has a duration of action that may be substantially shorter than that of insulin glargine but longer than those of intermediate-acting insulins.

Toujeo 300 U/mL is a newer dosage strength and form of insulin glargine than Lantus 100 U/mL, having been approved by the FDA in February 2016. Compared with those of Lantus 100 U/mL, the pharmacokinetic and pharmacodynamic profiles of Toujeo are more stable and prolonged; the duration of action exceeds 24 hours. Clinical trials showed comparable glycemic control between Lantus and Toujeo, although the trials noted the need for higher daily basal insulin doses (ie, 12-17.5%) with Toujeo. The risk for nocturnal hypoglycemia was lower with Toujeo in insulin-experienced patients with type 2 diabetes, but this was not the case for insulin-naïve patients with type 1 DM or for patients with type 2 DM.[108]

With its March 2018 approval by the FDA, Toujeo Max SoloStar became the highest capacity long-acting insulin pen on the market. Toujeo Max necessitates fewer refills and, for some diabetes patients, fewer injections to deliver the required Toujeo dosage.[109]

A new ultralong-acting basal insulin, insulin degludec (Tresiba), which has a duration of action beyond 42 hours, has also been approved by the FDA. It is indicated for diabetes mellitus types 1 and 2. A combination product of insulin degludec and the rapid-acting insulin aspart was also approved (Ryzodeg 70/30). Approval was based on results from the BEGIN trial[110, 111, 112] that showed noninferiority to comparator productions. The cardiovascular outcomes trial (DEVOTE) comparing cardiovascular safety of insulin degludec to that of insulin glargine in patients with type 2 DM is ongoing.

Mixtures of insulin preparations with different onsets and durations of action frequently are administered in a single injection by drawing measured doses of 2 preparations into the same syringe immediately before use. The exceptions are insulin glargine and insulin detemir, which should not be mixed with any other form of insulin. Preparations that contain a mixture of 70% NPH and 30% regular human insulin (eg, Novolin 70/30, Humulin 70/30, Ryzodeg 70/30) are available, but the fixed ratios of intermediate-acting or long-acting to rapid-acting insulin may restrict their use.

An ultrafast-acting insulin aspart formulation for mealtimes (Fiasp), for adult patients with type 1 or 2 DM, was approved in January 2017 for use in Canada and the Europe Union. The drug contains conventional mealtime insulin aspart in combination with two ingredients—vitamin B3 and the amino acid L-arginine—that are meant to allow faster insulin absorption so the medication can better mimic natural physiologic insulin. Unlike in the European Union, however, the new formulation is not approved for insulin pumps in Canada. The product has not yet been approved for use in the United States.[113]

Insulin glargine and cancer

Controversy has arisen over a disputed link between insulin glargine and cancer. On July 1, 2009, the FDA issued an early communication regarding a possible increased risk of cancer in patients using insulin glargine (Lantus).[114] The FDA communication was based on 4 observational studies that evaluated large patient databases and found some association between insulin glargine (and other insulin products) and various types of cancer.

The validity of the link remains in question, however. The duration of these observational studies was shorter than that considered necessary to evaluate for drug-related cancers. Additionally, findings were inconsistent within and across the studies, and patient characteristics differed across treatment groups.

In a study by Suissa et al, insulin glargine use was not associated with an increased risk of breast cancer during the first 5 years of use. The risk tended to increase after 5 years, however, and significantly so for the women who had taken other forms of insulin before starting insulin glargine.[115]

A study by Johnson et al found the same incidences for all cancers in patients receiving insulin glargine as in those not receiving insulin glargine. Overall, no increase in breast cancer rates was associated with insulin glargine use, although patients who used only insulin glargine had a higher rate of cancer than those who used another type of insulin. This finding was attributed to allocation bias and differences in baseline characteristics.[116]

Common insulin regimens

The goal of treatment in type 1 DM is to provide insulin in as physiologic a manner as possible. Insulin replacement is accomplished by giving a basal insulin and a preprandial (premeal) insulin. The basal insulin is either long-acting (glargine or detemir) or intermediate-acting (NPH). The preprandial insulin is either rapid-acting (lispro, aspart, or glulisine) or short-acting (regular). Currently, NPH insulin is being used less frequently, whereas insulin glargine and insulin detemir are being used more frequently.

For patients on intensive insulin regimens (multiple daily injections or insulin pumps), the preprandial dose is based on the carbohydrate content of the meal (the carbohydrate ratio) plus a correction dose if their blood glucose level is elevated (eg, an additional 2 U of rapid-acting insulin to correct the blood glucose from a level of 200 mg/dL to a target of 100 mg/dL). This method allows patients more flexibility in caloric intake and activity, but it requires more blood glucose monitoring and closer attention to the control of their diabetes.

Common insulin regimens include the following:

Insulin is sensitive to heat and exposure to oxygen. Once a bottle of insulin is open, it should be used for no more than 28 days and then discarded; even if there is still some insulin in the bottle, it may have lost its clinical effectiveness. Insulin kept in a pump reservoir for longer than 3 days may lose its clinical effectiveness (though insulin aspart has now been approved for use for as long as 6 days in a pump).

Sometimes, insulin distributed from the pharmacy has been exposed to heat or other environmental factors and therefore may be less active. If a patient is experiencing unexplained high blood sugar levels, new insulin vials should be opened and used.

Initiation of insulin therapy

The initial daily insulin dose is calculated on the basis of the patient’s weight. This dose is usually divided so that one half is administered before breakfast, one fourth before dinner, and one fourth at bedtime. After selecting the initial dose, adjust the amounts, types, and timing according to the plasma glucose levels. Adjust the dose to maintain preprandial plasma glucose at 80-150 mg/dL (ie, 4.44-8.33 mmol/L).

The insulin dose is often adjusted in increments of 10% at a time, and the effects are assessed over about 3 days before any further changes are made. More frequent adjustments of regular insulin can be made if a risk of hypoglycemia is present.

Carbohydrate counting may be used to determine the meal-time insulin dose. Because patients may experience hyperglycemic episodes despite strict adherence to carbohydrate counting, particularly after meals that are high in protein or fat, Australian researchers developed an algorithm for estimating the mealtime insulin dose on the basis of measurements of physiologic insulin demand evoked by foods in healthy adults. The researchers showed that use of this algorithm improved glycemic control.[117]

Initiation of insulin therapy in children

Children with moderate hyperglycemia but without ketonuria or acidosis may be started with a single daily subcutaneous injection of 0.3-0.5 U/kg of intermediate-acting insulin alone. Children with hyperglycemia and ketonuria but without acidosis or dehydration may be started on 0.5-0.7 U/kg of intermediate-acting insulin and subcutaneous injections of 0.1 U/kg of regular insulin at 4- to 6-hour intervals.

Multiple daily injections

Multiple subcutaneous insulin injections are administered to control hyperglycemia after meals and to maintain normal plasma glucose levels throughout the day. This may increase the risks of hypoglycemia. Therefore, patients should be well educated about their disease and about self-monitoring of plasma glucose levels.

About 25% of the total daily dose is administered as intermediate-acting insulin at bedtime, with additional doses of rapid-acting insulin before each meal (4-dose regimen). Where available, a basal insulin such as glargine or detemir is preferred to NPH. These patients may need additional intermediate- or long-acting insulin in the morning for all-day coverage.

Patients should adjust their daily dosage(s) on the basis of their self-monitoring of glucose levels before each meal and at bedtime. Patients should also assess their plasma glucose levels at 2:00-4:00 AM at least once per week during the first few weeks of treatment and thereafter as indicated.

Continuous subcutaneous insulin infusion

A small battery-operated infusion pump that administers a continuous subcutaneous infusion of rapid-acting insulin can provide selected, programmed basal rate(s) of insulin and a manually administered bolus dose before each meal. The patient self-monitors preprandial glucose levels to adjust the bolus dose(s).

The CSII method provides better control than the MDI method does. Initially, hypoglycemia is common with pump therapy, but once metabolic control is achieved, the risk is the same as with MDI. Bergenstal et al determined that sensor-augmented pump therapy led to better glycemic control and that more patients reached targets with this technology than with injection therapy.[118]

An Australian observational case-control study involving 690 children with type 1 diabetes found that CSII, in comparison with insulin injection therapy, yielded a long-term improvement in glycemic control, as well as a reduction in complications such as severe hypoglycemia and hospitalization for diabetic ketoacidosis (DKA).[119, 120] HbA1c improvement remained significant in the pump therapy cohort throughout 7 years of follow-up.

The rate of severe hypoglycemic events per 100 patient-years dropped from 14.7 to 7.2 with pump therapy but jumped from 6.8 to 10.2 events per 100 patient-years with injection therapy.[119, 120] Hospitalization rates for DKA were lower in children receiving pump therapy (2.3 per 100 patient-years) compared with those receiving injection therapy (4.7 per 100 patient-years) over 1160 patient-years of follow-up.

Increased bedtime doses of hypoglycemic agents with nighttime peaks in action may correct early morning hyperglycemia but may be associated with undesirable nocturnal hypoglycemia. Targeted CSII programming can facilitate the prevention of early-morning hyperglycemia in selected patients.

Changes in altitude may affect delivery from insulin pumps. During the flight of a commercial airliner (200 mm Hg pressure decrease), excess insulin delivery of 0.623% of cartridge volume was demonstrated as a result of bubble formation and expansion of preexisting bubbles.[121]

The American Association of Clinical Endocrinologists and American College of Endocrinology released a consensus statement on insulin pump management:[122]

Local allergic reactions

Generalized insulin allergy is rare. Symptoms occur immediately after the injection and include urticaria, angioedema, pruritus, bronchospasm, and, rarely, circulatory shock. As a rule, allergy may be treated with antihistamines. Some cases may require epinephrine and intravenous (IV) steroids.

Local allergic reactions can occur at the site of insulin injections and can cause pain, burning, local erythema, pruritus, and induration. These complications are less common with the human insulins now in use than with the animal insulins once widely employed. Such reactions usually resolve spontaneously without any intervention.

Local fat atrophy or hypertrophy at injection sites was common with animal insulins but is rare with human insulin and insulin analogues. Patients do not require any specific treatment of local fat hypertrophy, but injection sites should be rotated. Changing to a different insulin preparation may be necessary.[123]

Management of Hypoglycemia

Hypoglycemia may result from a change in insulin dose, a small or missed meal, or strenuous exercise. Regular insulin doses may cause hypoglycemia if the patient becomes anorectic or has another cause for reduced food intake, has gastroparesis, or is vomiting.

Common symptoms of hypoglycemia are light-headedness, dizziness, confusion, shakiness, sweating, and headache. Patients should be made aware of these symptoms and educated to respond rapidly with sugar intake. They should be advised to carry candy or sugar cubes. Family members can be taught to administer a subcutaneous injection of glucagon. In an emergency situation, initial treatment consists of a bolus injection of 25 mL of 50% glucose solution followed by a continuous glucose infusion.

Repeated hypoglycemia may be an aggravating factor in preclinical atherosclerosis. Thus, in the process of designing treatment plans aimed at reducing the glycemic burden and minimizing vascular complications, hypoglycemic episodes might negate some benefits.[124]

Controversy surrounds the question of whether severe hypoglycemia in youths with type 1 DM has lasting cognitive consequences. In a follow-up to the DCCT, recurrent and chronic hypoglycemia was linked to cognitive dysfunction.[82] In another study, however, electroencephalography (EEG) and cognition studies were performed at baseline and 16 years later in patients with type 1 DM, and no association between early severe hypoglycemia and subsequent reduced adult cognition or EEG changes was found.[125]

Management of Hyperglycemia

Acute hyperglycemia, even when not associated with DKA (or hyperosmolar hyperglycemic state [HHS], which occurs most commonly in type 2 DM), is harmful for a number of reasons. If the blood glucose level exceeds the renal threshold for glucose (which is typically 240 mg/dL in a healthy person but is lower in older patients, those with renal insufficiency, and pregnant women), an osmotic diuresis ensues, with loss of glucose, electrolytes, and water.

In addition, hyperglycemia impairs leukocyte function through a variety of mechanisms. Patients with diabetes have an increased rate of wound infection, and hyperglycemia impairs wound healing.

In patients with known, poorly controlled type 1 DM, no absolute level of blood glucose elevation mandates admission to the hospital or administration of insulin in the emergency department (ED). In general, lowering the patient’s glucose level in the ED does not correct the underlying cause and has no long-term effect on the patient’s glucose levels. Therefore, a plan for lowering and monitoring the patient’s glucose levels is needed.

Adequacy of follow-up is extremely important. Whether insulin is given in the ED is of less consequence and can be decided on an individual basis.

Patients with type 1 DM can have coexisting illnesses that aggravate hyperglycemia, such as infection, coronary artery disease (CAD), or fever. Additionally, certain medications can aggravate the condition.

Diabetic ketoacidosis

DKA involves acute metabolic changes in the body that develop as a result of lack of insulin or poor response to insulin arising from stress or illness. DKA is characterized by hyperglycemia, ketosis, and acidosis, leading to osmotic diuresis and dehydration. Volume repletion, insulin therapy, and specific metabolic corrections are the keys to treatment of DKA. (See Diabetic Ketoacidosis.)

Dawn and Somogyi phenomena

The dawn phenomenon is the normal tendency of the blood glucose to rise in the early morning before breakfast. This rise, which may result from the nocturnal spikes in growth hormone that cause insulin resistance, is probably enhanced by increased hepatic gluconeogenesis secondary to the diurnal rise in serum cortisol.

Augmented hepatic gluconeogenesis and glycogen cycling are known to occur in patients with type 1 DM. However, both abnormalities, regardless of the duration of diabetes, can be corrected with intensified insulin therapy.[126]

In some patients, however, nocturnal hypoglycemia may be followed by a marked increase in fasting plasma glucose with an increase in plasma ketones (the Somogyi phenomenon). Thus, both the dawn phenomenon and the Somogyi phenomenon are characterized by morning hyperglycemia, but the latter is considered to be rebound (counterregulation) hyperglycemia.

The existence of a true Somogyi phenomenon is a matter of debate. Most endocrinologists now believe this phenomenon reflects waning of insulin action with consequent hyperglycemia.

In cases of the dawn phenomenon, the patient should check blood glucose levels at 2:00-4:00 AM. The dawn and Somogyi phenomena can be ameliorated by administering intermediate insulin at bedtime.

Use of insulin

The insulin coverage, with a sliding scale for insulin administration, should not be the only intervention for correcting hyperglycemia, because it is reactive rather than proactive. Also, insulin may be used inappropriately when hyperglycemia reflects hepatic gluconeogenesis in response to previously uncorrected hypoglycemia.

Continue intermediate-acting (ie, NPH or Lente) insulin at 50-70% of the daily dose divided into 2 or, occasionally, 3-4 daily doses. Administer supplemental regular insulin on a sliding scale. Blood glucose should be monitored before meals and at bedtime.

Diet

One of the first steps in managing type 1 DM is diet control. According to ADA policy, dietary treatment is based upon nutritional assessment and treatment goals. Dietary recommendations should take into account the patient’s eating habits and lifestyle. For example, patients who participate in Ramadan may be at higher risk of acute diabetic complications. Although these patients do not eat during the annual observance, they should be encouraged to actively monitor their glucose, alter the dosage and timing of their medication, and seek dietary counseling and patient education to counteract these complications.[127]

Diet management includes education about how to adjust the timing, size, frequency, and composition of meals so as to avoid hypoglycemia or postprandial hyperglycemia. All patients on insulin should have a comprehensive diet plan, created with the help of a professional dietitian, that includes the following:

Caloric distribution is an important aspect of dietary planning in these patients. A recommended distribution consists of 20% of daily calories for breakfast, 35% for lunch, 30% for dinner, and 15% for a late-evening snack.

The minimum protein requirement for good nutrition is 0.9 g/kg/day (usual range, 1-1.5 g/kg/day), but a reduced protein intake is indicated in cases of nephropathy. Fat intake should be limited to no more than 30% of the total calories, and a low-cholesterol diet is recommended. Patients should minimize consumption of sugars and ensure that they have adequate fiber intake. In some cases, midmorning and midafternoon snacks are important to avoid hypoglycemia.

Activity

Exercise is an important aspect of diabetes management. Patients should be encouraged to exercise regularly.

Educate the patients about the effects of exercise on the blood glucose level. If patients participate in rigorous exercise for more than 30 minutes, they may develop hypoglycemia unless they either decrease the preceding insulin injection by 10-20% or have an extra snack. Patients must also make sure to maintain their hydration status during exercise.

Management of Complications

Infections

Diabetes predisposes patients to a number of infectious diseases (see Infections in Patients with Diabetes Mellitus). These include the following:

Ophthalmologic complications

Patients with preproliferative or proliferative retinopathy must immediately be referred for ophthalmologic evaluation. Laser therapy is effective in this condition, especially if it is provided before hemorrhage occurs.

Often, the first hemorrhage is small and is noted by the patient as a fleeting dark area (or “floater”) in the field of vision. Because subsequent hemorrhages can be larger and more serious, the patient should immediately be referred to an ophthalmologist for possible laser therapy. Patients with retinal hemorrhage should be advised to limit their activity and keep their head upright (even while sleeping), so that the blood settles to the inferior portion of the retina and thus obscures less of the central visual area.

Multifactorial intervention is important for slowing the progression of diabetic retinopathy. Metabolic control, smoking cessation, and blood pressure control are all protective. Patients with active proliferative diabetic retinopathy are at increased risk for retinal hemorrhage if they receive thrombolytic therapy; therefore, this condition is a relative contraindication to the use of thrombolytic agents. (See Diabetic Retinopathy and Macular Edema in Diabetes.)

Diabetic nephropathy

Extreme care should be exercised whenever any nephrotoxic agent is used in a patient with diabetes. Potentially nephrotoxic drugs should be avoided whenever possible. Renally excreted or potentially nephrotoxic drugs should be given at reduced doses appropriate to the patient’s serum creatinine level. (See Diabetic Nephropathy.)

In particular, caution should be exercised when contrast-enhanced radiologic studies are being considered in patients with diabetes who have a creatinine level higher than 2 mg/dL. Indeed, such studies should absolutely be avoided in patients with a creatinine level higher than 3 mg/dL.

Patients with diabetes who must undergo such studies should be well hydrated before, during, and after the study, and their renal function should be carefully monitored.[128] A better solution is to seek equivalent clinical information by using an alternative modality that does not require the use of contrast material (eg, ultrasonography, noncontrast computed tomography [CT], or magnetic resonance imaging [MRI]).

Current ADA guidelines recommend annual screening for nephropathy.[5] All adults with diabetes should have serum creatinine measured at least annually. In adults (and children aged 10 years or older) who have had type 1 DM for 5 or more years, annual assessment of urine albumin excretion is appropriate.

Microalbuminuria and macroalbuminuria are not permanent features in most diabetic children and adolescents.[129] Regression of microalbuminuria is common; female gender, absence of retinopathy, better glucose control, lower blood pressure, and better lipid control favor this outcome.[130] In patients with persistent microalbuminuria, the use of angiotensin-converting enzyme (ACE) inhibitors and good metabolic control can usually induce remission.

Progression and regression of kidney disease are common even after development of persistent microalbuminuria. Tight glycemic control, lower blood pressure, and a favorable lipid profile are associated with improved outcome.[130]

When chronic kidney disease is present, reduction of protein intake may improve renal function. If kidney disease is advanced or difficult to manage or its etiology is unclear, consider referral to a physician with experience in kidney disease patient care.

Control of blood pressure is a critical element of care. An ACE inhibitor or an angiotensin II receptor blocker (ARB) should be used because these classes of agents decrease proteinuria and slow the decline in renal function independent of the effect on blood pressure.[131] ACE inhibitors and ARBs tend to increase the serum potassium levels and therefore should be used with caution in patients with renal insufficiency or elevated serum potassium levels.

Diabetic neuropathy

Autonomic dysfunction can involve any part of the sympathetic or parasympathetic chains and produce myriad manifestations.[62, 132] Patients likely to seek care in the ED are those with diabetic gastroparesis and vomiting, severe diarrhea, bladder dysfunction and urinary retention, or symptomatic orthostatic hypotension. Treatment of gastroparesis is symptomatic, and symptoms tend to wax and wane. Patients with gastroparesis may benefit from metoclopramide or erythromycin.

Before these therapies are started, the degree of dehydration and metabolic imbalance must be assessed, and other serious causes of vomiting must be excluded. In severe cases, gastric pacing has been used. Patients with disabling orthostatic hypotension may be treated with salt tablets, support stockings, or fludrocortisone. Alleviating the functional abnormalities associated with the autonomic neuropathy is often difficult and frustrating for both doctor and patient. (See Diabetic Neuropathy and Diabetic Lumbosacral Plexopathy.)

Diabetic foot disease

Patients with diabetes who present with wounds, infections, or ulcers of the foot should be treated intensively.[133] In addition to appropriate use of antibiotics, the use of crutches, wheelchairs, or bed rest is mandatory for preventing further trauma to the healing foot. Patients should be treated by a podiatrist or an orthopedist with experience in the care of diabetic foot disease. (See Diabetic Foot and Diabetic Foot Infections.)

If bone or tendon is visible, osteomyelitis is present, and hospitalization for IV antibiotic therapy is often necessary. Many patients need a vascular evaluation in conjunction with local treatment of the foot ulcer because a revascularization procedure may be required to provide adequate blood flow for wound healing.

Because ulcers and foot infections are difficult to cure, their prevention is extremely important.[134] At one clinic, the rate of amputation was halved after patients were required to remove their shoes and socks at every visit. The emergency physician can facilitate this practice by briefly inspecting the feet of patients with diabetes and by educating them about the need for proper foot care.

Referral to a podiatrist is indicated for diabetic patients with any of the following:

Charcot joint, a type of arthropathy observed in people with diabetes, is a progressive deterioration of foot joints caused by underlying neuropathy. Tarsometatarsal and midtarsal joints are affected most commonly. Other neuromuscular foot deformities also may be present. Early diagnosis and treatment are important for preventing further joint degeneration.

Macrovascular disease

Hypercholesterolemia and hypertension increase the risk of specific late complications and require special attention and appropriate treatment. Although physicians can safely use beta blockers (eg, propranolol) in most patients, these agents can mask the adrenergic symptoms of insulin-induced hypoglycemia and can impair the normal counterregulatory response. ACE inhibitors are the drugs of choice for hypertension because of their renal protective action, especially early in the course of the disease.

The ADA advises that a systolic blood pressure below 130 mm Hg is an appropriate goal for most patients with diabetes and hypertension, but it also recommends modifying systolic blood pressure targets in accordance with individual patient characteristics. Diastolic blood pressure should be less than 80 mm Hg.[5]

Subtle differences in the pathophysiology of atherosclerosis in patients with diabetes result in both earlier development and a more malignant course. Therefore, lipid abnormalities must be treated aggressively to reduce the risk of serious atherosclerosis.[135] This is important from an epidemiologic point of view and has a bearing on the treatment strategies that must be used to mitigate the risk.[136]

Prediction of cardiovascular risk in diabetic patients on the basis of the lipid profile is not affected by the timing of blood specimen. Therefore, it may be unnecessary to insist on using fasting blood samples to determine the lipid profile.[137]

In a study involving diabetic adolescents and children, nocturnal hypertension was significantly associated with higher daytime blood pressure and carotid intima-media thickness, which could be precursors of atherosclerotic cardiovascular disease later in life; these findings warrant confirmation and longitudinal follow-up.[138]

Patients with diabetes may have increased incidence of silent ischemia.[65] However, silent ischemia is common in many patients with CAD, and the apparent increase in its incidence may come about because patients with diabetes are more likely than others to have CAD to begin with. Nevertheless, it is prudent to perform electrocardiography (ECG) in patients who have diabetes and a serious illness or who present with generalized weakness, malaise, or other nonspecific symptoms that are not usually expected to result from myocardial ischemia.

Persistent lipid abnormalities remain in patients with diabetes, despite evidence supporting the benefits of lipid-modifying drugs. Up-titration of the statin dose and addition of other lipid-modifying agents are needed.[139] Although metformin is used principally in type 2 DM because of its lipid-lowering effect, a placebo-controlled study by Lund et al found that metformin (1000 mg orally twice daily) significantly reduced total cholesterol and low-density lipoprotein (LDL) cholesterol in patients with type 1 DM.[140]

The American Diabetes Association (ADA) provided recommendations on the use of statins in patients with diabetes to align with those of the American College of Cardiology and the American Heart Association.[141]

Glycemic Control During Serious Medical Illness and Surgery

Serious medical illness and surgery produce a state of increased insulin resistance and relative insulin deficiency. Hyperglycemia can occur even in patients without diabetes as a consequence of stress-induced insulin resistance coupled with the use of dextrose-containing IV fluids. Increases in glucagon, catecholamines, cortisol, and growth hormone levels antagonize the effects of insulin, and the alpha-adrenergic effect of increased catecholamine levels inhibits insulin secretion. Counterregulatory hormones also directly increase hepatic gluconeogenesis.

Much less is known about optimal blood glucose levels in hospitalized patients with preexisting diabetes whose hyperglycemia reflects both their diabetes and a stress response to illness. Nonetheless, it is clear that management of hospitalized patients with preexisting diabetes requires modification of treatment regimens to compensate for both the decreased caloric intake and the increased physiologic stress. Near-normal blood glucose levels should be maintained in medical and surgical patients with diabetes, for the following reasons:

Patients with type 1 DM must take in insulin and carbohydrate at all times to prevent ketosis. It is strongly recommended that continuous IV infusions of dextrose and insulin be used in patients who are undergoing general anesthesia or who are critically ill.

Blood glucose levels must be measured with a glucose meter every hour, and the rates of insulin and dextrose infusion must be adjusted accordingly to prevent hypoglycemia or persistent hyperglycemia.[142] Algorithms are available for insulin infusions, and the use of a preprinted order facilitates administration and reduces dosing errors.

For patients who are less seriously ill or are undergoing minor surgery, frequent blood glucose monitoring is not always possible. These patients may do as well with subcutaneously injected insulin. A basal bolus insulin regimen, rather than a sliding-scale regular insulin regimen, should be used in these patients.

The same principles of providing a constant source of insulin and carbohydrate apply to patients with type 1 DM who must also take nothing by mouth for medical reasons. Patients should receive a basal insulin (eg, glargine or detemir insulin) with additional correction doses of regular insulin or a rapid-acting insulin. In many localities, regular insulin has been replaced by rapid-acting insulin (eg, lispro, aspart, or glulisine)

To prevent hypoglycemia, regular insulin should not be given more often than every 3-4 hours, because a dose is effective for up to 6 hours. Rapid-acting insulins may be given every 3 hours. Once the patient is eating, a preprandial insulin dose can be added.

Cardiovascular disease or renal dysfunction increases surgical morbidity and mortality, and diabetic autonomic neuropathy increases the risk of cardiovascular instability. The emergency physician caring for patients with diabetes who require emergency surgery must notify the surgeon and the anesthesiologist of the patient’s condition, consult medical specialists when appropriate, and promptly initiate a thorough medical evaluation.

Recent guidelines have trended away from stressing intensive glucose control in ill patients with diabetes. The ADA recommends that in critically ill patients, insulin therapy should be initiated if the glucose level exceeds 180 mg/dL (10 mmol/L), with a target range of 140-180 mg/dL (7.8-10 mmol/L) for the majority of critically ill patients.[5] More stringent goals, such as 110-140 mg/dL (6.1-7.8 mmol/L), may be appropriate for selected patients, provided that significant hypoglycemia can be avoided.

In the absence of clear evidence for specific blood glucose goals in non–critically ill patients, the ADA suggests that reasonable targets are premeal blood glucose levels lower than 140 mg/dL (7.8 mmol/L) with random blood glucose levels below 180 mg/dL (10.0 mmol/L), provided that these targets can be safely achieved.[5] It may be appropriate to use more stringent targets in stable patients with previous tight glycemic control and less stringent targets in patients with severe comorbidities.

The guidelines on glycemic control in hospitalized patients formulated by the American College of Physicians (ACP) recommend a target blood glucose level of 140-200 mg/dL if insulin therapy is used to manage patients with diabetes in nonsurgical (medical) intensive care units (ICUs).[143] These guidelines were based on a review of 21 trials in intensive care, perioperative care, myocardial infarction, stroke, or brain injury settings.[144]

The ACP found no convincing evidence that intensive insulin therapy reduced short-term or long-term mortality, infection rates, length of hospital stay, or the need for renal replacement therapy. In recommending 200 mg/dL as the upper target, the ACP guidelines depart from the 2009 AACE/ADA consensus statement on inpatient glycemic control, which recommended a target range of 140-180 mg/dL in critically ill patients.[145]

Nevertheless, in certain circumstances, such as after cardiovascular surgery and during treatment in a surgical ICU, it is very important to maintain near-normal blood glucose levels in patients with acute hyperglycemia of illness. These patients should receive sufficient insulin to maintain glucose levels around 100 mg/dL.[146]

Perioperative blood glucose management

Surgical procedures—including the preoperative emotional stress and the effects of general anesthesia as well as the trauma of the procedure itself—can markedly increase plasma glucose levels and induce DKA in patients with type 1 DM. (See Perioperative Management of the Diabetic Patient.) In patients going to surgery who have not received a dose of intermediate-acting insulin that day, injection of one third to one half of the total daily dose as NPH insulin or 80% of the dose as glargine or detemir insulin before surgery is often effective.

At the same time, an IV infusion containing 5% glucose in either 0.9% saline solution or water should be started at a rate of 1 L (50 g glucose) over 6-8 hours (or 125-150 mL/h). Blood glucose levels should be checked every 2 hours during the surgical procedure, and small doses of regular or rapid-acting insulin (eg, lispro, aspart, or glulisine) should be given if values exceed 140 mg/dL.

After the operation, check plasma glucose levels and assess for a reaction to ketones. Unless a change in dosage is indicated, repeat the preoperative dose of insulin when the patient recovers from the anesthesia, and continue the glucose infusion.

Monitor plasma glucose and ketones at 2- to 4-hour intervals, and administer regular insulin every 4-6 hours as needed to maintain the plasma glucose level in the range of 100-250 mg/dL (ie, 5.55-13.88 mmol/L). Continue until the patient can be switched to oral feedings and a 2- or 3-dose insulin schedule.

Some physicians prefer to withhold subcutaneous insulin on the day of the operation and to add 6-10 units of regular insulin to 1 L of 5% glucose in normal saline or water infused at 150 mL/h on the morning of the operation, depending on the plasma glucose level. The infusion is continued through recovery, with insulin adjustments depending on the plasma glucose levels obtained in the recovery room and at 2- to 4-hour intervals thereafter.

Postoperative IV insulin infusion after major surgical procedures is currently considered the standard of care in most hospitals.

Glycemic Control During Pregnancy

Because pregnant patients with type 1 DM are at risk for multiple poor maternal and fetal outcomes, it is essential to provide these patients with prepregnancy counseling, good glycemic control before and during pregnancy, and a complete medical evaluation. (See Diabetes Mellitus and Pregnancy.) High-risk possibilities include exacerbation of existing hypertension, renal insufficiency, retinopathy, and more frequent congenital anomalies. These patients should be referred to obstetricians specializing in high-risk pregnancies.

Despite advanced age, multiparity, obesity, and social disadvantage, patients with type 2 DM were found to have better glycemic control, fewer large-for-gestational-age infants, fewer preterm deliveries, and fewer neonatal care admissions than patients with type 1 DM.[147] This finding suggests that better tools are needed to improve glycemic control in patients with type 1 DM.

Prevention

Significant improvements in the prediction of type 1 DM have led to several trials of prevention. These include the Diabetes Prevention Trial–Type 1 (DPT-1) in the United States and the European Nicotinamide Diabetes Intervention Trial (ENDIT) in Europe and North America. Both trials have reported disappointing results.

In DPT-1, parenteral insulin failed to delay or prevent type 1 DM in subjects at elevated risk (as indicated by family history and the presence of islet cell antibodies). These subjects received low-dose subcutaneous Ultralente insulin twice daily, plus annual 4-day continuous IV infusions of insulin.[148] DPT-1 subjects who received oral insulin experienced considerable delays in the onset of diabetes, but once therapy was stopped, their rate of developing diabetes increased to a rate similar to that seen in the placebo group.[149]

In the ENDIT study, nicotinamide (which prevents autoimmune diabetes in animal models) did not prevent or delay the clinical onset of diabetes in people with a first-degree family history of type 1 DM. Subjects in the treatment arm received oral modified-release nicotinamide in a dose of 1.2 g/m2.[150]

Slowing progress of recent-onset type 1 DM

In animal models of autoimmunity, treatment with a target antigen can modulate aggressive autoimmunity. However, a trial of antigen-based immunotherapy with 2 or 3 doses of glutamic acid decarboxylase formulated with aluminum hydroxide (GAD-alum) vaccine for 4-12 weeks in patients with newly diagnosed type 1 DM did not alter the course of loss of insulin secretion during the first year.[151]

A phase 3 trial using an anti-CD3 monoclonal antibody, teplizumab, found an encouraging trend toward preservation of beta-cell function with reduction in daily insulin requirements in patients with recently diagnosed type 1 DM. However, rash was almost 3 times more common in treated patients than in those receiving placebo.[152]

A study by Orban et al found that costimulation modulation of activated T cells with abatacept slowed reduction in beta-cell function over a 2-year period of administration. However, this effect was reduced after 6 months of treatment, suggesting that T-cell activation lessens over time. Further studies are needed.[153]

Consultations

Patients with type 1 DM should be referred to an endocrinologist for multidisciplinary management. They should also undergo a complete retinal examination by an ophthalmologist at least once a year. Those patients with significant proteinuria or a reduced creatinine clearance should be referred to a nephrologist. Patients with significant foot involvement should see a podiatrist.

Guidelines Summary

ADA: Position statement on type 1 diabetes in children and adolescents

In August 2018, the American Diabetes Association released a position statement on type 1 diabetes in children and adolescents, which included the following guidelines[154, 155] :

ADA: Standards of Medical Care in Diabetes

The American Diabetes Association’s Standards of Medical Care in Diabetes-2018 include the following A-grade recommendations, ie, recommendations based on “[c]lear evidence from well-conducted, generalizable randomized controlled trials that are adequately powered”[76] :

ADA: hypertension guidelines

Guidelines published in 2017 by the American Diabetes Association on managing hypertension in patients with diabetes state the following[156, 157] :

ISPAD: Diabetic vascular complications in children and adolescents

In August 2018, the International Society for Pediatric and Adolescent Diabetes (ISPAD) released clinical practice consensus guidelines on diabetic microvascular and macrovascular complications in children and adolescents. These include the following[158] :

ISPAD: Glycemic control targets and glucose monitoring in children, adolescents, and young adults

In July 2018, the ISPAD released clinical practice consensus guidelines on glycemic control targets and glucose monitoring in children, adolescents, and young adults with diabetes. These include the following[159] :

Endocrine Society guidelines on diabetes management in older adults 

In 2019, the Endocrine Society released the following clinical practice guidelines on the diagnosis and management of diabetes and its comorbidities in older adults[160, 161] :

 

Medication Summary

Insulin injected subcutaneously is the first-line treatment of type 1 diabetes mellitus (DM). The different types of insulin vary with respect to onset and duration of action. Short-, intermediate-, and long-acting insulins are available. Short-acting and rapid-acting insulins are the only types that can be administered intravenously (IV). Human insulin currently is the only species of insulin available in the United States; it is less antigenic than the previously used animal-derived varieties.

Insulin aspart (NovoLog, Fiasp)

Clinical Context:  Insulin aspart has a rapid onset of action, 5-15 minutes. The peak effect occurs within 30-90 minutes, and the usual duration of action is 2-4 hours. Insulin aspart is approved by the US Food and Drug Administration (FDA) for use in insulin pumps.

Fiasp also has a rapid onset of action, with its first measurable effect occurring within 16-20 minutes. The peak effect occurs within 91-133 minutes, and the usual duration of action is 5-7 hours.

Insulin glulisine (Apidra)

Clinical Context:  Insulin glulisine has a rapid onset of action, 5-15 minutes. The peak effect occurs within 30-90 minutes, and the usual duration of action is 2-4 hours. Insulin glulisine is FDA-approved for use in insulin pumps.

Insulin lispro (Humalog)

Clinical Context:  Insulin lispro has a rapid onset of action, 5-15 minutes. The peak effect occurs within 30-90 minutes, and the usual duration of action is 2-4 hours.

Insulin inhaled (Afrezza)

Clinical Context:  Orally inhaled rapid-acting insulin in powder form. When 8 units were administered, maximum serum insulin concentration was reached by 12-15 minutes and declined to baseline by about 180 minutes.

Regular insulin (Humulin R, Novolin R, Myxredlin)

Clinical Context:  Regular insulin has a short onset of action, 0.5 hour. Its peak effect occurs within 2-4 hours, and its usual duration of action is 5-8 hours. Preparations that contain a mixture of 70% neutral protamine Hagedorn (NPH) insulin and 30% regular human insulin (eg, Novolin 70/30 and Humulin 70/30) are available, but the fixed ratios of intermediate-acting to rapid-acting insulin may restrict their use.

Insulin detemir (Levemir)

Clinical Context:  Insulin detemir is indicated for once-daily or twice-daily subcutaneous administration in individuals with type 1 DM who require long-acting basal insulin for hyperglycemia control. Its duration of action ranges from 5.7 hours (low dose) to 23.2 hours (high dose). The prolonged action results from slow systemic absorption of detemir molecules from the injection site. Its primary activity is regulation of glucose metabolism.

Insulin detemir binds to insulin receptors and lowers blood glucose levels by facilitating cellular uptake of glucose into skeletal muscle and fat; it also inhibits glucose output from the liver. The drug inhibits lipolysis in adipocytes, inhibits proteolysis, and enhances protein synthesis.

Insulin degludec (Tresiba)

Clinical Context:  Ultralong-acting basal insulin indicated to improve glycemic control in adults with diabetes mellitus who require basal insulin. It is highly protein bound, and following SC, the protein-binding provides a depot effect. The elimination half-life is 25 h and its duration of action is beyond 42 h.

Insulin aspart protamine/insulin aspart (NovoLog 70/30)

Clinical Context:  The combination of insulin aspart protamine with insulin aspart includes 30% rapid-onset insulin (ie, insulin aspart) and 70% intermediate-acting insulin (ie, insulin aspart protamine). Insulin aspart is absorbed more rapidly than regular human insulin, and insulin aspart protamine has a prolonged absorption profile after injection.

Insulin lispro protamine/insulin lispro (Humalog 75/25)

Clinical Context:  The combination of insulin lispro protamine with insulin lispro includes 75% insulin lispro protamine, which has a prolonged duration of action, and 25% insulin lispro, which is a rapid-onset insulin.

Insulin degludec/insulin aspart (Ryzodeg)

Clinical Context:  Combines the ultralong-acting basal insulin (degludec 70 units) and a rapid-acting insulin (aspart 30 units). It is indicated to improve glycemic control in adults with diabetes mellitus.

Insulin glargine (Lantus, Lantus SoloStar, Toujeo, Toujeo Max, Basaglar)

Clinical Context:  Insulin glargine stimulates proper utilization of glucose by the cells and reduces blood sugar levels. It has no pronounced peaks of action, because a small amount of insulin is gradually released at a constant rate over 24 hours. The amount of insulin in Toujeo and Toujeo Max SoloStar is three times greater (300 Units/mL) than in Lantus or Basaglar (100 Units/mL).

Class Summary

Rapid-acting insulins are used whenever a rapid onset and short duration are appropriate (eg, before meals or when the blood glucose level exceeds target and a correction dose is needed). Rapid-acting insulins are associated with less hypoglycemia than regular insulin.

Currently, short-acting insulins are less commonly used than the rapid-acting insulins in patients with type 1 DM. They are used when a slightly slower onset of action or a greater duration of action is desired.

Intermediate-acting insulins have a relatively slow onset of action and a relatively long duration of action. They are usually combined with faster-acting insulins to maximize the benefits of a single injection.

Long-acting and ultralong-acting insulins have a very long duration of action and, when combined with faster-acting insulins, provide better glucose control for some patients. In patients with type 1 DM, they must be used in conjunction with a rapid-acting or short-acting insulin given before meals.

Premixed insulins contain a fixed ratio of rapid-acting insulins with longer-acting insulin, which can restrict their use. Premixed insulin is usually not recommended in type 1 DM patients, because of their need for frequent adjustments of premeal insulin doses.

Pramlintide acetate (Symlin)

Clinical Context:  Pramlintide acetate is a synthetic analogue of human amylin, a naturally occurring hormone made in pancreatic beta cells that is deficient in people with type 1 DM. It slows gastric emptying, suppresses postprandial glucagon secretion, and regulates food intake through centrally mediated appetite modulation.

Class Summary

These amylinomimetic agents elicit endogenous amylin effects by delaying gastric emptying, decreasing postprandial glucagon release, and modulating appetite.

Glucagon (GlucaGen, Gvoke)

Clinical Context:  Glucagon elevates blood glucose levels by inhibiting glycogen synthesis and enhancing the formation of glucose from noncarbohydrate sources such as proteins and fats (gluconeogenesis). It increases hydrolysis of glycogen to glucose in the liver and accelerates hepatic glycogenolysis and lipolysis in adipose tissue. Glucagon also increases the force of contraction in the heart and has a relaxant effect on the gastrointestinal tract. It is available in a reconstitutable powder form. Glucagon is also available as a ready-to-use subcutaneous (SC) solution in prefilled syringes or an autoinjector.

Glucagon intranasal (Baqsimi)

Clinical Context:  This agent activates hepatic glucagon receptors, which stimulate cyclic adenosine monophosphate (cAMP) synthesis. Hepatic glycogenolysis and gluconeogenesis are thus accelerated, with blood glucose levels consequently increasing. Glucagon requires preexisting hepatic glycogen stores to effectively treat hypoglycemia. Glucagon intranasal is indicated for severe hypoglycemic reactions in adults and children (aged 4 years or older) with diabetes.

Class Summary

Pancreatic alpha cells of the islets of Langerhans produce glucagon, a polypeptide hormone. Glucagon increases blood glucose levels by promoting hepatic glycogenolysis and gluconeogenesis.

When is screening indicated for type 1 diabetes mellitus (DM)?What is type 1 diabetes mellitus (DM)?What are the classic symptoms of type 1 diabetes mellitus (DM)?What are non-classic symptoms of type 1 diabetes mellitus (DM)?How might type 1 diabetes mellitus (DM) initially present?What are the ADA diagnostic criteria for type 1 diabetes mellitus (DM)?Which lab tests are performed in the diagnosis of type 1 diabetes mellitus (DM)?How are glycemic goals established for type 1 diabetes mellitus (DM)?How are blood glucose levels monitored in type 1 diabetes mellitus (DM)?When is insulin therapy indicated for type 1 diabetes mellitus (DM)?What are common insulin regimens used for the treatment of type 1 diabetes mellitus (DM)?What is the role of diet in the treatment of type 1 diabetes mellitus (DM)?What is the role of exercise in the treatment of type 1 diabetes mellitus (DM)?What is type 1 diabetes mellitus (DM)?Which age groups are at highest risk for type 1 diabetes mellitus (DM)?How are type 1 and type 2 diabetes mellitus (DM) differentiated?What is the approach to treatment of type 1 diabetes mellitus (DM)?What is the medical cost of type 1 diabetes mellitus (DM) in the US?What is the pathophysiology of type 1 diabetes mellitus (DM)?What is the role of autoimmunity in the pathogenesis of type 1 diabetes mellitus (DM)?Which autoimmune diseases are associated with type 1 diabetes mellitus (DM)?What is the role of pancreatic volume in the pathogenesis of type 1 diabetes mellitus (DM)?What are the genetic determinants of type 1 diabetes mellitus (DM)?What is the cause of sensory and autonomic neuropathy in type 1 diabetes mellitus (DM)?What is the association between microvascular disease and type 1 diabetes mellitus (DM)?What is the source of renal lesions in type 1 diabetes mellitus (DM)?What is the significance of vitamin D deficiency in type 1 diabetes mellitus (DM)?What is the pathogenesis of nephropathy in type 1 diabetes mellitus (DM)?What is double diabetes?What is the significance of low estimated glucose disposal rate (eGDR) in type 1 diabetes mellitus (DM)?What causes type 1 diabetes mellitus (DM)?What is the sibling risk for type 1 diabetes mellitus (DM)?What is the parent-to-child risk for type 1 diabetes mellitus (DM)?How does the frequency of type 1 diabetes mellitus (DM) vary geographically?Which DR-DQ haplotypes increase the risk for type 1 diabetes mellitus (DM)?Which DR-DQ haplotypes decrease the risk for type 1 diabetes mellitus (DM)?Which DR-DQ haplotypes confers the highest risk for type 1 diabetes mellitus (DM)?Which ethnic groups carry high risk DR-DQ haplotypes for type 1 diabetes mellitus (DM)?Which genes are associated with susceptibility to type 1 diabetes mellitus (DM)?What are the nongenetic risk factors for type 1 diabetes mellitus (DM)?What is the relationship between maternal age and the susceptibility to type 1 diabetes mellitus (DM)?What is the association between vitamin D and type 1 diabetes mellitus (DM)?What is the association between upper respiratory infection and type 1 diabetes mellitus (DM)?What is the prevalence and incidence of type 1 diabetes mellitus (DM) in the US?What is the global prevalence of type 1 diabetes mellitus (DM)?How does the incidence of type 1 diabetes mellitus (DM) vary by age?How does the prevalence of type 1 diabetes mellitus (DM) vary by sex?How does the prevalence of type 1 diabetes mellitus (DM) vary by race?What are the morbidities of type 1 diabetes mellitus (DM)?What factors affect the long-term prognosis of type 1 diabetes mellitus (DM)?What are complications of type 1 diabetes mellitus (DM)?How does coronary vasodilator dysfunction affect the prognosis of type 1 diabetes mellitus (DM)?What is the association between type 1 diabetes mellitus (DM) and small-fiber neuropathy?What are the mortality differences between early-onset and late-onset type 1 diabetes mellitus (DM)?Which factors are indicators of poorer prognosis in type 1 diabetes mellitus (DM)?What is the role of patient education in the management of type 1 diabetes mellitus (DM)?What education about type 1 diabetes mellitus (DM) should patients be given at every clinical visit?What are the ADA recommendations for patient education of older adolescents with type 1 diabetes mellitus (DM)?What is key information to include in patient education about type 1 diabetes mellitus (DM)?What education should dietitians give patients with type 1 diabetes mellitus (DM)?What are the most common symptoms of type 1 diabetes mellitus (DM)?What causes polyuria in type 1 diabetes mellitus (DM)?What causes symptoms of type 1 diabetes mellitus (DM)?How soon after beta-cell destruction do patients with type 1 diabetes mellitus (DM) become symptomatic?How is the onset of symptomatic type 1 diabetes mellitus (DM) characterized?What are the signs and symptoms of type 1 diabetes mellitus (DM)?What are the GI symptoms of type 1 diabetes mellitus (DM)?What is frequency of neuropathy in type 1 diabetes mellitus (DM)?What is the focus of medical history of a patient with type 1 diabetes mellitus (DM)?What history is needed in patients with type 1 diabetes mellitus (DM) who use a pump or a multiple-injection regimen?What information should be included in a type 1 diabetes mellitus (DM) history?How is glycemic exposure in established type 1 diabetes mellitus (DM) assessed?Which questions regarding hypoglycemia and hyperglycemia symptoms should be asked of patients with type 1 diabetes mellitus (DM)?Which questions regarding microvascular complications should be asked of patients with type 1 diabetes mellitus (DM)?Which questions regarding macrovascular complications should be asked of patients with type 1 diabetes mellitus (DM)?Which questions regarding neuropathy should be asked of patients with type 1 diabetes mellitus (DM)?Which questions regarding foot disease should be asked of patients with type 1 diabetes mellitus (DM)?What are the physical findings characteristic of new-onset type 1 diabetes mellitus (DM)?How often should patients with type 1 diabetes mellitus (DM) be examined?What is included in a diabetes-focused physical exam?How are the vital signs assessed in type 1 diabetes mellitus (DM)?What is included in the funduscopic exam for type 1 diabetes mellitus (DM)?What is included in the foot exam for type 1 diabetes mellitus (DM)?Which factors increase the risk of infection in type 1 diabetes mellitus (DM)?What are the most common sites of infection in type 1 diabetes mellitus (DM)?Which infections occur almost exclusively in type 1 diabetes mellitus (DM)?What are ophthalmologic complications of type 1 diabetes mellitus (DM)?What is diabetic retinopathy?What are the stages in the progression of diabetic retinopathy?How does diabetic retinopathy progress in stage 1 and 2?How does diabetic retinopathy progress in stages 3, 4 and 5?What are renal complications of type 1 diabetes mellitus (DM)?What are the types of diabetic neuropathy?What is the most common type of diabetic neuropathy?What are acute-onset mononeuropathies of type 1 diabetes mellitus (DM)?What are cranial nerve complications of type 1 diabetes mellitus (DM)?What are macrovascular complications of type 1 diabetes mellitus (DM)?What are the risk factors for macrovascular disease in type 1 diabetes mellitus (DM)?Which conditions and factors should be considered in the differential diagnosis of type 1 diabetes mellitus (DM)?How are type 1 and type 2 diabetes mellitus (DM) differentiated?How is monogenic diabetes differentiated from type 1 diabetes mellitus (DM)?What are the differential diagnoses for Type 1 Diabetes Mellitus?What is the role of HbA1c measurement in the diagnosis of type 1 diabetes mellitus (DM)?What is the role of plasma glucose concentration in the diagnosis of type 1 diabetes mellitus (DM)?What is the role of fingerstick glucose test in the diagnosis of type 1 diabetes mellitus (DM)?How are glucose levels determined in the diagnosis of type 1 diabetes mellitus (DM)?What are the ADA recommendations for the measurement of HbA1c in type 1 diabetes mellitus (DM)?What are limitations of HbA1c levels in the diagnosis of type 1 diabetes mellitus (DM)?What is an HbA1c assay indicated in the diagnosis of type 1 diabetes mellitus (DM)?What are the advantages of HbA1c testing over glucose measurement for the diagnosis of type 1 diabetes mellitus (DM)?What is the HbA1c criterion for diagnosis of type 1 diabetes mellitus (DM)?What effect does seasonal variability have on HbA1c levels used for diagnosis of type 1 diabetes mellitus (DM)?What is the role of HbF in the diagnosis of type 1 diabetes mellitus (DM), and what are the concerns regarding the accuracy of HbA1c testing?What are the ADS guidelines for use of HbA1c in the management of type 1 diabetes mellitus (DM)?What is the role of fructosamine assay in the diagnosis of type 1 diabetes mellitus (DM)?What is the role of WBC count and blood and urine cultures in the diagnosis of type 1 diabetes mellitus (DM)?How is diabetic ketoacidosis (DKA) diagnosed?Who should be screened for type 1 diabetes mellitus (DM)?What are the AACE recommendations for distinguishing between type 1 and type 2 diabetes mellitus (DM)?What are some classical symptoms of type 1 diabetes mellitus (DM)?What is the role of islet-cell (IA2) levels in the diagnosis of type 1 diabetes mellitus (DM)?What is included in insulin therapy for type 1 diabetes mellitus (DM)?When is little or no insulin therapy needed for type 1 diabetes mellitus (DM)?What is the approach for treatment of new-onset type 1 diabetes mellitus (DM)?What are the American Disease Association (ADA) treatment guidelines for type 1 diabetes mellitus (DM)?What is the role of exogenous glucagonlike peptide 1 (GLP-1) in the treatment of type 1 diabetes mellitus (DM)?When is pancreatic transplantation indicated for the treatment of type 1 diabetes mellitus (DM)?What health risks are decreased with tight glycemic control in type 1 diabetes mellitus (DM)?What are the benefits to tight glycemic control in type 1 diabetes mellitus (DM)?How are HbA1c target levels determined in the treatment of type 1 diabetes mellitus (DM)?Which health risks increase with tight glycemic control of type 1 diabetes mellitus (DM)?What is the role of self-monitoring in the management of type 1 diabetes mellitus (DM)?How frequently should plasma glucose be monitored in type 1 diabetes mellitus (DM)?When is urine ketone testing indicated in type 1 diabetes mellitus (DM)?What is the role of continuous glucose monitors (CGMs) in the treatment of type 1 diabetes mellitus (DM)?What are the limitations of continuous glucose monitors (CGMs) for the treatment of type 1 diabetes mellitus (DM)?What is the benefit of continuous glucose monitors (CGMs) for the treatment of type 1 diabetes mellitus (DM)?When is the intermittent use of a continuous glucose monitor (CGM) indicated in the treatment of type 1 diabetes mellitus (DM)?Which continuous glucose monitor (CGM) is the most accurate for type 1 diabetes mellitus (DM)?What is the sensor-augmented insulin pump system for type 1 diabetes mellitus (DM)?Which continuous glucose monitors (CGMs) have recently been FDA approved for the management of type 1 diabetes mellitus (DM)?What is the role of artificial pancreases in the treatment of type 1 diabetes mellitus (DM)?What types of insulin preparations are used in the treatment of type 1 diabetes mellitus (DM)?What is the role of rapid-acting insulins in the treatment of type 1 diabetes mellitus (DM)?What is the efficacy of rapid-acting inhaled insulin powder (Afrezza) in the treatment of type 1 diabetes mellitus (DM)?What is the role of short-acting insulins in the treatment of type 1 diabetes mellitus (DM)?Which type of insulin is the most effective for lowering postprandial hyperglycemia in type 1 diabetes mellitus (DM)?What is the role of aspart (Fiasp) in the treatment of type 1 diabetes mellitus (DM)?What is the role of Semilente insulin in the treatment of type 1 diabetes mellitus (DM)?Which intermediate-acting insulins are used for the treatment of type 1 diabetes mellitus (DM)?What is the role of lente insulin in the treatment of type 1 diabetes mellitus (DM)?Which long-acting insulins are used for the treatment of type 1 diabetes mellitus (DM)?What is the role of degludec (Tresiba) in the treatment of type 1 diabetes mellitus (DM)?How are insulins mixed in the treatment of type 1 diabetes mellitus (DM)?What is the role of ultrafast-acting insulin in the treatment of type 1 diabetes mellitus (DM)?What is the evidence of increased cancer risk from insulin glargine (Lantus) treatment of type 1 diabetes mellitus (DM)?How is insulin administered for the treatment of type 1 diabetes mellitus (DM)?What are some common insulin regimens for the treatment of type 1 diabetes mellitus (DM)?How quickly does insulin used for the treatment of type 1 diabetes mellitus (DM) expire?How is the initial daily insulin dose calculated for type 1 diabetes mellitus (DM)?How is the initial daily insulin dose calculated for children with type 1 diabetes mellitus (DM)?What are the risks and benefits of administering multiple subcutaneous insulin injections for type 1 diabetes mellitus (DM)?Which types of insulin are administered for the treatment of type 1 diabetes mellitus (DM)?How should daily doses of insulin be adjusted in the treatment of type 1 diabetes mellitus (DM)?How are small battery-operated infusion pumps used in the treatment of type 1 diabetes mellitus (DM)?What is the role of continuous subcutaneous insulin infusion (CSII) in the treatment of type 1 diabetes mellitus (DM)?What are the benefits of pump therapy for the treatment of type 1 diabetes mellitus (DM)?How can early morning hyperglycemia be prevented in type 1 diabetes mellitus (DM)?How do altitude changes affect insulin pump delivery in the treatment of type 1 diabetes mellitus (DM)?What are the AACE/ACE guidelines for insulin pump management in type 1 diabetes mellitus (DM)?Which are the signs and symptoms of insulin allergy?What are the causes of hypoglycemia in type 1 diabetes mellitus (DM)?What are symptoms of hypoglycemia in type 1 diabetes mellitus (DM)?What are the health risks of repeated hypoglycemia in type 1 diabetes mellitus (DM)?What is the evidence of cognitive impairment as a result of hypoglycemia in type 1 diabetes mellitus (DM)?What are the risks of hyperglycemia in type 1 diabetes mellitus (DM)?What is the treatment for hyperglycemia in type 1 diabetes mellitus (DM)?What is the importance of follow-up following treatment of hyperglycemia in type 1 diabetes mellitus (DM)?Which type 1 diabetes mellitus (DM) comorbidities may aggravate hyperglycemia?What is diabetic ketoacidosis (DKA)?What is the dawn phenomenon in type 1 diabetes mellitus (DM)?What is the Somogyi phenomenon in type 1 diabetes mellitus (DM)?How are the dawn and Somogyi phenomena in type 1 diabetes mellitus (DM) managed?What is the role of insulin in the treatment of hyperglycemia in type 1 diabetes mellitus (DM)?What is the ADA recommendation for dietary treatment of type 1 diabetes mellitus (DM)?What is included in the diet management plan for type 1 diabetes mellitus (DM)?What is the recommended caloric distribution for management of type 1 diabetes mellitus (DM)?What are the recommended food group intakes for patients with type 1 diabetes mellitus (DM)?What are the activity recommendations for management of type 1 diabetes mellitus (DM)?Which infectious diseases are at higher risk in patients with type 1 diabetes mellitus (DM)?What are the ophthalmologic complications of type 1 diabetes mellitus (DM)?What are the dangers of nephrotoxic agents in patients with type 1 diabetes mellitus (DM)?What the alternative to contrast-enhanced imaging for patients with type 1 diabetes mellitus (DM)?What are the ADA recommendations for nephropathy screening in type 1 diabetes mellitus (DM)?What are complications of micro- and macroalbuminuria in type 1 diabetes mellitus (DM)?How is blood pressure controlled in type 1 diabetes mellitus (DM)?What are complications of diabetic neuropathy?What are complications of diabetic foot disease?When is referral to a podiatrist indicated in the treatment of type 1 diabetes mellitus (DM)?What is Charcot joint in type 1 diabetes mellitus (DM)?What are macrovascular disease complications in type 1 diabetes mellitus (DM)?What is the ADA recommended blood pressure goal for comorbid hypertension and type 1 diabetes mellitus (DM)?How is atherosclerosis prevented in type 1 diabetes mellitus (DM)?What is the risk of cardiovascular disease in patients with type 1 diabetes mellitus (DM)?What is the association between silent ischemia and type 1 diabetes mellitus (DM)?How are lipid abnormalities resolved in patients with type 1 diabetes mellitus (DM)?What are the ADA guidelines on the use of statins in patients with type 1 diabetes mellitus (DM)?How is type 1 diabetes mellitus (DM) managed during surgical procedures?What are complication of serious medical illness or surgical procedures in patients with type 1 diabetes mellitus (DM)?Why should near-normal blood glucose levels be maintained in medical and surgical patients with type 1 diabetes mellitus (DM)?How is ketosis prevented in hospitalized patients with type 1 diabetes mellitus (DM)?What insulin regimen should be used for patients with type 1 diabetes mellitus (DM) undergoing minor surgery?When are the different types of insulin indicated for the treatment of type 1 diabetes mellitus (DM) in patients with restricted food intake prior to medical procedures?Which types of insulin should be administered for prevention of hypoglycemia in hospitalized patients with type 1 diabetes mellitus (DM)?What effect does cardiovascular disease or renal dysfunction have on the morbidity and mortality of patients with type 1 diabetes mellitus (DM)?What are the insulin therapy requirements in critically ill patients with type 1 diabetes mellitus (DM)?What are the ADA recommended blood glucose goals for non-critically ill patients with type 1 diabetes mellitus (DM)?What are the ACP guidelines for glycemic control in hospitalized patients with type 1 diabetes mellitus (DM)?How are plasma glucose and ketones monitored in patients with type 1 diabetes mellitus (DM) undergoing surgical procedures?When is insulin administered for type 1 diabetes mellitus (DM) in patients undergoing surgical procedures?What is the postoperative standard of care for type 1 diabetes mellitus (DM)?What are the treatment options for type 1 diabetes mellitus (DM) during pregnancy?Which trials have been conducted on the prevention of type 1 diabetes mellitus (DM)?What were the results of the Diabetes Prevention Trial-Type 1 (DPT-1)?What is the role of nicotinamide in the prevention of type 1 diabetes mellitus (DM)?What is the role of antigen-based immunotherapy in the prevention of type 1 diabetes mellitus (DM)?What is the role of teplizumab in the prevention of type 1 diabetes mellitus (DM)?What is the role of abatacept in the prevention of type 1 diabetes mellitus (DM)?What are the recommended referrals for patients with type 1 diabetes mellitus (DM)?What are the A-grade recommendations from the American Diabetes Association’s Standards of Medical Care in Diabetes 2018?What is contained in the American Diabetes Association (ADA) position statement on type 1 diabetes in children and adolescents?What are the ADA guidelines on managing hypertension in type 1 diabetes mellitus (DM)?What are the ISPAD guidelines on diabetic vascular complications in children and adolescents with type 1 diabetes mellitus (DM)?What are the ISPAD clinical practice consensus guidelines on glycemic control targets and glucose monitoring in type 1 diabetes mellitus (DM)?What is the first-line treatment for type 1 diabetes mellitus (DM)?Which medications in the drug class Hypoglycemia Antidotes are used in the treatment of Type 1 Diabetes Mellitus?Which medications in the drug class Antidiabetics, Amylinomimetics are used in the treatment of Type 1 Diabetes Mellitus?Which medications in the drug class Antidiabetics, Insulins are used in the treatment of Type 1 Diabetes Mellitus?

Author

Romesh Khardori, MD, PhD, FACP, Professor of Endocrinology, Director of Training Program, Division of Endocrinology, Diabetes and Metabolism, Strelitz Diabetes and Endocrine Disorders Institute, Department of Internal Medicine, Eastern Virginia Medical School

Disclosure: Nothing to disclose.

Chief Editor

George T Griffing, MD, Professor Emeritus of Medicine, St Louis University School of Medicine

Disclosure: Nothing to disclose.

Acknowledgements

Howard A Bessen, MD Professor of Medicine, Department of Emergency Medicine, University of California, Los Angeles, David Geffen School of Medicine; Program Director, Harbor-UCLA Medical Center

Howard A Bessen, MD is a member of the following medical societies: American College of Emergency Physicians

Disclosure: Nothing to disclose.

Barry E Brenner, MD, PhD, FACEP Professor of Emergency Medicine, Professor of Internal Medicine, Program Director, Emergency Medicine, Case Medical Center, University Hospitals, Case Western Reserve University School of Medicine

Barry E Brenner, MD, PhD, FACEP is a member of the following medical societies: Alpha Omega Alpha, American Academy of Emergency Medicine, American College of Chest Physicians, American College of Emergency Physicians, American College of Physicians, American Heart Association, American Thoracic Society, Arkansas Medical Society, New York Academy of Medicine, New York Academy ofSciences,and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Aneela Naureen Hussain, MD, FAAFM Assistant Professor, Department of Family Medicine, State University of New York Downstate Medical Center; Consulting Staff, Department of Family Medicine, University Hospital of Brooklyn

Aneela Naureen Hussain, MD, FAAFM is a member of the following medical societies: American Academy of Family Physicians, American Medical Association, American Medical Women's Association, Medical Society of the State of New York, and Society of Teachers of Family Medicine

Disclosure: Nothing to disclose.

Anne L Peters, MD, CDE Director of Clinical Diabetes Programs, Professor, Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, Los Angeles County/University of Southern California Medical Center

Anne L Peters, MD, CDE is a member of the following medical societies: American College of Physicians and American Diabetes Association

Disclosure: Amylin Honoraria Speaking and teaching; AstraZeneca Consulting fee Consulting; Lilly Consulting fee Consulting; Takeda Consulting fee Consulting; Bristol Myers Squibb Honoraria Speaking and teaching; NovoNordisk Consulting fee Consulting; Medtronic Minimed Consulting fee Consulting; Dexcom Honoraria Speaking and teaching; Roche Honoraria Speaking and teaching

Don S Schalch, MD Professor Emeritus, Department of Internal Medicine, Division of Endocrinology, University of Wisconsin Hospitals and Clinics

Don S Schalch, MD is a member of the following medical societies: American Diabetes Association, American Federation for Medical Research, Central Society for Clinical Research, and Endocrine Society

Disclosure: Nothing to disclose.

Erik D Schraga, MD Staff Physician, Department of Emergency Medicine, Mills-Peninsula Emergency Medical Associates

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

Miriam T Vincent, MD, PhD Professor and Chair, Department of Family Practice, State University of New York Downstate Medical Center

Miriam T Vincent, MD, PhD is a member of the following medical societies: Alpha Omega Alpha, American Academy of Family Physicians, American Association for the Advancement of Science, Medical Society of the State of New York, North American Primary Care Research Group, Sigma Xi, and Society of Teachers of Family Medicine

Disclosure: Joslin Diabetes Group, Harvard Honoraria Speaking and teaching

Scott R Votey, MD Director of Emergency Medicine Residency, Ronald Reagan UCLA Medical Center; Professor of Medicine/Emergency Medicine, University of California, Los Angeles, David Geffen School of Medicine

Scott R Votey, MD is a member of the following medical societies: Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Frederick H Ziel, MD Associate Professor of Medicine, University of California, Los Angeles, David Geffen School of Medicine; Physician-In-Charge, Endocrinology/Diabetes Center, Director of Medical Education, Kaiser Permanente Woodland Hills; Chair of Endocrinology, Co-Chair of Diabetes Complete Care Program, Southern California Permanente Medical Group

Frederick H Ziel, MD is a member of the following medical societies: American Association of Clinical Endocrinologists, American College of Endocrinology, American College of Physicians, American College of Physicians-American Society of Internal Medicine, American Diabetes Association, American Federation for Medical Research, American Medical Association, American Society for Bone and Mineral Research, California Medical Association, Endocrine Society, andInternational Society for Clinical Densitometry

Disclosure: Nothing to disclose.

References

  1. Aathira R, Jain V. Advances in management of type 1 diabetes mellitus. World J Diabetes. 2014 Oct 15. 5 (5):689-96. [View Abstract]
  2. [Guideline] Diagnosis and classification of diabetes mellitus. Diabetes Care. 2010 Jan. 33 Suppl 1:S62-9. [View Abstract]
  3. International Expert Committee report on the role of the A1C assay in the diagnosis of diabetes. Diabetes Care. 2009 Jul. 32(7):1327-34. [View Abstract]
  4. Vehik K, Beam CA, Mahon JL, et al. Development of Autoantibodies in the TrialNet Natural History Study. Diabetes Care. 2011 Sep. 34(9):1897-1901. [View Abstract]
  5. [Guideline] American Diabetes Association. Standards of medical care in diabetes--2011. Diabetes Care. 2011 Jan. 34 Suppl 1:S11-61. [View Abstract]
  6. Nainggolan L. Continuous Glucose Monitoring: Navigator Beats Rival Devices. Medscape Medical News. January 14, 2013. Available at http://www.medscape.com/viewarticle/777607. Accessed: January 24, 2013.
  7. Damiano ER, El-Khatib FH, Zheng H, Nathan DM, Russell SJ. A Comparative Effectiveness Analysis of Three Continuous Glucose Monitors. Diabetes Care. 2013 Jan 3. [View Abstract]
  8. Tao B, Pietropaolo M, Atkinson M, Schatz D, Taylor D. Estimating the cost of type 1 diabetes in the U.S.: a propensity score matching method. PLoS One. 2010 Jul 9. 5(7):e11501. [View Abstract]
  9. Pilia S, Casini MR, Cambuli VM, et al. Prevalence of Type 1 diabetes autoantibodies (GAD and IA2) in Sardinian children and adolescents with autoimmune thyroiditis. Diabet Med. 2011 Aug. 28(8):896-9. [View Abstract]
  10. Philippe MF, Benabadji S, Barbot-Trystram L, et al. Pancreatic volume and endocrine and exocrine functions in patients with diabetes. Pancreas. 2011 Apr. 40(3):359-63. [View Abstract]
  11. Noble JA, Valdes AM. Genetics of the HLA region in the prediction of type 1 diabetes. Curr Diab Rep. 2011 Dec. 11(6):533-42. [View Abstract]
  12. Barchetta I, Riccieri V, Vasile M, et al. High prevalence of capillary abnormalities in patients with diabetes and association with retinopathy. Diabet Med. 2011 Sep. 28(9):1039-44. [View Abstract]
  13. Young KA, Snell-Bergeon JK, Naik RG, Hokanson JE, Tarullo D, Gottlieb PA, et al. Vitamin D deficiency and coronary artery calcification in subjects with type 1 diabetes. Diabetes Care. 2011 Feb. 34(2):454-8. [View Abstract]
  14. Joergensen C, Hovind P, Schmedes A, Parving HH, Rossing P. Vitamin d levels, microvascular complications, and mortality in type 1 diabetes. Diabetes Care. 2011 May. 34(5):1081-5. [View Abstract]
  15. Zhang D, Efendic S, Brismar K, Gu HF. Effects of MCF2L2, ADIPOQ and SOX2 genetic polymorphisms on the development of nephropathy in type 1 Diabetes Mellitus. BMC Med Genet. 2010 Jul 28. 11:116. [View Abstract]
  16. Busko M. Phenomenon of 'double diabetes' common among blacks. Medscape Medical News. April 25, 2013.
  17. Epstein EJ, Osman JL, Cohen HW, Rajpathak SN, Lewis O, Crandall JP. Use of the Estimated Glucose Disposal Rate (eGDR) as a Measure of Insulin Resistance in an Urban Multiethnic Population With Type 1 Diabetes. Diabetes Care. 2013 Apr 17. [View Abstract]
  18. Davies JL, Kawaguchi Y, Bennett ST, Copeman JB, Cordell HJ, Pritchard LE, et al. A genome-wide search for human type 1 diabetes susceptibility genes. Nature. 1994 Sep 8. 371(6493):130-6. [View Abstract]
  19. Steck AK, Barriga KJ, Emery LM, Fiallo-Scharer RV, Gottlieb PA, Rewers MJ. Secondary attack rate of type 1 diabetes in Colorado families. Diabetes Care. 2005 Feb. 28(2):296-300. [View Abstract]
  20. Redondo MJ, Jeffrey J, Fain PR, Eisenbarth GS, Orban T. Concordance for islet autoimmunity among monozygotic twins. N Engl J Med. 2008 Dec 25. 359(26):2849-50. [View Abstract]
  21. Borchers AT, Uibo R, Gershwin ME. The geoepidemiology of type 1 diabetes. Autoimmun Rev. 2010 Mar. 9(5):A355-65. [View Abstract]
  22. Diabetes Epidemiology Research International Group. Geographic patterns of childhood insulin-dependent diabetes mellitus. Diabetes Epidemiology Research International Group. Diabetes. 1988 Aug. 37(8):1113-9. [View Abstract]
  23. Erlich H, Valdes AM, Noble J, Carlson JA, Varney M, Concannon P, et al. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families. Diabetes. 2008 Apr. 57(4):1084-92. [View Abstract]
  24. Todd JA, Bell JI, McDevitt HO. HLA-DQ beta gene contributes to susceptibility and resistance to insulin-dependent diabetes mellitus. Nature. 1987 Oct 15-21. 329(6140):599-604. [View Abstract]
  25. Corper AL, Stratmann T, Apostolopoulos V, Scott CA, Garcia KC, Kang AS, et al. A structural framework for deciphering the link between I-Ag7 and autoimmune diabetes. Science. 2000 Apr 21. 288(5465):505-11. [View Abstract]
  26. Erlich H, Valdes AM, Noble J, Carlson JA, Varney M, Concannon P, et al. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families. Diabetes. 2008 Apr. 57(4):1084-92. [View Abstract]
  27. Noble JA, Johnson J, Lane JA, Valdes AM. Race-specific type 1 diabetes risk of HLA-DR7 haplotypes. Tissue Antigens. 2011 Nov. 78(5):348-51. [View Abstract]
  28. Rotwein P, Yokoyama S, Didier DK, Chirgwin JM. Genetic analysis of the hypervariable region flanking the human insulin gene. Am J Hum Genet. 1986 Sep. 39(3):291-9. [View Abstract]
  29. Pugliese A, Zeller M, Fernandez A Jr, Zalcberg LJ, Bartlett RJ, Ricordi C, et al. The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat Genet. 1997 Mar. 15(3):293-7. [View Abstract]
  30. Polychronakos C, Li Q. Understanding type 1 diabetes through genetics: advances and prospects. Nat Rev Genet. 2011 Oct 18. 12(11):781-92. [View Abstract]
  31. Concannon P, Chen WM, Julier C, Morahan G, Akolkar B, Erlich HA, et al. Genome-wide scan for linkage to type 1 diabetes in 2,496 multiplex families from the Type 1 Diabetes Genetics Consortium. Diabetes. 2009 Apr. 58(4):1018-22. [View Abstract]
  32. Yeung WC, Rawlinson WD, Craig ME. Enterovirus infection and type 1 diabetes mellitus: systematic review and meta-analysis of observational molecular studies. BMJ. 2011 Feb 3. 342:d35. [View Abstract]
  33. Paronen J, Knip M, Savilahti E, Virtanen SM, Ilonen J, Akerblom HK, et al. Effect of cow's milk exposure and maternal type 1 diabetes on cellular and humoral immunization to dietary insulin in infants at genetic risk for type 1 diabetes. Finnish Trial to Reduce IDDM in the Genetically at Risk Study Group. Diabetes. 2000 Oct. 49(10):1657-65. [View Abstract]
  34. Lempainen J, Tauriainen S, Vaarala O, Mäkelä M, Honkanen H, Marttila J, et al. Interaction of enterovirus infection and cow's milk-based formula nutrition in type 1 diabetes-associated autoimmunity. Diabetes Metab Res Rev. 2012 Feb. 28(2):177-85. [View Abstract]
  35. Cardwell CR, Stene LC, Joner G, Bulsara MK, Cinek O, Rosenbauer J, et al. Maternal age at birth and childhood type 1 diabetes: a pooled analysis of 30 observational studies. Diabetes. 2010 Feb. 59(2):486-94. [View Abstract]
  36. Henry EB, Patterson CC, Cardwell CR. A meta-analysis of the association between pre-eclampsia and childhood-onset Type 1 diabetes mellitus. Diabet Med. 2011 Aug. 28(8):900-5. [View Abstract]
  37. Simpson M, Brady H, Yin X, et al. No association of vitamin D intake or 25-hydroxyvitamin D levels in childhood with risk of islet autoimmunity and type 1 diabetes: the Diabetes Autoimmunity Study in the Young (DAISY). Diabetologia. 2011 Nov. 54(11):2779-88. [View Abstract]
  38. Melville N. Early Upper-Respiratory Infections Linked to Type 1 Diabetes. Medscape Medical News. Available at http://www.medscape.com/viewarticle/807205. Accessed: July 8, 2013.
  39. Beyerlein A, Wehweck F, Ziegler AG, Pflueger M. Respiratory Infections in Early Life and the Development of Islet Autoimmunity in Children at Increased Type 1 Diabetes Risk: Evidence From the BABYDIET Study. JAMA Pediatr. 2013 Jul 1. [View Abstract]
  40. U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, 2011. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. Available at http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf. Accessed: January 28, 2011.
  41. Mayer-Davis EJ, Lawrence JM, Dabelea D, et al. Incidence Trends of Type 1 and Type 2 Diabetes among Youths, 2002-2012. N Engl J Med. 2017 Apr 13. 376 (15):1419-29. [View Abstract]
  42. Imkampe AK, Gulliford MC. Trends in Type 1 diabetes incidence in the UK in 0- to 14-year-olds and in 15- to 34-year-olds, 1991-2008. Diabet Med. 2011 Jul. 28(7):811-4. [View Abstract]
  43. Thomas NJ, Jones SE, Weedon MN, Shields BM, Oram RA, Hattersley AT. Frequency and phenotype of type 1 diabetes in the first six decades of life: a cross-sectional, genetically stratified survival analysis from UK Biobank. Lancet Diabetes Endocrinol. 2017 Nov 30. [View Abstract]
  44. Tucker ME. Needle in a Haystack: Type 1 Diabetes Arises Equally in Adulthood. Medscape. 2017 Dec 4.
  45. Harjutsalo V, Maric C, Forsblom C, et al. Sex-related differences in the long-term risk of microvascular complications by age at onset of type 1 diabetes. Diabetologia. 2011 Aug. 54(8):1992-1999. [View Abstract]
  46. Murthy VL, Naya M, Foster CR, Gaber M, Hainer J, Klein J, et al. Association Between Coronary Vascular Dysfunction and Cardiac Mortality in Patients with and without Diabetes Mellitus. Circulation. 2012 Aug 23. [View Abstract]
  47. Tucker ME. Small-fiber neuropathy common at 40 years of type 1 diabetes. Medscape Medical News. September 18, 2013.
  48. Sveen KA, Karimé B, Jørum E, Mellgren SI, Fagerland MW, Monnier VM, et al. Small- and Large-Fiber Neuropathy After 40 Years of Type 1 Diabetes: Associations with glycemic control and advanced protein glycation: The Oslo Study. Diabetes Care. 2013 Sep 11. [View Abstract]
  49. Finne P, Reunanen A, Stenman S, Groop PH, Grönhagen-Riska C. Incidence of end-stage renal disease in patients with type 1 diabetes. JAMA. 2005 Oct 12. 294(14):1782-7. [View Abstract]
  50. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, et al. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med. 2005 Dec 22. 353(25):2643-53. [View Abstract]
  51. DCCT/EDIC Research Group, de Boer IH, Sun W, Cleary PA, Lachin JM, Molitch ME, et al. Intensive diabetes therapy and glomerular filtration rate in type 1 diabetes. N Engl J Med. 2011 Dec 22. 365(25):2366-76. [View Abstract]
  52. Harjutsalo V, Forsblom C, Groop PH. Time trends in mortality in patients with type 1 diabetes: nationwide population based cohort study. BMJ. 2011 Sep 8. 343:d5364. [View Abstract]
  53. Purnell JQ, Hokanson JE, Cleary PA, Nathan DM, Lachin JM, Zinman B, et al. The Effect of Excess Weight Gain with Intensive Diabetes Treatment on Cardiovascular Disease Risk Factors and Atherosclerosis in Type 1 Diabetes: Results from the Diabetes Control and Complications Trial / Epidemiology of Diabetes Interventions and Complications Study (DCCT/EDIC) Study. Circulation. 2012 Dec 4. [View Abstract]
  54. Zheng F, Yan L, Yang Z, Zhong B, Xie W. HbA1c, diabetes and cognitive decline: the English Longitudinal Study of Ageing. Diabetologia. 2018 Jan 25. [View Abstract]
  55. Melville NA. HbA1c Levels in Diabetes Linked to Cognitive Decline. Medscape Medical News. 2018 Jan 30.
  56. Joshi N, Caputo GM, Weitekamp MR, Karchmer AW. Infections in patients with diabetes mellitus. N Engl J Med. 1999 Dec 16. 341(25):1906-12. [View Abstract]
  57. Wong VH, Bui BV, Vingrys AJ. Clinical and experimental links between diabetes and glaucoma. Clin Exp Optom. 2011 Jan. 94(1):4-23. [View Abstract]
  58. Gillespie KM. Type 1 diabetes: pathogenesis and prevention. CMAJ. 2006 Jul 18. 175(2):165-70. [View Abstract]
  59. Harris SS. Vitamin D in type 1 diabetes prevention. J Nutr. 2005 Feb. 135(2):323-5. [View Abstract]
  60. Hammes HP, Kerner W, Hofer S, et al. Diabetic retinopathy in type 1 diabetes-a contemporary analysis of 8,784 patients. Diabetologia. 2011 Aug. 54(8):1977-1984. [View Abstract]
  61. Julius MC, Schatz DA, Silverstein JH. The prevention of type I diabetes mellitus. Pediatr Ann. 1999 Sep. 28(9):585-8. [View Abstract]
  62. Vinik AI, Mehrabyan A. Diabetic neuropathies. Med Clin North Am. 2004 Jul. 88(4):947-99, xi. [View Abstract]
  63. Chou KL, Galetta SL, Liu GT, Volpe NJ, Bennett JL, Asbury AK, et al. Acute ocular motor mononeuropathies: prospective study of the roles of neuroimaging and clinical assessment. J Neurol Sci. 2004 Apr 15. 219(1-2):35-9. [View Abstract]
  64. Gerstein HC, Islam S, Anand S, et al. Dysglycaemia and the risk of acute myocardial infarction in multiple ethnic groups: an analysis of 15,780 patients from the INTERHEART study. Diabetologia. 2010 Dec. 53(12):2509-17. [View Abstract]
  65. Falcone C, Nespoli L, Geroldi D, Gazzaruso C, Buzzi MP, Auguadro C, et al. Silent myocardial ischemia in diabetic and nondiabetic patients with coronary artery disease. Int J Cardiol. 2003 Aug. 90(2-3):219-27. [View Abstract]
  66. [Guideline] Handelsman Y, Mechanick JI, Blonde L, Grunberger G, Bloomgarden ZT, Bray GA, et al. American Association of Clinical Endocrinologists Medical Guidelines for Clinical Practice for developing a diabetes mellitus comprehensive care plan. Endocr Pract. 2011 Mar-Apr. 17 Suppl 2:1-53. [View Abstract]
  67. [Guideline] Hattersley A, Bruining J, Shield J, Njolstad P, Donaghue KC. The diagnosis and management of monogenic diabetes in children and adolescents. Pediatr Diabetes. 2009 Sep. 10 Suppl 12:33-42. [View Abstract]
  68. Hemoglobin A1c and Mean Glucose in Patients With Type 1 Diabetes: Analysis of data from the Juvenile Diabetes Research Foundation continuous glucose monitoring randomized trial. Diabetes Care. 2011 Mar. 34(3):540-4. [View Abstract]
  69. Mianowska B, Fendler W, Szadkowska A, Baranowska A, Grzelak-Agaciak E, Sadon J, et al. HbA(1c) levels in schoolchildren with type 1 diabetes are seasonally variable and dependent on weather conditions. Diabetologia. 2011 Apr. 54(4):749-56. [View Abstract]
  70. Suzuki S, Koga M, Amamiya S, et al. Glycated albumin but not HbA(1c) reflects glycaemic control in patients with neonatal diabetes mellitus. Diabetologia. 2011 Sep. 54(9):2247-53. [View Abstract]
  71. Brooks M. Hemoglobin A1c misses many cases of diabetes. Medscape. 2019 Mar 28.
  72. McDonald TJ, Colclough K, Brown R, et al. Islet autoantibodies can discriminate maturity-onset diabetes of the young (MODY) from Type 1 diabetes. Diabet Med. 2011 Sep. 28(9):1028-33. [View Abstract]
  73. [Guideline] Chiang JL, Kirkman MS, Laffel LM, Peters AL. Type 1 Diabetes Through the Life Span: A Position Statement of the American Diabetes Association. Diabetes Care. 2014 Jun 16. [View Abstract]
  74. Tucker M. First-Ever ADA Guidance Specifically for Type 1 Diabetes. Medscape Medical news. Available at http://www.medscape.com/viewarticle/826854. Accessed: June 20, 2014.
  75. Kielgast U, Holst JJ, Madsbad S. Antidiabetic actions of endogenous and exogenous GLP-1 in type 1 diabetic patients with and without residual ß-cell function. Diabetes. 2011 May. 60(5):1599-607. [View Abstract]
  76. [Guideline] American Diabetes Association. Standards of Medical Care in Diabetes-2018 Abridged for Primary Care Providers. Clin Diabetes. 2018 Jan. 36 (1):14-37. [View Abstract]
  77. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med. 1993 Sep 30. 329(14):977-86. [View Abstract]
  78. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, et al. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med. 2005 Dec 22. 353(25):2643-53. [View Abstract]
  79. 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 Jan-Feb. 12 Suppl 1:34-41. [View Abstract]
  80. Lind M, Bounias I, Olsson M, et al. Glycaemic control and incidence of heart failure in 20,985 patients with type 1 diabetes: an observational study. Lancet. 2011 Jul 9. 378(9786):140-6. [View Abstract]
  81. Tomlin A, Dovey S, Tilyard M. Health outcomes for diabetes patients returning for three annual general practice checks. N Z Med J. 2007 Apr 13. 120(1252):U2493. [View Abstract]
  82. Jacobson AM, Ryan CM, Cleary PA, Waberski BH, Weinger K, Musen G, et al. Biomedical risk factors for decreased cognitive functioning in type 1 diabetes: an 18 year follow-up of the Diabetes Control and Complications Trial (DCCT) cohort. Diabetologia. 2011 Feb. 54(2):245-55. [View Abstract]
  83. Asvold BO, Sand T, Hestad K, Bjørgaas MR. Cognitive function in type 1 diabetic adults with early exposure to severe hypoglycemia: a 16-year follow-up study. Diabetes Care. 2010 Sep. 33(9):1945-7. [View Abstract]
  84. Garg SK, Voelmle MK, Beatson CR, et al. Use of Continuous Glucose Monitoring in Subjects With Type 1 Diabetes on Multiple Daily Injections Versus Continuous Subcutaneous Insulin Infusion Therapy: A prospective 6-month study. Diabetes Care. 2011 Mar. 34(3):574-9. [View Abstract]
  85. Battelino T, Phillip M, Bratina N, Nimri R, Oskarsson P, Bolinder J. Effect of continuous glucose monitoring on hypoglycemia in type 1 diabetes. Diabetes Care. 2011 Apr. 34(4):795-800. [View Abstract]
  86. [Guideline] Klonoff DC, Buckingham B, Christiansen JS, et al. Continuous glucose monitoring: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2011 Oct. 96(10):2968-79. [View Abstract]
  87. [Guideline] Tucker ME. ADA 2018 Standards Address Diabetes Drugs With CV Benefit. Medscape. 2017 Dec 8.
  88. Medtronic, Inc. Medtronic gains approval of first artificial pancreas device system with threshold suspend automation [press release]. September 27, 2013. Available at http://newsroom.medtronic.com/phoenix.zhtml?c=251324&p=irol-newsArticle&ID=1859361&highlight. Accessed: October 7, 2013.
  89. Tucker ME. FDA OKs insulin pump with low-glucose suspend feature. Medscape Medical News. September 27, 2013.
  90. Tucker ME. FDA Okays Use of Dexcom G5 CGM for Insulin Dosing. Medscape Medical News. 2016 Dec 20.
  91. Tucker ME. FDA Approves New 'Smart' Continuous Glucose Monitor for Diabetes. Medscape Medical News. 2018 Mar 13.
  92. Nelson R, Tucker ME. FDA Approves FreeStyle Libre System for Patients. Medscape Medical News. 2017 Sep 27.
  93. Tucker ME. FDA Approves First Implantable Continuous Glucose Monitor. Medscape Medical News. 2018 Jun 21.
  94. FDA approves first continuous glucose monitoring system with a fully implantable glucose sensor and compatible mobile app for adults with diabetes. FDA. Available at https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm611454.htm. Jun 21, 2018; Accessed: Jun 25, 2018.
  95. What is the pancreas? What is an artificial pancreas device system?. US Food and Drug Administration. Available at http://www.fda.gov/MedicalDevices/ProductsandMedicalProcedures/HomeHealthandConsumer/ConsumerProducts/ArtificialPancreas/ucm259548.htm. May 16, 2016; Accessed: July 6, 2016.
  96. Tucker ME. Coming Soon: 'Artificial Pancreas' Options for Diabetes. Medscape Medical News. June 20, 2016.
  97. Boggs W. Round-the-Clock Closed-Loop Glucose Control Leads to Better Outcomes. Medscape. May 13, 2016.
  98. Renard E, Farret A, Kropff J, et al. Day-and-Night Closed-Loop Glucose Control in Patients With Type 1 Diabetes Under Free-Living Conditions: Results of a Single-Arm 1-Month Experience Compared With a Previously Reported Feasibility Study of Evening and Night at Home. Diabetes Care. 2016 Jul. 39 (7):1151-60. [View Abstract]
  99. US Food and Drug Administration. FDA approves first automated insulin delivery device for type 1 diabetes. FDA. Available at http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm522974.htm. Sep 28, 2016; Accessed: Sep 30, 2016.
  100. Busko M. FDA Approves Artificial Pancreas for Children With Type 1 Diabetes. Medscape Medical News. 2018 Jun 22.
  101. Tucker M. FDA Approves Inhaled Insulin Afrezza for Diabetes. Medscape Medical News. Available at http://www.medscape.com/viewarticle/827539.. Accessed: July 14, 2014.
  102. Afrezza (insulin inhaled) prescribing information [package insert]. Valencia CA, United States: MannKind Corporation. June, 2014. Available at
  103. US Food and Drug Administration. Mixups between Insulin U-100 and U-500 (April 2008). FDA Patient Safety News. Available at http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/psn/transcript.cfm?show=79. Accessed: January 28, 2012.
  104. de la Pena A, Riddle M, Morrow LA, et al. Pharmacokinetics and pharmacodynamics of high-dose human regular u-500 insulin versus human regular u-100 insulin in healthy obese subjects. Diabetes Care. 2011 Dec. 34(12):2496-501. [View Abstract]
  105. Garg S, Ampudia-Blasco FJ, Pfohl M. Rapid-acting insulin analogues in Basal-bolus regimens in type 1 diabetes mellitus. Endocr Pract. 2010 May-Jun. 16(3):486-505. [View Abstract]
  106. Fiasp Product Information [package insert]. 800 Scudders Mill Road, Plainsboro, NJ 08536: Novo Nordisk Inc. September 2017. Available at
  107. Nainggolan L. FDA Approves New Fast-Acting Insulin, Fiasp, for Diabetes in Adults. Medscape Medical News. 2017 Sep 29.
  108. Blair HA, Keating GM. Insulin Glargine 300 U/mL: A Review in Diabetes Mellitus. Drugs. 2016 Mar. 76 (3):363-74. [View Abstract]
  109. Toujeo. US Food and Drug Administration. Available at https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/206538s006lbl.pdf. Accessed: 2018 25 Apr.
  110. Birkeland KI, Home PD, Wendisch U, Ratner RE, Johansen T, Endahl LA, et al. Insulin Degludec in Type 1 Diabetes: A randomized controlled trial of a new-generation ultra-long-acting insulin compared with insulin glargine. Diabetes Care. 2011 Mar. 34(3):661-5. [View Abstract]
  111. Davies MJ, Gross JL, Ono Y, Sasaki T, Bantwal G, Gall MA, et al. Efficacy and safety of insulin degludec given as part of basal-bolus treatment with mealtime insulin aspart in type 1 diabetes: a 26-week randomized, open-label, treat-to-target non-inferiority trial. Diabetes Obes Metab. 2014 Oct. 16 (10):922-30. [View Abstract]
  112. Zinman B, DeVries JH, Bode B, Russell-Jones D, Leiter LA, Moses A, et al. Efficacy and safety of insulin degludec three times a week versus insulin glargine once a day in insulin-naive patients with type 2 diabetes: results of two phase 3, 26 week, randomised, open-label, treat-to-target, non-inferiority trials. Lancet Diabetes Endocrinol. 2013 Oct. 1 (2):123-31. [View Abstract]
  113. Nainggolan L. First Launch for Fiasp   : 'Ultrafast' Mealtime Insulin Aspart. Medscape. 2017 29 Mar.
  114. Heise T, Pieber TR. Towards peakless, reproducible and long-acting insulins. An assessment of the basal analogues based on isoglycaemic clamp studies. Diabetes Obes Metab. 2007 Sep. 9(5):648-59. [View Abstract]
  115. Suissa S, Azoulay L, Dell'aniello S, et al. Long-term effects of insulin glargine on the risk of breast cancer. Diabetologia. 2011 Sep. 54(9):2254-62. [View Abstract]
  116. Johnson JA, Bowker SL, Richardson K, Marra CA. Time-varying incidence of cancer after the onset of type 2 diabetes: evidence of potential detection bias. Diabetologia. 2011 Sep. 54(9):2263-71. [View Abstract]
  117. Bao J, Gilbertson HR, Gray R, et al. Improving the Estimation of Mealtime Insulin Dose in Adults With Type 1 Diabetes: The Normal Insulin Demand for Dose Adjustment (NIDDA) study. Diabetes Care. 2011 Oct. 34(10):2146-51. [View Abstract]
  118. Bergenstal RM, Tamborlane WV, Ahmann A, Buse JB, Dailey G, Davis SN, et al. Effectiveness of sensor-augmented insulin-pump therapy in type 1 diabetes. N Engl J Med. 2010 Jul 22. 363(4):311-20. [View Abstract]
  119. Busko M. Insulin pump therapy bests injection therapy in large study. Medscape Medical News. August 19, 2013.
  120. Johnson SR, Cooper MN, Jones TW, Davis EA. Long-term outcome of insulin pump therapy in children with type 1 diabetes assessed in a large population-based case-control study. Diabetologia. 2013 Aug 21. [View Abstract]
  121. King BR, Goss PW, Paterson MA, Crock PA, Anderson DG. Changes in Altitude Cause Unintended Insulin Delivery From Insulin Pumps: Mechanisms and implications. Diabetes Care. 2011 Sep. 34(9):1932-3. [View Abstract]
  122. Grunberger G, Abelseth JM, Bailey TS, Bode BW, Handelsman Y, Hellman R. Consensus statement by the american association of clinical endocrinologists/american college of endocrinology insulin pump management task force. Endocr Pract. 2014 May 1. 20(5):463-89. [View Abstract]
  123. Babiker A, Datta V. Lipoatrophy with insulin analogues in type I diabetes. Arch Dis Child. 2011 Jan. 96(1):101-2. [View Abstract]
  124. Giménez M, Gilabert R, Monteagudo J, Alonso A, Casamitjana R, Paré C, et al. Repeated episodes of hypoglycemia as a potential aggravating factor for preclinical atherosclerosis in subjects with type 1 diabetes. Diabetes Care. 2011 Jan. 34(1):198-203. [View Abstract]
  125. Asvold BO, Sand T, Hestad KA, Bjorgaas MR. Quantitative EEG in type 1 diabetic adults with childhood exposure to severe hypoglycaemia: a 16 year follow-up study. Diabetologia. 2011 Sep. 54(9):2404-8. [View Abstract]
  126. Kacerovsky M, Jones J, Schmid AI, et al. Postprandial and fasting hepatic glucose fluxes in long-standing type 1 diabetes. Diabetes. 2011 Jun. 60(6):1752-8. [View Abstract]
  127. Ahmedani MY, Haque MS, Basit A, Fawwad A, Alvi SF. Ramadan Prospective Diabetes Study: the role of drug dosage and timing alteration, active glucose monitoring and patient education. Diabet Med. 2012 Jun. 29(6):709-15. [View Abstract]
  128. Pannu N, Wiebe N, Tonelli M. Prophylaxis strategies for contrast-induced nephropathy. JAMA. 2006 Jun 21. 295(23):2765-79. [View Abstract]
  129. Salardi S, Balsamo C, Zucchini S, Maltoni G, Scipione M, Rollo A, et al. High rate of regression from micro-macroalbuminuria to normoalbuminuria in children and adolescents with type 1 diabetes treated or not with enalapril: the influence of HDL cholesterol. Diabetes Care. 2011 Feb. 34(2):424-9. [View Abstract]
  130. de Boer IH, Rue TC, Cleary PA, et al. Long-term Renal Outcomes of Patients With Type 1 Diabetes Mellitus and Microalbuminuria: An Analysis of the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Cohort. Arch Intern Med. 2011 Mar 14. 171(5):412-420. [View Abstract]
  131. Strippoli GF, Bonifati C, Craig M, Navaneethan SD, Craig JC. Angiotensin converting enzyme inhibitors and angiotensin II receptor antagonists for preventing the progression of diabetic kidney disease. Cochrane Database Syst Rev. 2006 Oct 18. CD006257. [View Abstract]
  132. Vinik AI, Ziegler D. Diabetic cardiovascular autonomic neuropathy. Circulation. 2007 Jan 23. 115(3):387-97. [View Abstract]
  133. Lipsky BA, Berendt AR, Deery HG, Embil JM, Joseph WS, Karchmer AW, et al. Diagnosis and treatment of diabetic foot infections. Clin Infect Dis. 2004 Oct 1. 39(7):885-910. [View Abstract]
  134. Singh N, Armstrong DG, Lipsky BA. Preventing foot ulcers in patients with diabetes. JAMA. 2005 Jan 12. 293(2):217-28. [View Abstract]
  135. Buse JB, Ginsberg HN, Bakris GL, Clark NG, Costa F, Eckel R, et al. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association. Diabetes Care. 2007 Jan. 30(1):162-72. [View Abstract]
  136. Margeirsdottir HD, Stensaeth KH, Larsen JR, Brunborg C, Dahl-Jørgensen K. Early signs of atherosclerosis in diabetic children on intensive insulin treatment: a population-based study. Diabetes Care. 2010 Sep. 33(9):2043-8. [View Abstract]
  137. van Dieren S, Nöthlings U, van der Schouw YT, Spijkerman AM, Rutten GE, van der A DL, et al. Non-fasting lipids and risk of cardiovascular disease in patients with diabetes mellitus. Diabetologia. 2011 Jan. 54(1):73-7. [View Abstract]
  138. Lee SH, Kim JH, Kang MJ, et al. Implications of nocturnal hypertension in children and adolescents with type 1 diabetes. Diabetes Care. 2011 Oct. 34(10):2180-5. [View Abstract]
  139. Leiter LA, Lundman P, da Silva PM, et al. Persistent lipid abnormalities in statin-treated patients with diabetes mellitus in Europe and Canada: results of the Dyslipidaemia International Study. Diabet Med. 2011 Nov. 28(11):1343-1351. [View Abstract]
  140. Lund SS, Tarnow L, Astrup AS, Hovind P, Jacobsen PK, Alibegovic AC, et al. Effect of adjunct metformin treatment on levels of plasma lipids in patients with type 1 diabetes. Diabetes Obes Metab. 2009 Oct. 11(10):966-77. [View Abstract]
  141. Tucker ME. ACC/AHA statin guidelines, with caveats. WebMD. Available at http://www.medscape.com/viewarticle/837138. Accessed: Dec 24, 2014.
  142. Marks JB. Perioperative management of diabetes. Am Fam Physician. 2003 Jan 1. 67(1):93-100. [View Abstract]
  143. [Guideline] Qaseem A, Humphrey LL, Chou R, Snow V, Shekelle P. Use of intensive insulin therapy for the management of glycemic control in hospitalized patients: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2011 Feb 15. 154(4):260-7. [View Abstract]
  144. Kansagara D, Fu R, Freeman M, Wolf F, Helfand M. Intensive insulin therapy in hospitalized patients: a systematic review. Ann Intern Med. 2011 Feb 15. 154(4):268-82. [View Abstract]
  145. [Guideline] Moghissi ES, Korytkowski MT, DiNardo M, Einhorn D, Hellman R, Hirsch IB, et al. American Association of Clinical Endocrinologists and American Diabetes Association consensus statement on inpatient glycemic control. Diabetes Care. 2009 Jun. 32(6):1119-31. [View Abstract]
  146. Vanhorebeek I, Langouche L, Van den Berghe G. Tight blood glucose control: what is the evidence?. Crit Care Med. 2007 Sep. 35(9 Suppl):S496-502. [View Abstract]
  147. Murphy HR, Steel SA, Roland JM, et al. Obstetric and perinatal outcomes in pregnancies complicated by Type 1 and Type 2 diabetes: influences of glycaemic control, obesity and social disadvantage. Diabet Med. 2011 Sep. 28(9):1060-7. [View Abstract]
  148. Diabetes Prevention Trial--Type 1 Diabetes Study Group. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med. 2002 May 30. 346(22):1685-91.
  149. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med. 2002 May 30. 346(22):1685-91. [View Abstract]
  150. Gale EA, Bingley PJ, Emmett CL, Collier T. European Nicotinamide Diabetes Intervention Trial (ENDIT): a randomised controlled trial of intervention before the onset of type 1 diabetes. Lancet. 2004 Mar 20. 363(9413):925-31. [View Abstract]
  151. Wherrett DK, Bundy B, Becker DJ, et al. Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial. Lancet. 2011 Jul 23. 378(9788):319-27. [View Abstract]
  152. Sherry N, Hagopian W, Ludvigsson J, et al. Teplizumab for treatment of type 1 diabetes (Protégé study): 1-year results from a randomised, placebo-controlled trial. Lancet. 2011 Aug 6. 378(9790):487-97. [View Abstract]
  153. Orban T, Bundy B, Becker DJ, et al. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet. 2011 Jul 30. 378(9789):412-9. [View Abstract]
  154. [Guideline] Tucker ME. ADA Issues New Guidance on Type 1 Diabetes in Youth. Medscape Medical News. 2018 Aug 10.
  155. [Guideline] Chiang JL, Maahs DM, Garvey KC, et al. Type 1 Diabetes in Children and Adolescents: A Position Statement by the American Diabetes Association. Diabetes Care. 2018 Sep. 41 (9):2026-44. [View Abstract]
  156. [Guideline] Jenkins K. ADA Updates Recommendations for Managing Hypertension in Diabetes. Medscape. 2017 Sep 4.
  157. [Guideline] de Boer IH, Bangalore S, Benetos A, et al. Diabetes and Hypertension: A Position Statement by the American Diabetes Association. Diabetes Care. 2017 Sep. 40 (9):1273-1284. [View Abstract]
  158. [Guideline] Donaghue KC, Marcovecchio ML, Wadwa RP, et al. ISPAD Clinical Practice Consensus Guidelines 2018: microvascular and macrovascular complications in children and adolescents. Pediatr Diabetes. 2018 Oct. 19 Suppl 27:262-74. [View Abstract]
  159. [Guideline] DiMeglio LA, Acerini CL, Codner E, et al. ISPAD Clinical Practice Consensus Guidelines 2018: glycemic control targets and glucose monitoring for children, adolescents, and young adults with diabetes. Pediatr Diabetes. 2018 Oct. 19 Suppl 27:105-14. [View Abstract]
  160. [Guideline] LeRoith D, Biessels GJ, Braithwaite SS, et al. Treatment of Diabetes in Older Adults: An Endocrine Society* Clinical Practice Guideline. J Clin Endocrinol Metab. 2019 May 1. 104 (5):1520-74. [View Abstract]
  161. [Guideline] Tucker ME. New Endocrine Society Guidelines Address Diabetes in Older Adults. Medscape Medical News. 2019 Mar 23.
  162. [Guideline] American Diabetes Association. Standards of medical care in diabetes-2015 abridged for primary care providers. Clin Diabetes. 2015 Apr. 33 (2):97-111. [View Abstract]
  163. Brooks M. FDA Clears Blood Test to Help Diagnose Type 1 Diabetes. Medscape Medical News. Aug 21 2014.
  164. Hsiao-Chuan L, et al. Enterovirus infection is associated with an increased risk of childhood type 1 diabetes in Taiwan: A nationwide population-based cohort study. Diabetologia. 2014.
  165. Leegaard A, Riis A, Kornum JB, et al. Diabetes, Glycemic Control, and Risk of Tuberculosis: A population-based case-control study. Diabetes Care. 2011 Dec. 34(12):2530-5. [View Abstract]
  166. Ludwig J, Sanbonmatsu L, Gennetian L, et al. Neighborhoods, obesity, and diabetes--a randomized social experiment. N Engl J Med. 2011 Oct 20. 365(16):1509-19. [View Abstract]
  167. [Guideline] Peters A, Laffel L. Diabetes Care for Emerging Adults: Recommendations for Transition From Pediatric to Adult Diabetes Care Systems: A position statement of the American Diabetes Association, with representation by the American College of Osteopathic Family Physicians, the American Academy of Pediatrics, the American Association of Clinical Endocrinologists, the American Osteopathic Association, the Centers for Disease Control and Prevention, Children with Diabetes, The Endocrine Society, the International Socie... Diabetes Care. 2011 Nov. 34(11):2477-85. [View Abstract]
  168. US Food and Drug Administration. Early Communication About Safety of Lantus (Insulin Glargine). Available at http://www.fda.gov/Drugs/DrugSafety/ucm239376.htm. Accessed: May 22, 2012.