The American Diabetes Association (ADA) “Standards of Medical Care in Diabetes” includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, a multidisciplinary expert committee (https://doi.org/10.2337/dc21-SPPC), are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations, please refer to the Standards of Care Introduction (https://doi.org/10.2337/dc21-SINT). Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.

Recommendations

  • 9.1 Most people with type 1 diabetes should be treated with multiple daily injections of prandial and basal insulin, or continuous subcutaneous insulin infusion. A

  • 9.2 Most individuals with type 1 diabetes should use rapid-acting insulin analogs to reduce hypoglycemia risk. A

  • 9.3 Patients with type 1 diabetes should receive education on how to match prandial insulin doses to carbohydrate intake, premeal blood glucose, and anticipated physical activity. C

Insulin Therapy

Because the hallmark of type 1 diabetes is absent or near-absent β-cell function, insulin treatment is essential for individuals with type 1 diabetes. In addition to hyperglycemia, insulinopenia can contribute to other metabolic disturbances like hypertriglyceridemia and ketoacidosis as well as tissue catabolism that can be life threatening. Severe metabolic decompensation can be, and was, mostly prevented with once or twice daily injections for the six or seven decades after the discovery of insulin. However, over the past three decades, evidence has accumulated supporting more intensive insulin replacement, using multiple daily injections of insulin or continuous subcutaneous administration through an insulin pump, as providing the best combination of effectiveness and safety for people with type 1 diabetes. The Diabetes Control and Complications Trial (DCCT) demonstrated that intensive therapy with multiple daily injections or continuous subcutaneous insulin infusion (CSII) reduced A1C and was associated with improved long-term outcomes (13). The study was carried out with short-acting (regular) and intermediate-acting (NPH) human insulins. In this landmark trial, lower A1C with intensive control (7%) led to ∼50% reductions in microvascular complications over 6 years of treatment. However, intensive therapy was associated with a higher rate of severe hypoglycemia than conventional treatment (62 compared with 19 episodes per 100 patient-years of therapy). Follow-up of subjects from the DCCT more than 10 years after the active treatment component of the study demonstrated less macrovascular as well as less microvascular complications in the group that received intensive treatment (2,4).

Over the last 25 years, rapid-acting and long-acting insulin analogs have been developed that have distinct pharmacokinetics compared with recombinant human insulins: basal insulin analogs have longer duration of action with flatter, more constant plasma concentrations and activity profiles than NPH insulin; rapid-acting analogs (RAA) have a quicker onset and peak and shorter duration of action than regular human insulin. In people with type 1 diabetes, treatment with analog insulins is associated with less hypoglycemia and weight gain as well as lower A1C compared with human insulins (57). More recently, two new injectable insulin formulations with enhanced rapid action profiles have been introduced. Inhaled human insulin has a rapid peak and shortened duration of action compared with RAA and may cause less hypoglycemia and weight gain (8), and faster-acting insulin aspart and insulin lispro-aabc may reduce prandial excursions better than RAA (9,9a,9b); further investigation is needed to establish a clear place for these agents in diabetes management. In addition, new longer-acting basal analogs (U-300 glargine or degludec) may confer a lower hypoglycemia risk compared with U-100 glargine in patients with type 1 diabetes (10,11). Despite the advantages of insulin analogs in patients with type 1 diabetes, for some patients the expense and/or intensity of treatment required for their use is prohibitive. There are multiple approaches to insulin treatment, and the central precept in the management of type 1 diabetes is that some form of insulin be given in a planned regimen tailored to the individual patient to keep them safe and out of diabetic ketoacidosis and to avoid significant hypoglycemia, with every effort made to reach the patient's glycemic targets.

Most studies comparing multiple daily injections with CSII have been relatively small and of short duration. However, a recent systematic review and meta-analysis concluded that pump therapy has modest advantages for lowering A1C (−0.30% [95% CI −0.58 to −0.02]) and for reducing severe hypoglycemia rates in children and adults (12). However, there is no consensus to guide the choice of injection or pump therapy in a given patient, and research to guide this decision-making is needed (13). The arrival of continuous glucose monitors to clinical practice has proven beneficial in specific circumstances. Reduction of nocturnal hypoglycemia in people with type 1 diabetes using insulin pumps with glucose sensors is improved by automatic suspension of insulin delivery at a preset glucose level (1315). When choosing among insulin delivery systems, patient preferences, cost, insulin type and dosing regimen, and self-management capabilities should be considered (See Section 7 “Diabetes Technology,” https://doi.org/10.2337/dc21-S007).

The U.S. Food and Drug Administration (FDA) has now approved two hybrid closed-loop pump systems. The safety and efficacy of hybrid closed-loop systems has been supported in the literature in adolescents and adults with type 1 diabetes (16,17), and recent evidence suggests that a closed-loop system is superior to sensor-augmented pump therapy for glycemic control and reduction of hypoglycemia over 3 months of comparison in children and adults with type 1 diabetes (18). In the International Diabetes Closed Loop (iDCL) trial, a 6-month trial in patients with type 1 diabetes at least 14 years of age, the use of a closed-loop system was associated with a greater percentage of time spent in the target glycemic range, reduced mean glucose and A1C levels, and lower percentage of time spent in hypoglycemia compared with use of a sensor-augmented pump (19).

Intensive insulin management using a version of CSII and continuous glucose monitoring should be considered in most patients. Automated insulin delivery systems may be considered in adults with type 1 diabetes who have the skills to use them in order to improve time in range and reduce A1C and hypoglycemia (19). See Section 7 “Diabetes Technology” (https://doi.org/10.2337/dc21-S007) for a full discussion of insulin delivery devices.

In general, patients with type 1 diabetes require 50% of their daily insulin as basal and 50% as prandial. Total daily insulin requirements can be estimated based on weight, with typical doses ranging from 0.4 to 1.0 units/kg/day. Higher amounts are required during puberty, pregnancy, and medical illness. The American Diabetes Association/JDRF Type 1 Diabetes Sourcebook notes 0.5 units/kg/day as a typical starting dose in patients with type 1 diabetes who are metabolically stable, with half administered as prandial insulin given to control blood glucose after meals and the other half as basal insulin to control glycemia in the periods between meal absorption (20); this guideline provides detailed information on intensification of therapy to meet individualized needs. In addition, the American Diabetes Association position statement “Type 1 Diabetes Management Through the Life Span” provides a thorough overview of type 1 diabetes treatment (21).

Typical multidose regimens for patients with type 1 diabetes combine premeal use of shorter-acting insulins with a longer-acting formulation, usually at night. The long-acting basal dose is titrated to regulate overnight, fasting glucose. Postprandial glucose excursions are best controlled by a well-timed injection of prandial insulin. The optimal time to administer prandial insulin varies, based on the pharmacokinetics of the formulation (regular, RAA, inhaled), the premeal blood glucose level, and carbohydrate consumption. Recommendations for prandial insulin dose administration should therefore be individualized. Physiologic insulin secretion varies with glycemia, meal size, and tissue demands for glucose. To approach this variability in people using insulin treatment, strategies have evolved to adjust prandial doses based on predicted needs. Thus, education of patients on how to adjust prandial insulin to account for carbohydrate intake, premeal glucose levels, and anticipated activity can be effective and should be offered to most patients (22,23). For individuals in whom carbohydrate counting is effective, estimates of the fat and protein content of meals can be incorporated into their prandial dosing for added benefit (24).

Insulin Injection Technique

Ensuring that patients and/or caregivers understand correct insulin injection technique is important to optimize glucose control and insulin use safety. Thus, it is important that insulin be delivered into the proper tissue in the correct way. Recommendations have been published elsewhere outlining best practices for insulin injection (25). Proper insulin injection technique includes injecting into appropriate body areas, injection site rotation, appropriate care of injection sites to avoid infection or other complications, and avoidance of intramuscular (IM) insulin delivery.

Exogenously delivered insulin should be injected into subcutaneous tissue, not intramuscularly. Recommended sites for insulin injection include the abdomen, thigh, buttock, and upper arm. Because insulin absorption from IM sites differs according to the activity of the muscle, inadvertent IM injection can lead to unpredictable insulin absorption and variable effects on glucose, with IM injection being associated with frequent and unexplained hypoglycemia in several reports. Risk for IM insulin delivery is increased in younger, leaner patients when injecting into the limbs rather than truncal sites (abdomen and buttocks) and when using longer needles. Recent evidence supports the use of short needles (e.g., 4-mm pen needles) as effective and well tolerated when compared with longer needles, including a study performed in adults with obesity (26).

Injection site rotation is additionally necessary to avoid lipohypertrophy, an accumulation of subcutaneous fat in response to the adipogenic actions of insulin at a site of multiple injections. Lipohypertrophy appears as soft, smooth raised areas several centimeters in breadth and can contribute to erratic insulin absorption, increased glycemic variability, and unexplained hypoglycemic episodes. Patients and/or caregivers should receive education about proper injection site rotation and to recognize and avoid areas of lipohypertrophy. As noted in Table 4.1, examination of insulin injection sites for the presence of lipohypertrophy, as well as assessment of injection device use and injection technique, are key components of a comprehensive diabetes medical evaluation and treatment plan. Proper insulin injection technique may lead to more effective use of this therapy and, as such, holds the potential for improved clinical outcomes.

Noninsulin Treatments for Type 1 Diabetes

Injectable and oral glucose-lowering drugs have been studied for their efficacy as adjuncts to insulin treatment of type 1 diabetes. Pramlintide is based on the naturally occurring β-cell peptide amylin and is approved for use in adults with type 1 diabetes. Results from randomized controlled studies show variable reductions of A1C (0–0.3%) and body weight (1–2 kg) with addition of pramlintide to insulin (27,28). Similarly, results have been reported for several agents currently approved only for the treatment of type 2 diabetes. The addition of metformin in adults with type 1 diabetes caused small reductions in body weight and lipid levels but did not improve A1C (29,30). The addition of the glucagon-like peptide 1 (GLP-1) receptor agonist (RA) liraglutide or exenatide to insulin therapy caused small (0.2%) reductions in A1C compared with insulin alone in people with type 1 diabetes and also reduced body weight by ∼3 kg (31). Similarly, the addition of a sodium–glucose cotransporter 2 (SGLT2) inhibitor to insulin therapy has been associated with improvements in A1C and body weight when compared with insulin alone (32,33); however, SGLT2 inhibitor use in type 1 diabetes is associated with a two- to fourfold increase in ketoacidosis. The risks and benefits of adjunctive agents continue to be evaluated, but only pramlintide is approved for treatment of type 1 diabetes.

Pancreas and Islet Transplantation

Successful pancreas and islet transplantation can normalize glucose levels and mitigate microvascular complications of type 1 diabetes. However, patients receiving these treatments require lifelong immunosuppression to prevent graft rejection and/or recurrence of autoimmune islet destruction. Given the potential adverse effects of immunosuppressive therapy, pancreas transplantation should be reserved for patients with type 1 diabetes undergoing simultaneous renal transplantation, following renal transplantation, or for those with recurrent ketoacidosis or severe hypoglycemia despite intensive glycemic management (34).

Recommendations

  • 9.4 Metformin is the preferred initial pharmacologic agent for the treatment of type 2 diabetes. A

  • 9.5 Once initiated, metformin should be continued as long as it is tolerated and not contraindicated; other agents, including insulin, should be added to metformin. A

  • 9.6 Early combination therapy can be considered in some patients at treatment initiation to extend the time to treatment failure. A

  • 9.7 The early introduction of insulin should be considered if there is evidence of ongoing catabolism (weight loss), if symptoms of hyperglycemia are present, or when A1C levels (>10% [86 mmol/mol]) or blood glucose levels (≥300 mg/dL [16.7 mmol/L]) are very high. E

  • 9.8 A patient-centered approach should be used to guide the choice of pharmacologic agents. Considerations include effect on cardiovascular and renal comorbidities, efficacy, hypoglycemia risk, impact on weight, cost, risk for side effects, and patient preferences (Table 9.1 and Fig. 9.1). E

  • 9.9 Among patients with type 2 diabetes who have established atherosclerotic cardiovascular disease or indicators of high risk, established kidney disease, or heart failure, a sodium–glucose cotransporter 2 inhibitor or glucagon-like peptide 1 receptor agonist with demonstrated cardiovascular disease benefit (Table 9.1, Table 10.3B, Table 10.3C) is recommended as part of the glucose-lowering regimen independent of A1C and in consideration of patient-specific factors (Fig. 9.1 and Section 10). A

  • 9.10 In patients with type 2 diabetes, a glucagon-like peptide 1 receptor agonist is preferred to insulin when possible. A

  • 9.11 Recommendation for treatment intensification for patients not meeting treatment goals should not be delayed. A

  • 9.12 The medication regimen and medication-taking behavior should be reevaluated at regular intervals (every 3–6 months) and adjusted as needed to incorporate specific factors that impact choice of treatment (Fig. 4.1 and Table 9.1). E

  • 9.13 Clinicians should be aware of the potential for overbasalization with insulin therapy. Clinical signals that may prompt evaluation of overbasalization include basal dose more than ∼0.5 IU/kg, high bedtime-morning or post-preprandial glucose differential, hypoglycemia (aware or unaware), and high variability. Indication of overbasalization should prompt reevaluation to further individualize therapy. E

The American Diabetes Association/European Association for the Study of Diabetes consensus report “Management of Hyperglycemia in Type 2 Diabetes, 2018” and the 2019 update (35,36) recommend a patient-centered approach to choosing appropriate pharmacologic treatment of blood glucose. This includes consideration of efficacy and key patient factors: 1) important comorbidities such as atherosclerotic cardiovascular disease (ASCVD) and indicators of high ASCVD risk, chronic kidney disease (CKD), and heart failure (see Section 10 “Cardiovascular Disease and Risk Management,” https://doi.org/10.2337/dc21-S010, and Section 11 “Microvascular Complications and Foot Care,” https://doi.org/10.2337/dc21-S011), 2) hypoglycemia risk, 3) effects on body weight, 4) side effects, 5) cost, and 6) patient preferences. Lifestyle modifications that improve health (see Section 5 “Facilitating Behavior Change and Well-being to Improve Health Outcomes,” https://doi.org/10.2337/dc21-S005) should be emphasized along with any pharmacologic therapy. Section 12 “Older Adults” (https://doi.org/10.2337/dc21-S012) and Section 13 “Children and Adolescents” (https://doi.org/10.2337/dc21-S013) have recommendations specific for older adults and for children and adolescents with type 2 diabetes, respectively. Section 10 “Cardiovascular Disease and Risk Management” (https://doi.org/10.2337/dc21-S010) and Section 11 “Microvascular Complications and Foot Care” (https://doi.org/10.2337/dc21-S011) have recommendations for the use of glucose-lowering drugs in the management of cardiovascular and renal disease, respectively.

Initial Therapy

Metformin should be started at the time type 2 diabetes is diagnosed unless there are contraindications; for many patients this will be monotherapy in combination with lifestyle modifications. Additional and/or alternative agents may be considered in special circumstances, such as in individuals with established or increased risk of cardiovascular or renal complications (see Section 10 “Cardiovascular Disease and Risk Management,” https://doi.org/10.2337/dc21-S010, and Fig. 9.1). Metformin is effective and safe, is inexpensive, and may reduce risk of cardiovascular events and death (37). Metformin is available in an immediate-release form for twice-daily dosing or as an extended-release form that can be given once daily. Compared with sulfonylureas, metformin as first-line therapy has beneficial effects on A1C, weight, and cardiovascular mortality (38); there is little systematic data available for other oral agents as initial therapy of type 2 diabetes.

The principal side effects of metformin are gastrointestinal intolerance due to bloating, abdominal discomfort, and diarrhea; these can be mitigated by gradual dose titration. The drug is cleared by renal filtration, and very high circulating levels (e.g., as a result of overdose or acute renal failure) have been associated with lactic acidosis. However, the occurrence of this complication is now known to be very rare, and metformin may be safely used in patients with reduced estimated glomerular filtration rates (eGFR); the FDA has revised the label for metformin to reflect its safety in patients with eGFR ≥30 mL/min/1.73 m2 (39). A randomized trial confirmed previous observations that metformin use is associated with vitamin B12 deficiency and worsening of symptoms of neuropathy (40). This is compatible with a report from the Diabetes Prevention Program Outcomes Study (DPPOS) suggesting periodic testing of vitamin B12 (41).

In patients with contraindications or intolerance to metformin, initial therapy should be based on patient factors; consider a drug from another class depicted in Fig. 9.1. When A1C is ≥1.5% (12.5 mmol/mol) above the glycemic target (see Section 6 “Glycemic Targets,” https://doi.org/10.2337/dc21-S006, for appropriate targets), many patients will require dual combination therapy to achieve their target A1C level (42). Insulin has the advantage of being effective where other agents are not and should be considered as part of any combination regimen when hyperglycemia is severe, especially if catabolic features (weight loss, hypertriglyceridemia, ketosis) are present. It is common practice to initiate insulin therapy for patients who present with blood glucose levels ≥300 mg/dL (16.7 mmol/L) or A1C >10% (86 mmol/mol) or if the patient has symptoms of hyperglycemia (i.e., polyuria or polydipsia) or evidence of catabolism (weight loss) (Fig. 9.2). As glucose toxicity resolves, simplifying the regimen and/or changing to oral agents is often possible. However, there is evidence that patients with uncontrolled hyperglycemia associated with type 2 diabetes can also be effectively treated with a sulfonylurea (43).

Combination Therapy

Because type 2 diabetes is a progressive disease in many patients, maintenance of glycemic targets with monotherapy is often possible for only a few years, after which combination therapy is necessary. Current recommendations have been to use stepwise addition of medications to metformin to maintain A1C at target. This allows a clearer assessment of the positive and negative effects of new drugs and reduces patient risk and expense (44); based on these factors, sequential addition of oral agents to metformin has been the standard of care. However, there are data to support initial combination therapy for more rapid attainment of glycemic goals (45,46) and later combination therapy for longer durability of glycemic effect (47). The VERIFY (Vildagliptin Efficacy in combination with metfoRmIn For earlY treatment of type 2 diabetes) trial demonstrated that initial combination therapy is superior to sequential addition of medications for extending primary and secondary failure (48). In the VERIFY trial, participants receiving the initial combination of metformin and the dipeptidyl peptidase 4 (DPP-4) inhibitor vildagliptin had a slower decline of glycemic control compared with metformin alone and with vildagliptin added sequentially to metformin. These results have not been generalized to oral agents other than vildagliptin, but they suggest that more intensive early treatment has some benefits and should be considered through a shared decision-making process with patients, as appropriate. Moreover, since the absolute effectiveness of most oral medications rarely exceeds 1%, initial combination therapy should be considered in patients presenting with A1C levels 1.5–2.0% above target.

Recommendations for treatment intensification for patients not meeting treatment goals should not be delayed. Shared decision-making is important in discussions regarding treatment intensification. The choice of medication added to metformin is based on the clinical characteristics of the patient and their preferences. Important clinical characteristics include the presence of established ASCVD or indicators of high ASCVD risk, heart failure, CKD, other comorbidities, and risk for specific adverse drug effects, as well as safety, tolerability, and cost. Although there are numerous trials comparing dual therapy with metformin alone, there is little evidence to support one combination over another. A comparative effectiveness meta-analysis suggests that each new class of noninsulin agents added to initial therapy with metformin generally lowers A1C approximately 0.7–1.0% (49,50). If the A1C target is not achieved after approximately 3 months, metformin can be combined with any one of the preferred six treatment options: sulfonylurea, thiazolidinedione, DPP-4 inhibitor, SGLT2 inhibitor, GLP-1 RA, or basal insulin; the choice of which agent to add is based on drug-specific effects and patient factors (Fig. 9.1 and Table 9.1).

For patients with established ASCVD or indicators of high ASCVD risk (such as patients ≥55 years of age with coronary, carotid, or lower-extremity artery stenosis >50% or left ventricular hypertrophy), heart failure, or CKD, an SGLT2 inhibitor or GLP-1 RA with demonstrated CVD benefit (Table 9.1, Table 10.3B, Table 10.3C, and Section 10 “Cardiovascular Disease and Risk Management,” https://doi.org/10.2337/dc21-S010) is recommended as part of the glucose-lowering regimen independent of A1C, independent of metformin use, and in consideration of patient-specific factors (Fig. 9.1). For patients without established ASCVD, indicators of high ASCVD risk, heart failure, or CKD, the choice of a second agent to add to metformin is not yet guided by empiric evidence comparing across multiple classes. Rather, drug choice is based on efficacy, avoidance of side effects (particularly hypoglycemia and weight gain), cost, and patient preferences (51). Similar considerations are applied in patients who require a third agent to achieve glycemic goals. A recent systematic review and network meta-analysis suggests greatest reductions in A1C level with insulin regimens and specific GLP-1 RAs added to metformin-based background therapy (52). In all cases, treatment regimens need to be continuously reviewed for efficacy, side effects, and patient burden (Table 9.1). In some instances, patients will require medication reduction or discontinuation. Common reasons for this include ineffectiveness, intolerable side effects, expense, or a change in glycemic goals (e.g., in response to development of comorbidities or changes in treatment goals). Section 12 “Older Adults (https://doi.org/10.2337/dc21-S012) has a full discussion of treatment considerations in older adults, in whom changes of glycemic goals and de-escalation of therapy are common.

The need for the greater potency of injectable medications is common, particularly in people with a longer duration of diabetes. The addition of basal insulin, either human NPH or one of the long-acting insulin analogs, to oral agent regimens is a well-established approach that is effective for many patients. In addition, recent evidence supports the utility of GLP-1 RAs in patients not at glycemic goal. While most GLP-1 RAs are injectable, an oral formulation of semaglutide is now commercially available (53). In trials comparing the addition of an injectable GLP-1 RA or insulin in patients needing further glucose lowering, glycemic efficacy of injectable GLP-1 RA was similar or greater than that of basal insulin (5460). GLP-1 RAs in these trials had a lower risk of hypoglycemia and beneficial effects on body weight compared with insulin, albeit with greater gastrointestinal side effects. Thus, trial results support GLP-1 RAs as the preferred option for patients requiring the potency of an injectable therapy for glucose control (Fig. 9.2). However, high costs and tolerability issues are important barriers to GLP-1 RA use.

Cost for diabetes medicine has increased dramatically over the past two decades, and an increasing proportion is now passed on to patients and their families (61). Table 9.2 provides cost information for currently approved noninsulin therapies. Of note, prices listed are average wholesale prices (AWP) (62) and National Average Drug Acquisition Costs (NADAC) (63), separate measures to allow for a comparison of drug prices, but do not account for discounts, rebates, or other price adjustments often involved in prescription sales that affect the actual cost incurred by the patient. Medication costs can be a major source of stress for patients with diabetes and contribute to worse adherence to medications (64); cost-reducing strategies may improve adherence in some cases (65).

Cardiovascular Outcomes Trials

There are now multiple large randomized controlled trials reporting statistically significant reductions in cardiovascular events in patients with type 2 diabetes treated with an SGLT2 inhibitor (empagliflozin, canagliflozin, dapagliflozin) or GLP-1 RA (liraglutide, semaglutide, dulaglutide); see Section 10 “Cardiovascular Disease and Risk Management” (https://doi.org/10.2337/dc21-S010) for details. The subjects enrolled in the cardiovascular outcomes trials using empagliflozin, canagliflozin, dapagliflozin, liraglutide, and semaglutide had A1C ≥6.5%, and more than 70% were taking metformin at baseline. Thus, a practical extension of these results to clinical practice is to use these drugs preferentially in patients with type 2 diabetes and established ASCVD or indicators of high ASCVD risk. For these patients, incorporating one of the SGLT2 inhibitors or GLP-1 RAs that have been demonstrated to have cardiovascular disease benefit is recommended (Table 9.1). In cardiovascular outcomes trials, empagliflozin, canagliflozin, dapagliflozin, liraglutide, semaglutide, and dulaglutide all had beneficial effects on indices of CKD, while dedicated renal outcomes studies have demonstrated benefit of specific SGLT2 inhibitors. See Section 11 “Microvascular Complications and Foot Care” (https://doi.org/10.2337/dc21-S011) for discussion of how CKD may impact treatment choices. Additional large randomized trials of other agents in these classes are ongoing.

Insulin Therapy

Many patients with type 2 diabetes eventually require and benefit from insulin therapy (Fig. 9.2). See the section insulin injection technique, above, for guidance on how to administer insulin safely and effectively. The progressive nature of type 2 diabetes should be regularly and objectively explained to patients, and providers should avoid using insulin as a threat or describing it as a sign of personal failure or punishment. Rather, the utility and importance of insulin to maintain glycemic control once progression of the disease overcomes the effect of other agents should be emphasized. Educating and involving patients in insulin management is beneficial. For example, instruction of patients in self-titration of insulin doses based on glucose monitoring improves glycemic control in patients with type 2 diabetes initiating insulin (66). Comprehensive education regarding self-monitoring of blood glucose, diet, and the avoidance and appropriate treatment of hypoglycemia are critically important in any patient using insulin.

Basal Insulin

Basal insulin alone is the most convenient initial insulin regimen and can be added to metformin and other oral agents. Starting doses can be estimated based on body weight (0.1–0.2 units/kg/day) and the degree of hyperglycemia, with individualized titration over days to weeks as needed. The principal action of basal insulin is to restrain hepatic glucose production and limit hyperglycemia overnight and between meals (67,68). Control of fasting glucose can be achieved with human NPH insulin or a long-acting insulin analog. In clinical trials, long-acting basal analogs (U-100 glargine or detemir) have been demonstrated to reduce the risk of symptomatic and nocturnal hypoglycemia compared with NPH insulin (6974), although these advantages are modest and may not persist (75). Longer-acting basal analogs (U-300 glargine or degludec) may convey a lower hypoglycemia risk compared with U-100 glargine when used in combination with oral agents (7682). Despite evidence for reduced hypoglycemia with newer, longer-acting basal insulin analogs in clinical trial settings, in practice these effects may be modest compared with NPH insulin (83). Clinicians should be aware of the potential for overbasalization with insulin therapy. Clinical signals that may prompt evaluation of overbasalization include basal dose greater than ∼0.5 IU/kg, high bedtime-morning or post-preprandial glucose differential (e.g. bedtime-morning glucose differential ≥50 mg/dL), hypoglycemia (aware or unaware), and high variability. Indication of overbasalization should prompt reevaluation to further individualize therapy (84).

The cost of insulin has been rising steadily over the past two decades, at a pace several fold that of other medical expenditures (85). This expense contributes significant burden to patients as insulin has become a growing “out-of-pocket” cost for people with diabetes, and direct patient costs contribute to treatment nonadherence (85). Therefore, consideration of cost is an important component of effective management. For many patients with type 2 diabetes (e.g., individuals with relaxed A1C goals, low rates of hypoglycemia, and prominent insulin resistance, as well as those with cost concerns), human insulin (NPH and regular) may be the appropriate choice of therapy, and clinicians should be familiar with its use (83). Human regular insulin, NPH, and 70/30 NPH/regular products can be purchased for considerably less than the AWP and NADAC prices listed in Table 9.3 at select pharmacies.

Prandial Insulin

Many individuals with type 2 diabetes require doses of insulin before meals, in addition to basal insulin, to reach glycemic targets. A dose of 4 units or 10% of the amount of basal insulin at the largest meal or the meal with the greatest postprandial excursion is a safe estimate for initiating therapy. The prandial insulin regimen can then be intensified based on patient needs (see Fig. 9.2). People with type 2 diabetes are generally more insulin resistant than those with type 1 diabetes, require higher daily doses (∼1 unit/kg), and have lower rates of hypoglycemia (86). Titration can be based on home glucose monitoring or A1C. With significant additions to the prandial insulin dose, particularly with the evening meal, consideration should be given to decreasing basal insulin. Meta-analyses of trials comparing rapid-acting insulin analogs with human regular insulin in patients with type 2 diabetes have not reported important differences in A1C or hypoglycemia (87,88).

Concentrated Insulins

Several concentrated insulin preparations are currently available. U-500 regular insulin is, by definition, five times more concentrated than U-100 regular insulin. Regular U-500 has distinct pharmacokinetics with delayed onset and longer duration of action, has characteristics more like an intermediate-acting (NPH) insulin, and can be used as two or three daily injections (89). U-300 glargine and U-200 degludec are three and two times as concentrated as their U-100 formulations and allow higher doses of basal insulin administration per volume used. U-300 glargine has a longer duration of action than U-100 glargine but modestly lower efficacy per unit administered (90,91). The FDA has also approved a concentrated formulation of rapid-acting insulin lispro, U-200 (200 units/mL) and insulin lispro-aabc (U-200). These concentrated preparations may be more convenient and comfortable for patients to inject and may improve adherence in those with insulin resistance who require large doses of insulin. While U-500 regular insulin is available in both prefilled pens and vials, other concentrated insulins are available only in prefilled pens to minimize the risk of dosing errors.

Inhaled Insulin

Inhaled insulin is available as a rapid-acting insulin; studies in people with type 1 diabetes suggest rapid pharmacokinetics (8). A pilot study found evidence that compared with injectable rapid-acting insulin, supplemental doses of inhaled insulin taken based on postprandial glucose levels may improve blood glucose management without additional hypoglycemia or weight gain (92), although results from a larger study are needed for confirmation. Inhaled insulin is contraindicated in patients with chronic lung disease, such as asthma and chronic obstructive pulmonary disease, and is not recommended in patients who smoke or who recently stopped smoking. All patients require spirometry (forced expiratory volume in 1 s [FEV1]) testing to identify potential lung disease prior to and after starting inhaled insulin therapy.

Combination Injectable Therapy

If basal insulin has been titrated to an acceptable fasting blood glucose level (or if the dose is >0.5 units/kg/day with indications of need for other therapy) and A1C remains above target, consider advancing to combination injectable therapy (Fig. 9.2). This approach can use a GLP-1 RA added to basal insulin or multiple doses of insulin. The combination of basal insulin and GLP-1 RA has potent glucose-lowering actions and less weight gain and hypoglycemia compared with intensified insulin regimens (9395), with one study suggesting greater durability of glycemic effect compared with addition of basal insulin alone (47). Two different once-daily, fixed dual-combination products containing basal insulin plus a GLP-1 RA are available: insulin glargine plus lixisenatide and insulin degludec plus liraglutide.

Intensification of insulin treatment can be done by adding doses of prandial to basal insulin. Starting with a single prandial dose with the largest meal of the day is simple and effective, and it can be advanced to a regimen with multiple prandial doses if necessary (96). Alternatively, in a patient on basal insulin in whom additional prandial coverage is desired, the regimen can be converted to two doses of a premixed insulin. Each approach has advantages and disadvantages. For example, basal/prandial regimens offer greater flexibility for patients who eat on irregular schedules. On the other hand, two doses of premixed insulin is a simple, convenient means of spreading insulin across the day. Moreover, human insulins, separately, self-mixed, or as premixed NPH/regular (70/30) formulations, are less costly alternatives to insulin analogs. Figure 9.2 outlines these options as well as recommendations for further intensification, if needed, to achieve glycemic goals. When initiating combination injectable therapy, metformin therapy should be maintained, while sulfonylureas and DPP-4 inhibitors are typically weaned or discontinued. In patients with suboptimal blood glucose control, especially those requiring large insulin doses, adjunctive use of a thiazolidinedione or an SGLT2 inhibitor may help to improve control and reduce the amount of insulin needed, though potential side effects should be considered. Once a basal/bolus insulin regimen is initiated, dose titration is important, with adjustments made in both mealtime and basal insulins based on the blood glucose levels and an understanding of the pharmacodynamic profile of each formulation (pattern control). As people with type 2 diabetes get older, it may become necessary to simplify complex insulin regimens because of a decline in self-management ability (see Section 12 “Older Adults,” https://doi.org/10.2337/dc21-S012).

Suggested citation: American Diabetes Association. 9. Pharmacologic approaches to glycemic treatment: Standards of Medical Care in Diabetes—2021. Diabetes Care 2021;44(Suppl. 1):S111–S124

1.
Cleary
PA
,
Orchard
TJ
,
Genuth
S
, et al.;
DCCT/EDIC Research Group
.
The effect of intensive glycemic treatment on coronary artery calcification in type 1 diabetic participants of the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) study
.
Diabetes
2006
;
55
:
3556
3565
2.
Nathan
DM
,
Cleary
PA
,
Backlund
J-YC
, et al.;
Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group
.
Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes
.
N Engl J Med
2005
;
353
:
2643
2653
3.
Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) Study Research Group
.
Mortality in type 1 diabetes in the DCCT/EDIC versus the general population
.
Diabetes Care
2016
;
39
:
1378
1383
4.
Writing Team for the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Research Group
.
Effect of intensive therapy on the microvascular complications of type 1 diabetes mellitus
.
JAMA
2002
;
287
:
2563
2569
5.
Tricco
AC
,
Ashoor
HM
,
Antony
J
, et al
.
Safety, effectiveness, and cost effectiveness of long acting versus intermediate acting insulin for patients with type 1 diabetes: systematic review and network meta-analysis
.
BMJ
2014
;
349
:
g5459
6.
Bartley
PC
,
Bogoev
M
,
Larsen
J
,
Philotheou
A
.
Long-term efficacy and safety of insulin detemir compared to Neutral Protamine Hagedorn insulin in patients with type 1 diabetes using a treat-to-target basal-bolus regimen with insulin aspart at meals: a 2-year, randomized, controlled trial
.
Diabet Med
2008
;
25
:
442
449
7.
DeWitt
DE
,
Hirsch
IB
.
Outpatient insulin therapy in type 1 and type 2 diabetes mellitus: scientific review
.
JAMA
2003
;
289
:
2254
2264
8.
Bode
BW
,
McGill
JB
,
Lorber
DL
,
Gross
JL
,
Chang
PC
,
Bregman
DB
;
Affinity 1 Study Group
.
Inhaled technosphere insulin compared with injected prandial insulin in type 1 diabetes: a randomized 24-week trial
.
Diabetes Care
2015
;
38
:
2266
2273
9.
Russell-Jones
D
,
Bode
BW
,
De Block
C
, et al
.
Fast-acting insulin aspart improves glycemic control in basal-bolus treatment for type 1 diabetes: results of a 26-week multicenter, active-controlled, treat-to-target, randomized, parallel-group trial (onset 1)
.
Diabetes Care
2017
;
40
:
943
950
9a.
Klaff
L
,
Cao
D
,
Dellva
MA
, et al
.
Ultra rapid lispro improves postprandial glucose control compared with lispro in patients with type 1 diabetes: results from the 26-week PRONTO-T1D study
.
Diabetes Obes Metab
2020
;
22
:
1799
1807
9b.
Blevins
T
,
Zhang
Q
,
Frias
JP
,
Jinnouchi
H
,
Chang
AM
;
PRONTO-T2D Investigators. Randomized double-blind clinical trial comparing ultra rapid lispro with lispro in a basal-bolus regimen in patients with type 2 diabetes: PRONTO-T2D
.
Diabetes Care 2020;43:2991–2998
10.
Lane
W
,
Bailey
TS
,
Gerety
G
, et al.;
Group Information
;
SWITCH 1
.
Effect of insulin degludec vs insulin glargine U100 on hypoglycemia in patients with type 1 diabetes: the SWITCH 1 randomized clinical trial
.
JAMA
2017
;
318
:
33
44
11.
Home
PD
,
Bergenstal
RM
,
Bolli
GB
, et al
.
New insulin glargine 300 units/mL versus glargine 100 units/mL in people with type 1 diabetes: a randomized, phase 3a, open-label clinical trial (EDITION 4)
.
Diabetes Care
2015
;
38
:
2217
2225
12.
Yeh
H-C
,
Brown
TT
,
Maruthur
N
, et al
.
Comparative effectiveness and safety of methods of insulin delivery and glucose monitoring for diabetes mellitus: a systematic review and meta-analysis
.
Ann Intern Med
2012
;
157
:
336
347
13.
Pickup
JC
.
The evidence base for diabetes technology: appropriate and inappropriate meta-analysis
.
J Diabetes Sci Technol
2013
;
7
:
1567
1574
14.
Bergenstal
RM
,
Klonoff
DC
,
Garg
SK
, et al.;
ASPIRE In-Home Study Group
.
Threshold-based insulin-pump interruption for reduction of hypoglycemia
.
N Engl J Med
2013
;
369
:
224
232
15.
Buckingham
BA
,
Raghinaru
D
,
Cameron
F
, et al.;
In Home Closed Loop Study Group
.
Predictive low-glucose insulin suspension reduces duration of nocturnal hypoglycemia in children without increasing ketosis
.
Diabetes Care
2015
;
38
:
1197
1204
16.
Bergenstal
RM
,
Garg
S
,
Weinzimer
SA
, et al
.
Safety of a hybrid closed-loop insulin delivery system in patients with type 1 diabetes
.
JAMA
2016
;
316
:
1407
1408
17.
Garg
SK
,
Weinzimer
SA
,
Tamborlane
WV
, et al
.
Glucose outcomes with the in-home use of a hybrid closed-loop insulin delivery system in adolescents and adults with type 1 diabetes
.
Diabetes Technol Ther
2017
;
19
:
155
163
18.
Tauschmann
M
,
Thabit
H
,
Bally
L
, et al.;
APCam11 Consortium
.
Closed-loop insulin delivery in suboptimally controlled type 1 diabetes: a multicentre, 12-week randomised trial
.
Lancet
2018
;
392
:
1321
1329
19.
Brown
SA
,
Kovatchev
BP
,
Raghinaru
D
, et al
.; iDCL Trial Research Group. Six-month randomized, multicenter trial of closed-loop control in type 1 diabetes. N Engl J Med
2019
;381:1707–1717
20.
Peters
AL
,
Laffel
L
(Eds.).
American Diabetes Association/JDRF Type 1 Diabetes Sourcebook
.
Alexandria, VA
,
American Diabetes Association
,
2013
21.
Chiang
JL
,
Kirkman
MS
,
Laffel
LMB
,
Peters
AL
;
Type 1 Diabetes Sourcebook Authors
.
Type 1 diabetes through the life span: a position statement of the American Diabetes Association
.
Diabetes Care
2014
;
37
:2034–2054
22.
Bell
KJ
,
Barclay
AW
,
Petocz
P
,
Colagiuri
S
,
Brand-Miller
JC
.
Efficacy of carbohydrate counting in type 1 diabetes: a systematic review and meta-analysis
.
Lancet Diabetes Endocrinol
2014
;
2
:
133
140
23.
Vaz
EC
,
Porfírio
GJM
,
Nunes
HRC
,
Nunes-Nogueira
VDS
.
Effectiveness and safety of carbohydrate counting in the management of adult patients with type 1 diabetes mellitus: a systematic review and meta-analysis
.
Arch Endocrinol Metab
2018
;
62
:
337
345
24.
Bell
KJ
,
Smart
CE
,
Steil
GM
,
Brand-Miller
JC
,
King
B
,
Wolpert
HA
.
Impact of fat, protein, and glycemic index on postprandial glucose control in type 1 diabetes: implications for intensive diabetes management in the continuous glucose monitoring era
.
Diabetes Care
2015
;
38
:
1008
1015
25.
Frid
AH
,
Kreugel
G
,
Grassi
G
, et al
.
New insulin delivery recommendations
.
Mayo Clin Proc
2016
;
91
:
1231
1255
26.
Bergenstal
RM
,
Strock
ES
,
Peremislov
D
,
Gibney
MA
,
Parvu
V
,
Hirsch
LJ
.
Safety and efficacy of insulin therapy delivered via a 4mm pen needle in obese patients with diabetes
.
Mayo Clin Proc
2015
;
90
:
329
338
27.
Ratner
RE
,
Dickey
R
,
Fineman
M
, et al
.
Amylin replacement with pramlintide as an adjunct to insulin therapy improves long-term glycaemic and weight control in type 1 diabetes mellitus: a 1-year, randomized controlled trial
.
Diabet Med
2004
;
21
:
1204
1212
28.
Edelman
S
,
Garg
S
,
Frias
J
, et al
.
A double-blind, placebo-controlled trial assessing pramlintide treatment in the setting of intensive insulin therapy in type 1 diabetes
.
Diabetes Care
2006
;
29
:
2189
2195
29.
Meng
H
,
Zhang
A
,
Liang
Y
,
Hao
J
,
Zhang
X
,
Lu
J
.
Effect of metformin on glycaemic control in patients with type 1 diabetes: a meta-analysis of randomized controlled trials
.
Diabetes Metab Res Rev
2018
;
34
:
e2983
30.
Petrie
JR
,
Chaturvedi
N
,
Ford
I
, et al.;
REMOVAL Study Group
.
Cardiovascular and metabolic effects of metformin in patients with type 1 diabetes (REMOVAL): a double-blind, randomised, placebo-controlled trial
.
Lancet Diabetes Endocrinol
2017
;
5
:
597
609
31.
Wang
W
,
Liu
H
,
Xiao
S
,
Liu
S
,
Li
X
,
Yu
P
.
Effects of insulin plus glucagon-like peptide-1 receptor agonists (GLP-1RAs) in treating type 1 diabetes mellitus: a systematic review and meta-analysis
.
Diabetes Ther
2017
;
8
:
727
738
32.
Dandona
P
,
Mathieu
C
,
Phillip
M
, et al.;
DEPICT-1 Investigators
.
Efficacy and safety of dapagliflozin in patients with inadequately controlled type 1 diabetes (DEPICT-1): 24 week results from a multicentre, double-blind, phase 3, randomised controlled trial
.
Lancet Diabetes Endocrinol
2017
;
5
:
864
876
33.
Rosenstock
J
,
Marquard
J
,
Laffel
LM
, et al
.
Empagliflozin as adjunctive to insulin therapy in type 1 diabetes: the EASE trials
.
Diabetes Care
2018
;
41
:
2560
2569
34.
Dean
PG
,
Kukla
A
,
Stegall
MD
,
Kudva
YC
. Pancreas transplantation. BMJ.
2017
;357:j1321
35.
Davies
MJ
,
D’Alessio
DA
,
Fradkin
J
, et al
.
Management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)
.
Diabetes Care
2018
;
41
:
2669
2701
36.
Buse
JB
,
Wexler
DJ
,
Tsapas
A
, et al
.
2019 update to: management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)
.
Diabetes Care
2020
;
43
:
487
493
37.
Holman
RR
,
Paul
SK
,
Bethel
MA
,
Matthews
DR
,
Neil
HAW
.
10-year follow-up of intensive glucose control in type 2 diabetes
.
N Engl J Med
2008
;
359
:
1577
1589
38.
Maruthur
NM
,
Tseng
E
,
Hutfless
S
, et al
.
Diabetes medications as monotherapy or metformin-based combination therapy for type 2 diabetes: a systematic review and meta-analysis
.
Ann Intern Med
2016
;
164
:
740
751
39.
U.S. Food and Drug Administration
.
FDA Drug Safety Communication: FDA revises warnings regarding use of the diabetes medicine metformin in certain patients with reduced kidney function. Accessed 6 March 2020
. Available from https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-fda-revises-warnings-regarding-use-diabetes-medicine-metformin-certain
40.
Out
M
,
Kooy
A
,
Lehert
P
,
Schalkwijk
CA
,
Stehouwer
CDA
.
Long-term treatment with metformin in type 2 diabetes and methylmalonic acid: post hoc analysis of a randomized controlled 4.3 year trial
.
J Diabetes Complications
2018
;
32
:
171
178
41.
Aroda
VR
,
Edelstein
SL
,
Goldberg
RB
, et al.;
Diabetes Prevention Program Research Group
.
Long-term metformin use and vitamin B12 deficiency in the diabetes prevention program outcomes study
.
J Clin Endocrinol Metab
2016
;
101
:
1754
1761
42.
Henry
RR
,
Murray
AV
,
Marmolejo
MH
,
Hennicken
D
,
Ptaszynska
A
,
List
JF
.
Dapagliflozin, metformin XR, or both: initial pharmacotherapy for type 2 diabetes, a randomised controlled trial
.
Int J Clin Pract
2012
;
66
:
446
456
43.
Babu
A
,
Mehta
A
,
Guerrero
P
, et al
.
Safe and simple emergency department discharge therapy for patients with type 2 diabetes mellitus and severe hyperglycemia
.
Endocr Pract
2009
;
15
:
696
704
44.
Cahn
A
,
Cefalu
WT
.
Clinical considerations for use of initial combination therapy in type 2 diabetes
.
Diabetes Care
2016
;
39
(
Suppl. 2
):
S137
S145
45.
Abdul-Ghani
MA
,
Puckett
C
,
Triplitt
C
, et al
.
Initial combination therapy with metformin, pioglitazone and exenatide is more effective than sequential add-on therapy in subjects with new-onset diabetes. Results from the Efficacy and Durability of Initial Combination Therapy for Type 2 Diabetes (EDICT): a randomized trial
.
Diabetes Obes Metab
2015
;
17
:
268
275
46.
Phung
OJ
,
Sobieraj
DM
,
Engel
SS
,
Rajpathak
SN
.
Early combination therapy for the treatment of type 2 diabetes mellitus: systematic review and meta-analysis
.
Diabetes Obes Metab
2014
;
16
:
410
417
47.
Aroda
VR
,
González-Galvez
G
,
Grøn
R
, et al
.
Durability of insulin degludec plus liraglutide versus insulin glargine U100 as initial injectable therapy in type 2 diabetes (DUAL VIII): a multicentre, open-label, phase 3b, randomised controlled trial
.
Lancet Diabetes Endocrinol
2019
;
7
:
596
605
48.
Matthews
DR
,
Paldánius
PM
,
Proot
P
,
Chiang
Y
,
Stumvoll
M
,
Prato
SD
. Glycaemic durability of an early combination therapy with vildagliptin and metformin versus sequential metformin monotherapy in newly diagnosed type 2 diabetes (VERIFY): a 5-year, multicentre, randomised, double-blind trial. Lancet
2019
;394:1519–1529
49.
Bennett
WL
,
Maruthur
NM
,
Singh
S
, et al
.
Comparative effectiveness and safety of medications for type 2 diabetes: an update including new drugs and 2-drug combinations
.
Ann Intern Med
2011
;
154
:
602
613
50.
Maloney
A
,
Rosenstock
J
,
Fonseca
V
.
A model-based meta-analysis of 24 antihyperglycemic drugs for type 2 diabetes: comparison of treatment effects at therapeutic doses
.
Clin Pharmacol Ther
2019
;
105
:
1213
1223
51.
Vijan
S
,
Sussman
JB
,
Yudkin
JS
,
Hayward
RA
.
Effect of patients’ risks and preferences on health gains with plasma glucose level lowering in type 2 diabetes mellitus
.
JAMA Intern Med
2014
;
174
:
1227
1234
52.
Tsapas
A
,
Avgerinos
I
,
Karagiannis
T
, et al
.
Comparative effectiveness of glucose-lowering drugs for type 2 diabetes: a systematic review and network meta-analysis
.
Ann Intern Med
2020
;
173
:
278
286
53.
Pratley
R
,
Amod
A
,
Hoff
ST
, et al.;
PIONEER 4 investigators
.
Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial
.
Lancet
2019
;
394
:
39
50
54.
Singh
S
,
Wright
EE
 Jr
,
Kwan
AYM
, et al
.
Glucagon-like peptide-1 receptor agonists compared with basal insulins for the treatment of type 2 diabetes mellitus: a systematic review and meta-analysis
.
Diabetes Obes Metab
2017
;
19
:
228
238
55.
Levin
PA
,
Nguyen
H
,
Wittbrodt
ET
,
Kim
SC
.
Glucagon-like peptide-1 receptor agonists: a systematic review of comparative effectiveness research
.
Diabetes Metab Syndr Obes
2017
;
10
:
123
139
56.
Abd El Aziz
MS
,
Kahle
M
,
Meier
JJ
,
Nauck
MA
.
A meta-analysis comparing clinical effects of short- or long-acting GLP-1 receptor agonists versus insulin treatment from head-to-head studies in type 2 diabetic patients
.
Diabetes Obes Metab
2017
;
19
:
216
227
57.
Giorgino
F
,
Benroubi
M
,
Sun
J-H
,
Zimmermann
AG
,
Pechtner
V
.
Efficacy and safety of once-weekly dulaglutide versus insulin glargine in patients with type 2 diabetes on metformin and glimepiride (AWARD-2)
.
Diabetes Care
2015
;
38
:
2241
2249
58.
Aroda
VR
,
Bain
SC
,
Cariou
B
, et al
.
Efficacy and safety of once-weekly semaglutide versus once-daily insulin glargine as add-on to metformin (with or without sulfonylureas) in insulin-naive patients with type 2 diabetes (SUSTAIN 4): a randomised, open-label, parallel-group, multicentre, multinational, phase 3a trial
.
Lancet Diabetes Endocrinol
2017
;
5
:
355
366
59.
Davies
M
,
Heller
S
,
Sreenan
S
, et al
.
Once-weekly exenatide versus once- or twice-daily insulin detemir: randomized, open-label, clinical trial of efficacy and safety in patients with type 2 diabetes treated with metformin alone or in combination with sulfonylureas
.
Diabetes Care
2013
;
36
:
1368
1376
60.
Diamant
M
,
Van Gaal
L
,
Stranks
S
, et al
.
Once weekly exenatide compared with insulin glargine titrated to target in patients with type 2 diabetes (DURATION-3): an open-label randomised trial
.
Lancet
2010
;
375
:
2234
2243
61.
Riddle
MC
,
Herman
WH
.
The cost of diabetes care—an elephant in the room
.
Diabetes Care
2018
;
41
:
929
932
62.
Truven Health Analytics
. Micromedex 2.0: Introduction to RED BOOK Online. Accessed 10 September 2020. Available from https://www.micromedexsolutions.com/micromedex2/4.34.0/WebHelp/RED_BOOK/Introduction_to_REDB_BOOK_Online.htm
63.
Centers for Medicare & Medicaid Services
. NADAC (National Average Drug Acquisition Cost) drug pricing and payment. Accessed 2 October 2020. Available from https://data.medicaid.gov/Drug-Pricing-and-Payment/NADAC-National-Average-Drug-Acquisition-Cost-/a4y5-998d
64.
Kang
H
,
Lobo
JM
,
Kim
S
,
Sohn
M-W
.
Cost-related medication non-adherence among U.S. adults with diabetes
.
Diabetes Res Clin Pract
2018
;
143
:
24
33
65.
Patel
MR
,
Piette
JD
,
Resnicow
K
,
Kowalski-Dobson
T
,
Heisler
M
.
Social determinants of health, cost-related nonadherence, and cost-reducing behaviors among adults with diabetes: findings from the National Health Interview Survey
.
Med Care
2016
;
54
:
796
803
66.
Blonde
L
,
Merilainen
M
,
Karwe
V
,
Raskin
P
;
TITRATE Study Group
.
Patient-directed titration for achieving glycaemic goals using a once-daily basal insulin analogue: an assessment of two different fasting plasma glucose targets - the TITRATE study
.
Diabetes Obes Metab
2009
;
11
:
623
631
67.
Porcellati
F
,
Lucidi
P
,
Cioli
P
, et al
.
Pharmacokinetics and pharmacodynamics of insulin glargine given in the evening as compared with in the morning in type 2 diabetes
.
Diabetes Care
2015
;
38
:
503
512
68.
Wang
Z
,
Hedrington
MS
,
Gogitidze Joy
N
, et al
.
Dose-response effects of insulin glargine in type 2 diabetes
.
Diabetes Care
2010
;
33
:
1555
1560
69.
Singh
SR
,
Ahmad
F
,
Lal
A
,
Yu
C
,
Bai
Z
,
Bennett
H
.
Efficacy and safety of insulin analogues for the management of diabetes mellitus: a meta-analysis
.
CMAJ
2009
;
180
:
385
397
70.
Horvath
K
,
Jeitler
K
,
Berghold
A
, et al
.
Long-acting insulin analogues versus NPH insulin (human isophane insulin) for type 2 diabetes mellitus
.
Cochrane Database Syst Rev
2007
;
2
:
CD005613
71.
Monami
M
,
Marchionni
N
,
Mannucci
E
.
Long-acting insulin analogues versus NPH human insulin in type 2 diabetes: a meta-analysis
.
Diabetes Res Clin Pract
2008
;
81
:
184
189
72.
Owens
DR
,
Traylor
L
,
Mullins
P
,
Landgraf
W
.
Patient-level meta-analysis of efficacy and hypoglycaemia in people with type 2 diabetes initiating insulin glargine 100U/mL or neutral protamine Hagedorn insulin analysed according to concomitant oral antidiabetes therapy
.
Diabetes Res Clin Pract
2017
;
124
(
Suppl. C
):
57
65
73.
Riddle
MC
,
Rosenstock
J
,
Gerich
J
;
Insulin Glargine 4002 Study Investigators
.
The treat-to-target trial: randomized addition of glargine or human NPH insulin to oral therapy of type 2 diabetic patients
.
Diabetes Care
2003
;
26
:
3080
3086
74.
Hermansen
K
,
Davies
M
,
Derezinski
T
,
Martinez Ravn
G
,
Clauson
P
,
Home
P
.
A 26-week, randomized, parallel, treat-to-target trial comparing insulin detemir with NPH insulin as add-on therapy to oral glucose-lowering drugs in insulin-naive people with type 2 diabetes
.
Diabetes Care
2006
;
29
:
1269
1274
75.
Yki-Järvinen
H
,
Kauppinen-Mäkelin
R
,
Tiikkainen
M
, et al
.
Insulin glargine or NPH combined with metformin in type 2 diabetes: the LANMET study
.
Diabetologia
2006
;
49
:
442
451
76.
Bolli
GB
,
Riddle
MC
,
Bergenstal
RM
, et al.;
on behalf of the EDITION 3 study investigators
.
New insulin glargine 300 U/ml compared with glargine 100 U/ml in insulin-naïve people with type 2 diabetes on oral glucose-lowering drugs: a randomized controlled trial (EDITION 3)
.
Diabetes Obes Metab
2015
;
17
:
386
394
77.
Terauchi
Y
,
Koyama
M
,
Cheng
X
, et al
.
New insulin glargine 300 U/ml versus glargine 100 U/ml in Japanese people with type 2 diabetes using basal insulin and oral antihyperglycaemic drugs: glucose control and hypoglycaemia in a randomized controlled trial (EDITION JP 2)
.
Diabetes Obes Metab
2016
;
18
:
366
374
78.
Yki-Järvinen
H
,
Bergenstal
RM
,
Bolli
GB
, et al
.
Glycaemic control and hypoglycaemia with new insulin glargine 300 U/ml versus insulin glargine 100 U/ml in people with type 2 diabetes using basal insulin and oral antihyperglycaemic drugs: the EDITION 2 randomized 12-month trial including 6-month extension
.
Diabetes Obes Metab
2015
;
17
:
1142
1149
79.
Marso
SP
,
McGuire
DK
,
Zinman
B
, et al.;
DEVOTE Study Group
.
Efficacy and safety of degludec versus glargine in type 2 diabetes
.
N Engl J Med
2017
;
377
:723–732
80.
Rodbard
HW
,
Cariou
B
,
Zinman
B
, et al.;
BEGIN Once Long trial investigators
.
Comparison of insulin degludec with insulin glargine in insulin-naive subjects with type 2 diabetes: a 2-year randomized, treat-to-target trial
.
Diabet Med
2013
;
30
:
1298
1304
81.
Wysham
C
,
Bhargava
A
,
Chaykin
L
, et al
.
Effect of insulin degludec vs insulin glargine u100 on hypoglycemia in patients with type 2 diabetes: the SWITCH 2 randomized clinical trial
.
JAMA
2017
;
318
:
45
56
82.
Zinman
B
,
Philis-Tsimikas
A
,
Cariou
B
, et al.;
NN1250-3579 (BEGIN Once Long) Trial Investigators
.
Insulin degludec versus insulin glargine in insulin-naive patients with type 2 diabetes: a 1-year, randomized, treat-to-target trial (BEGIN Once Long)
.
Diabetes Care
2012
;
35
:
2464
2471
83.
Lipska
KJ
,
Parker
MM
,
Moffet
HH
,
Huang
ES
,
Karter
AJ
.
Association of initiation of basal insulin analogs vs neutral protamine hagedorn insulin with hypoglycemia-related emergency department visits or hospital admissions and with glycemic control in patients with type 2 diabetes
.
JAMA
2018
;
320
:
53
62
84.
Cowart
K
.
Overbasalization: addressing hesitancy in treatment intensification beyond basal insulin
.
Clin Diabetes
2020
;
38
:
304
310
85.
Cefalu
WT
,
Dawes
DE
,
Gavlak
G
, et al.;
Insulin Access and Affordability Working Group
.
Insulin Access and Affordability Working Group: conclusions and recommendations
.
Diabetes Care
2018
;
41
:
1299
1311
86.
McCall
AL
.
Insulin therapy and hypoglycemia
.
Endocrinol Metab Clin North Am
2012
;
41
:
57
87
87.
Mannucci
E
,
Monami
M
,
Marchionni
N
.
Short-acting insulin analogues vs. regular human insulin in type 2 diabetes: a meta-analysis
.
Diabetes Obes Metab
2009
;
11
:
53
59
88.
Heller
S
,
Bode
B
,
Kozlovski
P
,
Svendsen
AL
.
Meta-analysis of insulin aspart versus regular human insulin used in a basal-bolus regimen for the treatment of diabetes mellitus
.
J Diabetes
2013
;
5
:
482
491
89.
Wysham
C
,
Hood
RC
,
Warren
ML
,
Wang
T
,
Morwick
TM
,
Jackson
JA
.
Effect of total daily dose on efficacy, dosing, and safety of 2 dose titration regimens of human regular U500 insulin in severely insulin-resistant patients with type 2 diabetes
.
Endocr Pract
2016
;
22
:
653
665
90.
Riddle
MC
,
Yki-Järvinen
H
,
Bolli
GB
, et al
.
One-year sustained glycaemic control and less hypoglycaemia with new insulin glargine 300 U/ml compared with 100 U/ml in people with type 2 diabetes using basal plus meal-time insulin: the EDITION 1 12-month randomized trial, including 6-month extension
.
Diabetes Obes Metab
2015
;
17
:
835
842
91.
Yki-Järvinen
H
,
Bergenstal
R
,
Ziemen
M
, et al.;
EDITION 2 Study Investigators
.
New insulin glargine 300 units/mL versus glargine 100 units/mL in people with type 2 diabetes using oral agents and basal insulin: glucose control and hypoglycemia in a 6-month randomized controlled trial (EDITION 2)
.
Diabetes Care
2014
;
37
:
3235
3243
92.
Akturk
HK
,
Snell-Bergeon
JK
,
Rewers
A
, et al
.
Improved postprandial glucose with inhaled technosphere insulin compared with insulin aspart in patients with type 1 diabetes on multiple daily injections: the STAT study
.
Diabetes Technol Ther
2018
;
20
:
639
647
93.
Diamant
M
,
Nauck
MA
,
Shaginian
R
, et al.;
4B Study Group
.
Glucagon-like peptide 1 receptor agonist or bolus insulin with optimized basal insulin in type 2 diabetes
.
Diabetes Care
2014
;
37
:
2763
2773
94.
Eng
C
,
Kramer
CK
,
Zinman
B
,
Retnakaran
R
.
Glucagon-like peptide-1 receptor agonist and basal insulin combination treatment for the management of type 2 diabetes: a systematic review and meta-analysis
.
Lancet
2014
;
384
:
2228
2234
95.
Maiorino
MI
,
Chiodini
P
,
Bellastella
G
,
Capuano
A
,
Esposito
K
,
Giugliano
D
.
Insulin and glucagon-like peptide 1 receptor agonist combination therapy in type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials
.
Diabetes Care
2017
;
40
:
614
624
96.
Rodbard
HW
,
Visco
VE
,
Andersen
H
,
Hiort
LC
,
Shu
DHW
.
Treatment intensification with stepwise addition of prandial insulin aspart boluses compared with full basal-bolus therapy (FullSTEP Study): a randomised, treat-to-target clinical trial
.
Lancet Diabetes Endocrinol
2014
;
2
:
30
37
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.